Recent Advances in NGF and Related Molecules: Laura Calzà Luigi Aloe Luciana Giardino Editors

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 302

Advances in Experimental Medicine and Biology 1331

Cellular Neuroscience, Neural Circuits and Systems Neuroscience

Laura Calzà
Luigi Aloe
Luciana Giardino Editors

Recent Advances
in NGF and Related
Molecules
The Continuum of the NGF “Saga”
Advances in Experimental Medicine
and Biology

Cellular Neuroscience, Neural Circuits


and Systems Neuroscience

Volume 1331

Series Editors
Adalberto Merighi, Department of Veterinary Sciences, University of Turin,
Grugliasco, Italy
Laura Lossi, Department of Veterinary Science, University of Turin,
Grugliasco, Italy
Alberto Granato, Department of Psychology, Catholic University, Milan, Italy
This book subseries publishes reviews and original articles covering all
aspects of cellular and systems neuroscience, particularly neural circuits. It
is also open to contributions on neurological disorders and their molecular and
biochemical basis. A subseries of the highly successful Advances in Experi-
mental Medicine and Biology, it provides a publication vehicle for articles
focusing on the latest research, developments and methods, as well as
opinions and principles. Contributors are experienced neuroscientists and
researchers with extensive knowledge of the embryology, histology, anatomy,
physiology and pharmacology of central and peripheral neurons. It endeavors
to highlight the importance of the correct choice of animal models and the
implementation of the 3Rs principles to translate molecular and cellular
findings of basic research into clinical applications. This series is a valuable
source of state-of-art knowledge and inspiration for future research ideas for a
wide range of researchers and animal care professionals working in the field of
neuroscience.

More information about this subseries at http://www.springer.com/series/


16028
Laura Calzà • Luigi Aloe •
Luciana Giardino
Editors

Recent Advances in NGF


and Related Molecules
The Continuum of the NGF “Saga”
Editors
Laura Calzà Luigi Aloe
University of Bologna IRET Foundation
Ozzano dell’Emilia, Bologna, Italy Ozzano dell’Emilia, Bologna, Italy

Luciana Giardino
University of Bologna
Ozzano dell’Emilia, Bologna, Italy

ISSN 0065-2598 ISSN 2214-8019 (electronic)


Advances in Experimental Medicine and Biology
ISSN 2524-6577 ISSN 2524-6585 (electronic)
Cellular Neuroscience, Neural Circuits and Systems Neuroscience
ISBN 978-3-030-74045-0 ISBN 978-3-030-74046-7 (eBook)
https://doi.org/10.1007/978-3-030-74046-7

# The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Switzerland AG 2021
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher,
whether the whole or part of the material is concerned, specifically the rights of translation,
reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any
other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this
publication does not imply, even in the absence of a specific statement, that such names are
exempt from the relevant protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in
this book are believed to be true and accurate at the date of publication. Neither the publisher nor
the authors or the editors give a warranty, expressed or implied, with respect to the material
contained herein or for any errors or omissions that may have been made. The publisher remains
neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This Springer imprint is published by the registered company Springer Nature Switzerland AG.
The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland
Preface: Rita Levi-Montalcini

Rita Levi-Montalcini received her medical degree and specialization in neu-


rology and psychiatry in 1936 from the Faculty of Medicine at the University
of Turin, Italy. She then entered the Institute of Anatomy as a postgraduate
student in the laboratory of Giuseppe Levi, a well-known anatomist and tutor
of two other future winners of the Nobel Prize in Physiology or Medicine—
Salvatore Luria (who won in 1969) and Renato Dulbecco (who won in 1975).
During her early postgraduate studies, Levi-Montalcini investigated the rela-
tionship between the developing central nervous system (CNS) and its periph-
eral targets, the results of which led her to suggest that the failure of neurons to
thrive in the absence of peripheral target tissues was due to a degenerative
process, rather than a failure of differentiation as had previously been
hypothesized by Victor Hamburger, a renowned embryologist and develop-
mental neuroscientist.
The NGF story began in 1949 when Hamburger showed Levi-Montalcini
the results achieved by one of his postgraduate students, Elmer Bueker.
Bueker had observed that implantation of a small fragment of malignant
mouse tumor into the body wall of 3-day chick embryos caused sensory fibers
to invade the tumor. Bueker hypothesized that this effect was due to the
rapidly expanding tumor allowing the sensory fibers to branch into the
much larger field offered by the neoplastic cells which replaced the embryonic
tissue, a hypothesis which did not convince Rita Levi-Montalcini, who
decided to study the effect more extensively. The results of her reinvestigation
led Levi-Montalcini to the hypothesis that the mouse sarcoma tissues 180 and
37, when transplanted into a chick embryo, produce and release a diffusible
agent which stimulates the growth and differentiation of developing nerve
cells. Using a variety of in vitro methodologies, she then demonstrated that
these tumor tissues produce and release a molecule able to stimulate neuritis
outgrowth from sensory and sympathetic nerve cells, and in 1952, she named
this molecule nerve growth factor (NGF).
In collaboration with Stanley Cohen, co-winner of the Nobel Prize in 1986,
she discovered that NGF is stored in snake venom and in the submaxillary
salivary gland of mice.
During the early 1950s, Levi-Montalcini—then Professor at the Depart-
ment of Zoology at the Washington University of St. Louis—intensified her
in vivo and in vitro studies of NGF on small rodents, while two other
scientists—first Stanley Cohen and later P. U. Angeletti—investigated the

v
vi Preface: Rita Levi-Montalcini

chemical and biochemical properties of NGF, both devising a methodology of


purifying large quantities of NGF from snake venom and from mouse salivary
glands. The availability of large amounts of NGF in these glands provided an
additional opportunity to carry out numerous and collaborative in vivo studies
and to investigate the structural, biochemical, and pharmacological action of
NGF on sympathetic and sensory nerve cells, as well as permitting the
production of anti-NGF antibodies, and to further characterize the effects of
NGF on the development of the peripheral nervous system by injecting this
antibody into newborn rodents. The immune deprivation of NGF via the
exogenous administration of NGF antibodies to these rodents, and the obser-
vation that inhibition of circulating NGF levels results in the death of
NGF-target cells, described in the late 1960s, became known as
immunosympathectomy.
In addition to this continued scientific activity, Levi-Montalcini wrote
several books. The first, In Praise of Imperfection, is an inspiring autobio-
graphical work where she tells of her success, but also of the delays and
distractions affecting researchers, the potential mistakes they can make, and
their tendency to underestimate difficulties. A second volume entitled Il tuo
futuro (“Your future”) is dedicated to young people who during their early
lives often face the need to make vital choices, while her third book, Senza
Olio e contro Vento (“Out of oil and against the wind”), contains ten true
stories of well-known and everyday people, united by the courage and honesty
they showed at very difficult times of their lives. In her most recent book
L’asso nella manica a brandelli (“The ace up a tattered sleeve”), she discusses
the skills and creativity of famous elderly people, and how to maintain and
improve cognitive abilities in later life. From a scientific point of view,
however, her most important book is perhaps the The NGF Saga.
More recently, basic preclinical studies have indicated that purified NGF
has potential clinical application in ocular disorders, such as corneal ulcers,
neurotrophic keratitis, maculopathy, glaucoma, and cutaneous ulcers such as
diabetic and rheumatoid ulcers and pressure sores.
The articles published in this issue are part of a scientific contribution
presented and discussed at the Second International NGF scientific meeting
dedicated to Rita Levi-Montalcini, held in Bologna on June 13–14, 2019, and
organized by Laura Calzà, Luigi Aloe, and Luciana Giardino. A major aim of
this international NGF meeting was to bring attendees up to date on the past,
recent, and ongoing studies in the field of the Italian Nobel Laureate in
Physiology or Medicine who discovered the first neurotrophic factor,
providing neurologists with a powerful tool for investigating the growth,
differentiation, and plasticity of nerve cells, as well as paving the way for
the identification of other specific growth factors for other cell types. As
progress is achieved by moving continuously forward toward further scientific
knowledge, we were happy that the invited speakers presented and discussed
the most recent findings in the field of NGF and other neurotrophic factors,
including the recently discovered recombinant human NGF with clinical
applications for certain human ocular diseases. It is a great pity that Rita
Levi-Montalcini is no longer with us, as she was at our first International NGF
meeting held in Modena in 2002. Finally, we owe a debt of gratitude to all the
Preface: Rita Levi-Montalcini vii

participants who contributed to the success of the NGF meeting conference in


Bologna, to the financial support provided by the IRET Foundation in
Ozzano, Bologna, and to the professional collaboration of all logistical
IRET staff. This meeting was also part of the “Italian Regenerative Medicine
Infrastructure—IRMI” project training program (MIUR,
CTN01_00177_888744 to LC).

Bologna, Italy Laura Calzà


Luigi Aloe
Luciana Giardino
Contents

Part I Basic Science


1 Nerve Growth Factor: The First Molecule of the
Neurotrophin Family . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Luca Lorenzini, Vito Antonio Baldassarro, Agnese Stanzani,
and Luciana Giardino
2 The First and Last Time I Met Rita Levi-Montalcini:
From the Insect Neurotrophic Factor to the Nerve Growth
Factor Clinical Application . . . . . . . . . . . . . . . . . . . . . . . . . . 11
Luigi Aloe
3 NGF Signaling in Endosomes . . . . . . . . . . . . . . . . . . . . . . . . . 19
Naoya Yamashita
4 The NGF Metabolic Pathway: New Opportunities for
Biomarker Research and Drug Target Discovery . . . . . . . . . . 31
Rowan Pentz and M. Florencia Iulita

Part II Development and Regeneration


5 NGF and Endogenous Regeneration: From Embryology
Toward Therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
Vito Antonio Baldassarro, Luca Lorenzini, Andrea Bighinati,
Alessandro Giuliani, Giuseppe Alastra, Micaela Pannella,
Mercedes Fernandez, Luciana Giardino, and Laura Calzà
6 The Versatile Roles of Nerve Growth Factor in Neuronal
Attraction, Odontoblast Differentiation, and Mineral
Deposition in Human Teeth . . . . . . . . . . . . . . . . . . . . . . . . . . 65
Thimios A. Mitsiadis and Pierfrancesco Pagella
7 An Overview of Adult Neurogenesis . . . . . . . . . . . . . . . . . . . . 77
Filipa F. Ribeiro and Sara Xapelli
8 Intervention of Brain-Derived Neurotrophic Factor
and Other Neurotrophins in Adult Neurogenesis . . . . . . . . . . 95
Filipa F. Ribeiro and Sara Xapelli

ix
x Contents

Part III Alzheimer and Central Nervous System


9 Rita Levi-Montalcini, NGF Metabolism in Health and in the
Alzheimer’s Pathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 119
A. Claudio Cuello
10 NGF and the Amyloid Precursor Protein in Alzheimer’s
Disease: From Molecular Players to Neuronal Circuits . . . . . 145
Viviana Triaca, Francesca Ruberti, and Nadia Canu
11 A Review of Techniques for Biodelivery of Nerve Growth
Factor (NGF) to the Brain in Relation to Alzheimer’s
Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 167
Sumonto Mitra, Ruchi Gera, Bengt Linderoth, Göran Lind,
Lars Wahlberg, Per Almqvist, Homira Behbahani,
and Maria Eriksdotter
12 NGF Released from Blood Cells or Collagen Hydrogels
as a Therapeutic Target in Alzheimer’s Disease? . . . . . . . . . . 193
Christian Humpel

Part IV Complex Behaviors


13 When Nerve Growth Factor Met Behavior . . . . . . . . . . . . . . 205
Daniela Santucci, Arianna Racca, and Enrico Alleva
14 Cross Talk of BDNF and GDNF in Spinal Substantia
Gelatinosa (Lamina II): Focus on Circuitry . . . . . . . . . . . . . . 215
Adalberto Merighi

Part V Clinical Medicine


15 BDNF Gene as a Precision Skill of Obesity Management . . . . 233
Helena Marcos-Pasero, Elena Aguilar-Aguilar,
Maria P. Ikonomopoulou, and Viviana Loria-Kohen
16 The Science of Love: State of the Art . . . . . . . . . . . . . . . . . . . 249
Donatella Marazziti, Stefania Palermo, and Federico Mucci
17 NGF and Retinitis Pigmentosa: Structural and Molecular
Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 255
Maria Luisa Rocco, Laura Calzà, and Luigi Aloe
18 NGF in Inflammatory and Neurodegenerative Diseases
of the Eye: New Findings Supporting Neuroprotection and
Proper Tissue Remodeling in Vitreoretinal Disorders . . . . . . 265
Graziana Esposito, Bijorn Omar Balzamino, Luca Bruno,
Andrea Cacciamani, and Alessandra Micera
Contents xi

Part VI Veterinary Medicine


19 Nerve Growth Factor (NGF) and Animal Reproduction . . . . . 277
Margherita Maranesi, Cristiano Boiti, and Massimo Zerani
20 Neurotrophins in the Brain of Teleost Fish: The State
of the Art . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289
Paolo de Girolamo and Livia D’Angelo
Part I
Basic Science
Nerve Growth Factor: The First Molecule
of the Neurotrophin Family 1
Luca Lorenzini, Vito Antonio Baldassarro, Agnese Stanzani,
and Luciana Giardino

Abstract endocrine, and skeletal system, vascular


Neurotrophins (NTs) are molecules regulating districts, inflammation, etc. To date, many
differentiation, maintenance, and functional biological aspects of NTs have been clarified,
plasticity of vertebrate nervous systems. but the new discoveries are still opening new
Nerve growth factor (NGF) was the first to be insights on molecular and cellular mechanisms
identified in the neurotrophin family. The long and systemic effects, also affecting the possi-
scientific history of NTs provided not only ble therapeutic application of NTs. This short
advancement in the neuroscience field but review summarizes the main aspects of NGF
opened new scenarios involving different biology and biochemistry, including the role
body districts in physiological and pathologi- of the NGF precursor molecule, high- and
cal conditions, which include the immune, low-affinity receptors and related intracellular
pathways, and target cells.

L. Lorenzini
Department of Veterinary Medical Sciences, University of
Bologna, Bologna, Italy 1.1 Introduction
Health Science and Technologies Interdepartmental
Center for Industrial Research (HST-ICIR), University of The pioneering work of Rita Levi-Montalcini in
Bologna, Bologna, Italy the first half of the last century led to a revolu-
e-mail: luca.lorenzini8@unibo.it tionary view of neuroembryology, when she first
V. A. Baldassarro · A. Stanzani speculated (in collaboration with Viktor Ham-
Health Science and Technologies Interdepartmental burger) and then proved (in collaboration with
Center for Industrial Research (HST-ICIR), University of
Stanley Cohen) that a target-derived soluble “fac-
Bologna, Bologna, Italy
e-mail: vito.baldassarro2@unibo.it; agnese. tor” was responsible not only for the appropriate
stanzani2@unibo.it development of sensory and sympathetic innerva-
L. Giardino (*) tion of the chicken embryo but also for cell num-
Department of Veterinary Medical Sciences, University of ber control and, therefore, for the appropriate
Bologna, Bologna, Italy shaping of neural pathways during development
Health Science and Technologies Interdepartmental (Levi-Montalcini 1997). This soluble factor was
Center for Industrial Research (HST-ICIR), University of then isolated in 1954 and initially named “nerve
Bologna, Bologna, Italy
growth-promoting factor.” Subsequently
IRET Foundation, Bologna, Italy abbreviated to nerve growth factor (NGF), it led
e-mail: luciana.giardino@unibo.it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 3


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_1
4 L. Lorenzini et al.

to the neurotrophic theory, postulating that NGF like domains. The NGF binding domain is located
(and subsequently all neurotrophins—NTs) is in the Trk-d5(Ig2) domain. According to a classi-
responsible for regulating the balance between cal view, the ligand-receptor complex is
neuronal survival and neuronal death, its absence internalized in endosomes after binding to Trk
or withdrawal being the causal link to induce receptors and retrogradely transported to the cell
neuronal death (Ichim et al. 2012). body regulating pro-apoptotic and anti-apoptotic
NGF was the first molecule of the gene expression (Mok et al. 2009). Target-
“neurotrophin family” to be identified (Franco derived NGF is also responsible for the trophic
et al. 2020): brain-derived neurotrophic factor support of selected neural populations in the cen-
(BDNF) was discovered a few years later, with tral nervous system (CNS) during adulthood, the
neurotrophin-3 (NT-3) and neurotrophin-4 cholinergic basal forebrain neurons.
(NT-4) subsequently included in the family In addition, all NTs bind to the low-affinity
(Hennigan et al. 2007). p75 receptor (p75NTR), a member of the TNF
NTs are synthesized in the endoplasmic retic- receptor superfamily (Huang and Reichardt
ulum as precursor proteins of around 250 amino 2001). The p75NTR receptor is a monomer and
acids (proNT, 35 kDa) which associate in a consists of three parts: a cysteine-rich extracellu-
homodimer. ProNGF is subsequently processed lar domain involved in binding to NTs, a trans-
by different proteases (e.g., furin), the presence of membrane region containing the cleavage site for
a 103-residue N-terminal pro-part in proNGF gamma-secretase, and an intracellular domain
constituting the difference between the primary containing what is known as the death domain,
structure of NGF and proNGF (Trabjerg et al. responsible for the receptor’s pro-apoptotic activ-
2017). The mature forms (12–15 kDa) then end ity. In some cells, NT binding to p75NTR triggers
up in two types of vesicles within the Golgi a programmed cell death process via the c-Jun
network, belonging to the constitutive or N-terminal kinase (JNK) or caspase signaling
regulated secretory pathways (Costa et al. 2018). pathways (Chao 2003).
In the former, vesicles release their cargos when Over the years, many more molecular and
they reach the plasma membrane, whereas in the cellular aspects of NT biology and mechanisms
regulated mode, release is calcium-dependent. of action have been discovered and clarified,
The cellular action of the neurotrophins, i.e., including (1) the biological activity of proNTs,
the biological effect of each NT, is mediated (2) the multiple transmembrane pathways
through the activation of high- and low-affinity activated by Trk receptors, and (3) the pleiotropic
receptors. High-affinity receptors belong to the nature of NTs, which act on many different cell
tropomyosin-related kinase (Trk) family of recep- types also in adulthood. In this short review, we
tor tyrosine kinases, NGF being the specific will review these aspects with regard to NGF.
ligand for TrkA, BDNF for TrkB, and NT-3/
NT-4 for TrkC. NGF specifically binds to TrkA,
while BDNF preferentially binds to TrkB. NT-3 1.2 NGF and proNGF: Who Is Doing
preferentially binds to TrkC but can also activate What?
TrkA and TrkB, while NT-4 preferentially binds
to TrkB (Skaper 2012). TrkA has five extracellu- NGF is a product of a single gene found on
lar binding domains that connect the extracellular chromosome 1 and is produced according to the
portion of the receptor to the single transmem- aforementioned process of synthesis and release
brane domain and a juxtamembrane intracellular from proNGF. In its mature form, NGF is assem-
region that is connected to the kinase domain. The bled in a non-covalent homodimer, denoted by a
first (Trk-d1) is a leucine-rich region and is ring structure formed by two disulfide bridges
flanked by two cysteine-rich domains (Trk-d2 penetrated by a third disulfide bridge (Trabjerg
and Trk-d3). The fourth and fifth domains et al. 2017). However, proNGF is not an inactive
(Trk-d4 and Trk-d5) are immunoglobulin (Ig)- precursor but takes active part in the biological
1 Nerve Growth Factor: The First Molecule of the Neurotrophin Family 5

activity of this NT, often producing opposite co-receptor (Skeldal et al. 2012), and is a required
effects to the mature NGF form. element for proNGF cell death signaling. More-
Substantial contributions clarifying the over, while most of the data excludes the possi-
biological role of proNGF have come from the bility of Trk activation by proNTs, recent studies
study of the cholinergic neurons of the basal suggest that proNGF can bind TrkA receptors
forebrain in both human and animal brains. with low affinity (5.4 times lower than mature
Observations on these neurons have NGF). Thus, the biological effect of NGF must
demonstrated that degeneration in Alzheimer’s be stated according to a number of concurrent
disease, or in animal models such as the Down biochemical, cell-specific players.
syndrome mice Ts65Dn, is related to a defect in
the retrograde axonal transport of proNGF. This
is associated with a deficit in the NGF metabolic 1.3 TrkA- and p75NTR-Mediated
pathway responsible for processing proNGF to Intracellular Pathways
mature NGF (Iulita and Cuello 2014; Costa
et al. 2018; Fahnestock and Shekari 2019). The By interaction with TrkA, NGF regulates many
ratio between pro- and mature forms of NGF, neuronal functions, including cell survival and
responsible for determining neuronal fate, there- differentiation, axonal and dendritic growth, and
fore derives from complex biochemical pathways synaptic formation and plasticity (Fig. 1.1).
including not only intracellular synthesis and Ligand-receptor binding induces phosphorylation
release but also intracellular distribution, extracel- of specific tyrosine residues, and the docking site
lular proteases, and regulation of high- and recruits pathway-activating proteins for Ras, Rac,
low-affinity receptor expression. For example, PI3 kinase, and PLC-γ1 and their downstream
the proNGF and NGF homodimers have a differ- effectors (Franco et al. 2020). These include
ent distribution within the neuron, with proNGF mitogen-activated protein (MAP) kinase cascades
localized in the cell body and mature NGF in the (stimulated by Ras), Akt stimulation by PI3, and
cell body and neuronal processes. Moreover, the PLC-γ1-dependent generation of IP3 and
cleavage site of proNGF, whether intracellular or diacylglycerol (DAG), which results in the mobi-
extracellular, is still disputed, and extracellular lization of Ca2+ stores and the activation of Ca2+
matrix enzymes, including MMP7 and TIMP1, and DAG-regulated protein kinases (Reichardt
seem to promote NGF biochemical maturation 2006).
(Costa et al. 2018). Transmembrane pathway activation does not
In the meantime, proNT and proNGF receptors obscure gene transcription regulation through ret-
continue to be a matter of debate. Since the origi- rograde transport of the NT-Trk endosomal com-
nal demonstration that proNTs are secreted and plex. This process is regulated by ubiquitination,
cleaved extracellularly and bind to receptors in involves Shc, and is required for persistent acti-
order to produce their biological effects (Lee et al. vation of the MAP kinase cascade. Lastly, the
2001), the p75NTR/sortilin high-affinity receptor recruitment of distinct populations of endosomes,
complex has been identified as responsible for such as the recruitment of the receptor in the
mediating proNGF-induced neuronal death by appropriate cell compartment, i.e., the plasma
apoptosis (Nykjaer et al. 2004; Teng et al. membrane or the nucleus, may represent further
2010). p75NTR expression is high during develop- levels for control of NT biological activity.
ment, while very few neural populations express The p75NTR receptor also mediates the activa-
this receptor in the mature brain. However, recent tion of several signaling pathways, including
evidence indicates that p75NTR expression can be Traf6, NT receptor-interacting factor (NRIF),
induced in other neurons by lesion stimuli. melanoma-associated antigen (MAGE), p75NTR-
Sortilin is a member of the VPS10 domain trans- interacting MAGE homolog (NRAGE), Schwann
membrane receptor family. It creates heterodimer cell factor 1 (SC1), RhoGDI, and other proteins
complexes with p75NTR, thus functioning as (Fig. 1.2). A major pathway activated by p75NTR
6 L. Lorenzini et al.

Fig. 1.1 NGF/TrkA signaling transduction pathways. facilitating the trafficking and leading to neuronal survival
TrkA mediates its action by phosphorylation, activating and differentiation. Rabs are involved in biogenesis and
MAPK, PI3K, and PLC-γ1 pathways, resulting in activa- regulation of the transport of the endosomes.
tion of gene expression, neuronal survival, and neurite Abbreviations: Akt protein kinase B, Bcl2 B-cell lym-
outgrowth. Activation of Ras activates MAP kinase sig- phoma 2, C1 cysteine cluster 1, C2 cysteine cluster
naling cascade, promoting neuronal differentiation and 2, Erk extracellular signal-regulated kinases, Frs2 fibro-
neurite outgrowth. Ras and Gab1 activate also PI3, blast growth factor receptor substrate 2, GAB1/2 growth
which promotes survival pathway activation and neuronal factor receptor-bound protein 2-associated protein 1, Gbr2
growth. PLC-γ1 activation leads to Ca2+- and protein growth factor receptor-bound protein 2, IAPs inhibitors of
kinase C-regulated pathway activation promoting synaptic apoptosis, Ig1 immunoglobulin-like domain 1, Ig2
plasticity. TrkA acts also in endosomes moving through immunoglobulin-like domain 2, LRR1–3 extracellular
the axon on microtubules by dynein binding. NGF remains ligand-binding domain comprising leucine-rich motif,
bound to TrkA keeping it activated, sustaining signals MAPK mitogen-activated protein kinase, MEKK MAP
1 Nerve Growth Factor: The First Molecule of the Neurotrophin Family 7

is the c-Jun kinase signaling cascade, resulting in are largely suppressed as a result of Trk
p53/bax-mediated apoptosis. Notably, the recruit- receptor-mediated signaling when mature NTs
ment of these pathways seems to occur via the bind to Trk receptors (Reichardt 2006).
p75NTR death domain (DD). This 10 kDa glob-
ular protein is present in many members of the
tumor necrosis factor receptor superfamily and 1.4 NGF Target Cells
can recruit several interactor proteins which act
as pathway activators (Yuan et al. 2019). Currently, NGF remains the best-characterized
The receptor p75NTR also promotes NF-κB- neurotrophic factor for its role in regulating sym-
dependent neuronal survival via p62 and the pathetic and sensory neuron differentiation, in
kinase activity of IRAK. This mechanism, not phenotype maintenance and protection of target
yet fully understood, is TrkA-independent and neuronal populations, in the sensitivity of periph-
has been demonstrated in several cell types eral nerve fibers to heat and pain, and in
including Schwann cells and trigeminal and hip- mediating physiological and abnormal pain per-
pocampal neurons (Franco et al. 2020). Other ception. In the CNS, NGF-producing and NGF
pathways activated by NTs binding to p75NTR target cells are present in the basal forebrain,
include acidic sphingomyelinase, which results striatum, thalamus, and several cortical regions
in the generation of ceramide, and the Rho family and in dentate granule cells in the hippocampus
GTPase activity, which regulates cytoskeleton as subpopulations of GABAergic interneurons.
assembly and stability and neurite outgrowth. NGF expression in the hippocampus is regulated
The role of the p75NTR intracellular domain by neuronal activity, being increased by activa-
(ICD), produced by cleavage of the receptor by tion of glutamatergic and cholinergic neurotrans-
gamma-secretase, is also unclear. The specific mission and decreased by GABAergic
29-amino acid peptide derived from the intracel- transmission (Sofroniew et al. 2001).
lular cleavage of p75NTR interacts with and NGF is also produced by glial cells, as
potentiates the binding of NGF to TrkA- astrocytes and microglia, and NGF expression in
expressing cells. Examples are the potentiation both cell types is markedly upregulated by local
of the effect of NGF on neurite outgrowth in tissue injury, inflammation, and cytokines. This
sympathetic neurons as a result of enhanced was observed in the periinfarct area in ischemic
Erk1/2 and Akt signaling (Matusica et al. 2013) cortex after transient stroke, after mechanical
or the increase in the expression of vascular endo- injury in the hippocampus and in the cortex
thelial growth factor, for example, after retinal surrounding the wound, and in Parkinson’s dis-
hypoxia via HIF1α (Le Moan et al. 2011). ease models at lesioned striatum (Pöyhönen et al.
Lastly, TrkA and p75NTR can signal synergis- 2019).
tically, antagonistically, or independently of each These major physiological roles have also
other in different cellular contexts (Lu et al. prompted studies of the possible role of NGF
2005), meaning that the final fate produced by defects or signaling dysfunctions in pathological
NGF or proNGF exposure is cell-specific. In a conditions such as metabolic diseases—periph-
general sense, most studies suggest that in the eral neuropathy in diabetes mellitus, for exam-
case of TrkA and p75NTR co-expression in the ple—or in infectious and inflammatory diseases,
same cell, the pro-apoptotic signals of p75NTR due to the role of the neurotrophin in

ä
Fig. 1.1 (continued) kinase kinase kinase, NGF nerve activating system, Shc SH2 domain and collagen homolo-
growth factor, PDKs pyruvate dehydrogenase kinases, gous region, SOS mammalian son of sevenless, TrkA
PI3K phosphatidylinositol 3 kinase, PLC-γ1 phospholi- tropomyosin receptor kinase A
pase Cγ, Raf rapid accelerated fibrosarcoma, Ras reticular
8 L. Lorenzini et al.

Fig. 1.2 NGF/p75NTR signaling transduction pathways. interleukin-1 receptor-associated kinase, JNK c-Jun N-ter-
P75NTR signaling may induce survival or apoptosis minal kinase, NADE p75NTR-associated death executor,
response, depending on the cell type and activated cas- NF-κB nuclear factor kappa-light-chain-enhancer of
cade. The receptor interacts with different proteins activated B cells, NGF nerve growth factor, NIK
(TRAF6, RhoA, NRAGE, SC-1, NRIF) regulating gene NF-kappa-B-inducing kinase, NRAGE neurotrophin
expression, cell cycle, apoptosis, mitogenic responses, and receptor-interacting MAGE homolog, NRIF neurotrophin
axonal growth. It activates three main pathways, NF-κB, receptor-interacting factor, p62 sequestosome 1, p75NTR
c-Jun, and Rho, which promotes, respectively, cell sur- neurotrophin low-affinity receptor, RhoA Ras homolog
vival, apoptosis, and growth cone motility. The family member A, RhoGDI Rho GDP-dissociation inhibi-
pro-apoptotic c-Jun effect can be inhibited by Ras-PI3K tor, RIP2 receptor-interacting-serine/threonine-protein
pathway activated by TrkA. Abbreviations: Ask1 apopto- kinase 2, SC-1 Schwann cell factor 1, TRAF6 TNF
sis signal-regulating kinase 1, Cd42 glycoprotein lb, receptor-associated factor 6
CR1–4 complement receptor 1–4, IKK IκB kinase, IRAK

inflammatory and immune cells (Aarão et al. inflammatory cells or epithelial cells, leading to
2018). In fact, NGF production and biological the general definition of NGF as a “pleiotropic”
actions have been described in many molecule (Aloe and Calzà 2004; Micera et al.
non-nervous cell types, such as immune 2007).
1 Nerve Growth Factor: The First Molecule of the Neurotrophin Family 9

NGF high- and low-affinity receptors are pres- part in a process known as “tumor neurogenesis”
ent in not only nerve cells and CNS tumors but in (Griffin et al. 2018). This hypothesis refers to the
immune cells such as macrophages, lymphocytes, major role of peripheral innervation in the homeo-
and mast cells. These cells synthesize, store, and static regulation of the microenvironment and its
secrete NGF, suggesting a possible role for NGF adaptation in pathological conditions, suggesting
in the regulation of immune responses during that NGF may play a role in cancer pain.
inflammatory, infectious, and autoimmune pro- This evidence brings the research full circle to
cesses (Aarão et al. 2018). the discoveries made by Rita Levi-Montalcini, a
Other tissues and cell types expressing NGF mouse sarcoma being the tissue from which NGF
and related receptors include the skin (e.g., was originally isolated (Cohen et al. 1954).
keratinocytes, fibroblasts, and melanocytes
(Gostynska et al. 2020)), the vascular system
(endothelial cells (Gostynska et al. 2020)), the
References
salivary (e.g., submandibular) glands, and various
endocrine organs/cells, such as the pituitary, the Aarão TL d S, de Sousa JR, ASC F, LFM F, JAS Q (2018)
thyroid, and the endocrine elements in the testes Nerve growth factor and pathogenesis of leprosy:
and ovaries. Most of these cells continue to pro- review and update. Front Immunol 9:939
Aloe L, Calzà L (2004) NGF and related molecules in
duce NGF during adulthood and modulate NGF
health and disease: preface. Prog Brain Res 146:1–544
production in response to stimuli (Sofroniew et al. Chao MV (2003) Neurotrophins and their receptors: a
2001). convergence point for many signalling pathways. Nat
This process also leads to local events, includ- Rev Neurosci 4:299–309. https://doi.org/10.1038/
nrn1078
ing activation-induced histamine release, and an
Cohen S, Levi-Montalcini R, Hamburger V (1954) A
increase in the number of mast cells and other nerve growth-stimulating factor isolated from
immune system cells (Aarão et al. 2018). SARCOM as 37 and 180. Proc Natl Acad Sci
Several inflammatory, infectious, and autoim- 40:1014–1018. https://doi.org/10.1073/pnas.40.10.
1014
mune diseases also lead to an altered NGF physi-
Costa RO, Perestrelo T, Almeida RD (2018) PROneuro-
ology, as indicated by altered plasma levels and trophins and CONSequences. Mol Neurobiol
by the presence of anti-NGF antibody levels in 55:2934–2951
patients with rheumatoid arthritis, systemic lupus Fahnestock M, Shekari A (2019) ProNGF and
neurodegeneration in Alzheimer’s disease. Front
erythematosus, and thyroiditis (Skaper 2017;
Neurosci 13:129
Aarão et al. 2018). Franco ML, Comaposada-Baró R, Vilar M (2020)
Finally, a special mention should be reserved Neurotrophins and Neurotrophin receptors. In: Hor-
for the role of NGF, other neurotrophins, and monal signaling in biology and medicine. Elsevier,
Amsterdam, pp 83–106
related receptors in cancer. The emerging concept
Gostynska N, Pannella M, Rocco ML, Giardino L, Aloe L,
of “nerve dependency” in oncology has gained Calzà L (2020) The pleiotropic molecule NGF
consensus in recent years thanks to a large body regulates the in vitro properties of fibroblasts,
of data demonstrating that NGF and related keratinocytes, and endothelial cells: implications for
wound healing. Am J Physiol – Cell Physiol 318:
receptors can be expressed in human tumors,
C360–C371. https://doi.org/10.1152/ajpcell.00180.
including ovarian, breast, lung, pancreatic, skin, 2019
liver, gastric, and thyroid malignancies. In these Griffin N, Faulkner S, Jobling P, Hondermarck H (2018)
context, neurotrophins can stimulate cancer cell Targeting neurotrophin signaling in cancer: the renais-
sance. Pharmacol Res 135:12–17
growth (Griffin et al. 2018), also leading to the
Hennigan A, O’Callaghan RM, Kelly ÁM (2007)
hypothesis of using the Trk receptor expression Neurotrophins and their receptors: roles in plasticity,
level as a prognostic marker. A more recent neurodegeneration and neuroprotection. Biochem Soc
hypothesis, however, has suggested that nerves Trans 35(2):424–427. Portland Press
Huang EJ, Reichardt LF (2001) Neurotrophins: roles in
infiltrated in the tumor microenvironment secrete
neuronal development and function. Annu Rev
neurotransmitters, which can stimulate both Neurosci 24:677–736. https://doi.org/10.1146/
growth of tumor cells and angiogenesis, taking annurev.neuro.24.1.677
10 L. Lorenzini et al.

Ichim G, Tauszig-Delamasure S, Mehlen P (2012) Nature 427:843–848. https://doi.org/10.1038/


Neurotrophins and cell death. Exp Cell Res nature02319
318:1221–1228 Pöyhönen S, Er S, Domanskyi A, Airavaara M (2019)
Iulita MF, Cuello AC (2014) Nerve growth factor meta- Effects of neurotrophic factors in glial cells in the
bolic dysfunction in Alzheimer’s disease and down central nervous system: expression and properties in
syndrome. Trends Pharmacol Sci 35:338–348. https:// neurodegeneration and injury. Front Physiol 10:486
doi.org/10.1016/j.tips.2014.04.010 Reichardt LF (2006) Neurotrophin-regulated signalling
Le Moan N, Houslay DM, Christian F, Houslay MD, pathways. Philos Trans R Soc B Biol Sci
Akassoglou K (2011) Oxygen-dependent cleavage of 361:1545–1564
the p75 neurotrophin receptor triggers stabilization of Skaper SD (2012) The neurotrophin family of
HIF-1α. Mol Cell 44:476–490. https://doi.org/10. neurotrophic factors: an overview. Methods Mol Biol
1016/j.molcel.2011.08.033 846:1–12
Lee R, Kermani P, Teng KK, Hempstead BL (2001) Reg- Skaper SD (2017) Nerve growth factor: a neuroimmune
ulation of cell survival by secreted proneurotrophins. crosstalk mediator for all seasons. Immunology
Science (80-) 294:1945–1948. https://doi.org/10.1126/ 151:1–15
science.1065057 Skeldal S, Sykes AM, Glerup S, Matusica D, Palstra N,
Levi-Montalcini R (1997) The Saga of the nerve growth Autio H, Boskovic Z, Madsen P, Castrén E, Nykjaer A,
factor. World Scientific, Singapore Coulson EJ (2012) Mapping of the interaction site
Lu B, Pang PT, Woo NH (2005) The yin and yang of between sortilin and the p75 neurotrophin receptor
neurotrophin action. Nat Rev Neurosci 6:603–614 reveals a regulatory role for the sortilin intracellular
Matusica D, Skeldal S, Sykes AM, Palstra N, Sharma A, domain in p75 neurotrophin receptor shedding and
Coulson EJ (2013) An intracellular domain fragment apoptosis. J Biol Chem 287:43798–43809. https://doi.
of the p75 neurotrophin receptor (p75NTR) enhances org/10.1074/jbc.M112.374710
tropomyosin receptor kinase a(TrkA) receptor func- Sofroniew MV, Howe CL, Mobley WC (2001) Nerve
tion. J Biol Chem 288:11144–11154. https://doi.org/ growth factor signaling, neuroprotection, and neural
10.1074/jbc.M112.436469 repair. Annu Rev Neurosci 24:1217–1281. https://doi.
Micera A, Lambiase A, Stampachiacchiere B, Bonini S, org/10.1146/annurev.neuro.24.1.1217
Bonini S, Levi-Schaffer F (2007) Nerve growth factor Teng KK, Felice S, Kim T, Hempstead BL (2010) Under-
and tissue repair remodeling: trkANGFR and p75NTR, standing proneurotrophin actions: recent advances and
two receptors one fate. Cytokine Growth Factor Rev challenges. Dev Neurobiol 70:350–359
18:245–256 Trabjerg E, Kartberg F, Christensen S, Rand KD (2017)
Mok SA, Lund K, Campenot RB (2009) A retrograde Conformational characterization of nerve growth fac-
apoptotic signal originating in NGF-deprived distal tor- reveals that its regulatory pro-part domain
axons of rat sympathetic neurons in compartmented stabilizes three loop regions in its mature part. J Biol
cultures. Cell Res 19:546–560. https://doi.org/10. Chem 292:16665–16676. https://doi.org/10.1074/jbc.
1038/cr.2009.11 M117.803320
Nykjaer A, Lee R, Teng KK, Jansen P, Madsen P, Nielsen Yuan W, Ibáñez CF, Lin Z (2019) Death domain of p75
MS, Jacobsen C, Kliemannel M, Schwarz E, Willnow neurotrophin receptor: a structural perspective on an
TE, Hempstead BL, Petersen CM (2004) Sortilin is intracellular signalling hub. Biol Rev 94:1282–1293.
essential for proNGF-induced neuronal cell death. https://doi.org/10.1111/brv.12502
The First and Last Time I Met Rita
Levi-Montalcini: From the Insect 2
Neurotrophic Factor to the Nerve
Growth Factor Clinical Application
RLM and the Discovery of Two Neurotrophic Factors

Luigi Aloe

Abstract cardiometabolic diseases; and finally (4) NGF


The neurotrophic factor nerve growth factor potential clinical application.
(NGF) has been discovered in the 1950s by
Rita Levi-Montalcini, first in a neoplastic tis-
sue and, later, in the mouse salivary gland (see
1A). Levi-Montalcini characterized its action 2.1 Introduction
in the sensory and sympathetic neurons
(1B) and, a few years later, in central nervous, Rita Levi-Montalcini (RLM) received her medi-
endocrine, and immune systems. Nerve cal degree in 1936 from the University of Turin
growth factor plays its trophic role both during and entered in the Institute of Anatomy as a
development and in adulthood, ensuring the postgraduate student in the laboratory of
maintenance of phenotypic and functional Giuseppe Levi, a well-known anatomist and men-
characteristic of several populations of tor of other two future Nobel Prize winners in
neurons as well as immune cells. The aim of Physiology or Medicine, Salvatore Luria, in
the present overview is to describe my per- 1969, and Renato Dulbecco in 1975. During the
sonal scientific and human experiences work- early postgraduate years, RLM studied the rela-
ing with Rita Levi-Montalcini for over tionship between the developing nerve system
45 years, first at Washington University in and peripheral targets and observed that many
St. Louis, Missouri, USA, searching (1) the sensory neurons died during normal development
invertebrate neurotrophic factor in the and that the limb bud extirpation in chick
cockroaches and, later, at the Institute of Neu- embryos caused an increase of cell death. These
robiology of the National Research Council observations led to the hypothesis that failure of
(CNR) in Rome studying (2) the role of NGF neurons to thrive in the absence of peripheral
for various neuronal and non-neuronal target was because of a degenerative process
functions; (3) the potential involvement of rather than a failure of differentiation. In 1946,
NGF in the pathobiology of human cutaneous, Viktor Hamburger, a well-known and famous
ocular, neurodegenerative, and neuroembryologist working at the Department
of Zoology, Washington University in St. Louis,
Missouri, USA, invited RLM in his laboratory.
L. Aloe (*) Working with Hamburger, she demonstrated that
IRET Foundation, Tecnopolo-Bologna “Rita Levi- nerve cells die during the course of normal devel-
Montalcini”, Bologna, Italy opment and that limb bud extirpation causes an
e-mail: luigi.aloe@cnr.it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 11


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_2
12 L. Aloe

increase of neuronal death, suggesting the international scientific community for her studies
hypothesis that the developing neurons depend on NGF. At that time, I personally did not know
on feedback signals from the peripheral tissues almost anything about NGF studies, but only that
and that neuronal target provides a specific mole- she was a famous international scientist. Only
cule required for neuronal survival, growth, and later, I believe I understood her motivation. She
differentiation. In 1950, she discovered the first isolated and purified the molecule with her grad-
neurotrophic factor, the nerve growth factor uate student Stanley Cohen and demonstrated that
(NGF), in a mouse sarcoma tissue and later in it was able to promote growth and differentiation
the snake venom and mouse submaxillary gland of sensory and sympathetic neurons in vivo and
(Fig. 2.1a). In collaboration with a young bio- in vitro and that the administration of an antibody
chemist, Stanley Cohen (who discovered the epi- against the purified NGF in developing rodents
dermal growth factor, co-winner of the Nobel caused immunosympathectomy, but no lethal
Prize in 1986), RLM developed a series of exper- actions, nor an effect on their lifespan. Moreover,
imental approaches to purify the factor from the after removing the largest mouse source of NGF,
mouse salivary gland that produces and stores the salivary gland, she observed neither adverse
high amounts of NGF that was effective in nor lethal effect. Finally, she tested the action of
stimulating the growth and differentiation of neu- the purified molecule in invertebrates, particularly
ritis outgrowth from sensory and sympathetic in young and adult insects, and, again, found no
neurons, both in vivo and in vitro (Fig. 2.1b). action on their nervous system. All these negative
Using the NGF purified from this source and observations led her to believe that NGF might
antibodies against NGF, they then demonstrated not have any potential clinical development and
the functional biological significance of NGF on future applications and hypothesized that insects
mammals’ peripheral sensory and sympathetic should produce their own neurotrophic factors, as
nerve cells (Cohen 1960; Levi-Montalcini 1952; mammals. Convinced about this hypothesis,
Levi-Montalcini 1964; Levi-Montalcini and RLM decided to take a sabbatical leave from the
Booker 1960; Levi-Montalcini and Hamburger NGF studies and to invest her time searching for
1951). insect neurotropic factors.

2.2 How I Met Rita Levi-Montalcini 2.3 Flight to St. Louis, MO, USA

In the early June 1965, I received a letter from In May 1966, I took the airplane for St. Louis,
Levi-Montalcini from St. Louis, MO, USA, for USA, with RLM. To carry studies on insects’
fixing an interview in Rome during her next trip nervous system, she selected adult developing
to Rome. During the interview, she briefly told cockroaches because of their long lifespan and
me: “I was told that you are an expert of taking the properties to lay bean-like eggs every week
care and raising Grasshoppers and I am sure that containing about 12–14 embryos having all the
you would be also able to take care and raise same stage of development. Once I raised a stable
cockroaches (Periplaneta Americana) in my Lab- colony of cockroaches and collected a sufficient
oratory in St. Louis.” She then continued: “I number of eggs and embryos, I started with RLM
would give you 400 dollars a month for six to remove several different embryo tissues,
months and I need an answer within two weeks.” making tissue extracts and searching for the
A week later, I decided to accept and traveled unknown new insect neurotropic factor. Less
with her to St. Louis 2 weeks later. The 6 months than 1 year later, we obtained the first positive
became then nearly 45 years. Why did she decide and novel result. We identified an NGF-like
to leave the study of NGF? I was not told why she neurotrophic factor in the digestive tract of
decided stopping her studies on NGF, in spite of cockroaches (Fig. 2.2a) with properties of pro-
she was already well-known among the moting outgrowth and differentiation of insect
2 The First and Last Time I Met Rita Levi-Montalcini: From the Insect. . . 13

Fig. 2.1 (a) Photomicrograph of immunohistochemical- developing rats cultured for 24 h in the presence of
stained section of adult mouse salivary gland storing and 10 nanograms of purified NGF showing neurite outgrowth
releasing the nerve growth factor. Magnification: 95X. (b) induced by NGF. Magnification: 45X
Photomicrograph of dissociated sympathetic neurons of

brain nerve cells in developing and adult and molecular analyses, the commercial cost of
cockroaches (Fig. 2.2b). These findings were cockroaches became quite high, and the recombi-
published (Aloe and Levi-Montalcini 1972a, b). nant methodologies were not yet available. There-
Unfortunately, the amounts and release of the fore, she decided not to invest further time on
factor in the digestive tract of this insect were insect studies.
not sufficient for the molecule to be purified in
large amounts for further structural, biochemical,

Fig. 2.2 (a) Photomicrograph of adult cockroach intes- ganglia of developing cockroaches in culture in the pres-
tine producing and releasing insect neurotrophic factor. ence of cockroach intestine extract inducing neurite
Magnification: 25X. (b) Photomicrograph showing outgrowth after 48 h in vitro. Magnification: 200X
14 L. Aloe

2.4 In 1972 RLM Decided 2.5 NGF and the Central Nervous
to Organize Her Return System
in Rome
In 1984, Siler and Schwab published the first
I remained in St. Louis to continue the studies of evidence that NGF was expressed in the central
the action of insect growth factor on the intestine nervous system of rodents. They demonstrated
motility and to explore the role of NGF on rodent that radiolabeled NGF, injected in very small
peripheral neuropathies, behavioral response, and quantities into the frontal or dorsal anterior occip-
potential pharmacotherapeutic properties. These ital cortex of adult rats, was specifically taken up
studies revealed that NGF was able to prevent the and transported retrogradely to large, presumably
peripheral neuropathies induced by chemical cholinergic neurons in the region of the nucleus
drugs and by surgical axotomy. These findings basalis (lateral preoptic nucleus, anterior lateral
reinforced the hypothesis that NGF might have hypothalamic nucleus, substantia innominata,
indeed a potential clinical future. Furthermore, ventral globus pallidus, and internal capsule), as
other findings revealed that the NGF not only revealed by light microscopic autoradiography.
prevented peripheral neuropathies but also pro- They thus suggested that NGF might be
moted the growth, differentiation, and degranula- implicated in the protection of the brain choliner-
tion of mast cells. Moreover, we showed that mast gic neurons that degenerated in aging and mem-
cells had the properties of synthesizing, storing, ory deficits (Aloe et al. 1986). Few years later, the
and releasing NGF, which could exert profound studies published by Hefti showed that NGF
effects of autoimmune action, neuroimmune receptors were localized in the rat forebrain and
interactions, tissue inflammation, and modulation that NGF promoted the survival of these neurons,
of lymphocytes’ memory abilities (Levi- both in vivo and in vitro (Spillantini et al. 1989).
Montalcini et al. 1975). Furthermore, in vivo Other studies, published a few years later,
administration of neutralizing anti-NGF demonstrated that intraventricular injections of
antibodies caused marked reduction in the titer NGF attenuated the degeneration of these
of specific IgGs in mice immunized with the neurons, suggesting that NGF might be able to
tetanus toxoid, nitrophen. Together these attenuate and/or prevent the degeneration of the
observations indicated that NGF played a critical forebrain cholinergic neurons involved in
role as an autocrine survival factor for memory B Alzheimer’s disease (Hefti and Will 1987; Seiler
lymphocytes. and Schwab 1984). The role of NGF in physiopa-
In Rome, collaborating with Enrico Alleva thology of brain nerve cells was subsequently
from the Italian National Institute of Health, we extended by the studies published by Laura
found that the action of NGF was involved in Calzà in collaboration with RLM (Calza et al.
aggressive behavior, enhancing circulating NGF 2001; Calza et al. 2003; Fischer et al. 1987;
levels, and promotes its synthesis in forebrain and Giardino et al. 2002; Tuszynski et al. 1991).
hypothalamic areas. These findings and other In 1995, I was invited by Dr George
numerous observations suggested that the Chaldakov to present a lecture on NGF and mast
increase of NGF levels preceded the increase of cells at the Biomedical Forum organized by the
plasma cortisol and adrenocorticotropic hormone Medical University of Varna, Bulgaria. Since
and could be involved in the activation of cells of then, we had a fruitful scientific collaboration on
the immune system, most probably associated the possible significance of NGF and brain-
with homeostatic (homeodynamic, a term pro- derived neurotrophic factor (BDNF) in multiple
posed by RLM) mechanisms, as previously biological phenomena, ranging from
shown for stressed rodents (Aloe and Levi- neurotrophic, immunotrophic, to metabotropic
Montalcini 1977; Torcia et al. 1996). effects. We have for the first time reported data
that demonstrated significantly reduced local
2 The First and Last Time I Met Rita Levi-Montalcini: From the Insect. . . 15

and/or blood circulating levels of the two then followed up for up to 12 months. Corneal
neurotrophins in human cardiometabolic dis- integrity and sensitivity were maintained during
eases, including coronary atherosclerosis and the follow-up period (Lambiase et al. 1998). Best-
the metabolic syndrome and acute coronary corrected visual acuity increased progressively
syndromes. We also suggested that TrkA and during treatment and follow-up in all patients,
TrkB agonists might have therapeutic benefit in without systemic or local side effects of
patients with these conditions. Further, we have treatment.
demonstrated that both NGF and BDNF were
secreted by adipose tissue cells.
2.8 NGF and Glaucoma
2.6 Studies on NGF and Clinical One key event leading to glaucoma (GL) is the
Potentiality elevated intraocular pressure. However, at that
time, whether the ocular application of NGF
The first study to test the role of NGF on visual could prevent or reduce the damage to retinal
cells was carried out with intravitreal administra- ganglion cells (RGCs) was unknown. To test the
tion of the factor on a mouse strain C3H, affected action of NGF, we induced GL in adult rats by the
by inherited retinitis pigmentosa, which is injection of hypertonic saline into the episcleral
characterized by a single gene mutation that vein of the right eye and used the left eye as
leads to photoreceptor degeneration. It was control. The injection led to a progressive degen-
found that the intraocular injection of NGF eration of the RGCs, with a loss of nearly 40% of
induced a significant increase in the thickness of these cells after 7 weeks of treatment. This cellu-
the retinal compared to controls, indicating that lar loss was associated with the downregulation
NGF administration protected and/or reduced the of NGF and NGF receptor expression in the retina
degeneration of retinal cells (Lambiase and Aloe and the glaucomatous eye, but ocular treatment
1996). with NGF significantly reduced the deficit
induced by GL. These results unequivocally
revealed that NGF administration was protective
2.7 NGF and Human Corneal against RGC degeneration (Coassin et al. 2008;
Neurotrophic Ulcer Colafrancesco et al. 2011; Lambiase et al. 2009a;
Lambiase et al. 2009b). To further explore the
Corneal neurotrophic ulcer is known to be role of NGF on retinal degeneration, we used a
associated with impairment of sensory innerva- rodent model of intraocular hypertension. In this
tion that can lead to loss of vision. At that time, study, we again investigated the effect of NGF
we started to address the potential usefulness of administration on RGCs. We found that both
NGF in ameliorating the condition. Unfortu- animal models displayed a progressive degenera-
nately, no effective treatment was available. As tion of RGCs and altered NGF and VEGF levels
other available treatments were ineffective, based in the retina and optic nerve. We then proved that
on the results obtained with animal models NGF eye drop administration exerted a protective
indicating that no undesired side effects occurred effect on the two models of retinal degeneration.
after the topic administration of NGF to the eye, In brief, our findings indicated that NGF can play
after approval of an ethical committee and a protective role against RGC degeneration
informed consent of the patient, we topically occurring in GL not only in rodents but also in
administered NGF to the eye of a patient affected humans, suggesting that ocular topical NGF
by the disorder. The treatment continued for administration could be a suitable pharmacologic
2 weeks, the ulcers healed, and the patients were agent to counteract RGC degeneration in GL.
16 L. Aloe

2.9 NGF and Macular Degeneration a powerful pharmacological tool for human cuta-
neous ulcers, including in cutaneous ulcer healing
Age-related macular degeneration (ARMD) is a in “non-responder” transplanted skin (Aloe
severe disease affecting visual function in the 2004b; Aloe and Chaldakov 2013; Chaldakov
elderly. Currently available surgical and medical 2011; Chaldakov et al. 2001; Generini et al.
options do not guarantee a protective outcome of 2004; Lambiase et al. 2009b; Landi et al. 2003;
RGC degeneration, and visual acuity is progres- Tuveri et al. 2000).
sively worsening despite surgical and medical
treatments. RLM suffered from a maculopathy
for over 5 years, and the laser therapy and other 2.11 Concluding Remarks
available therapies had no effect. Therefore, in
2002, RLM asked me to purify the NGF and to During the last three decades, despite some doubt
the ophthalmologist, A. Lambiase, to monitor the by RLM regarding the effective development of
action of the factor on her macular degeneration. potential clinical applications of NGF, I provided
After signing the informed consent, she was convincing demonstration, first in laboratory ani-
treated with NGF eye drops for six consecutive mal and later in human visual cells and cutaneous
months. Daily comments from the patient and ulcers, of the beneficial therapeutic effects of
weekly clinical examination from the ophthal- NGF in clinical settings. Indeed, the Italian phar-
mologist revealed the absence of NGF-related maceutical company Dompè has recently
undesired effects. Moreover, a clear improvement registered the recombinant NGF as Cenegermin,
of visual acuity and functional parameters were and the molecule has now been approved for
consistently monitored 3 months after the begin- medical application in Italy, Switzerland,
ning of the treatment (Tuveri et al. 2000). In brief, Germany, and Canada.
RLM discovered and used for her ocular disorder
the NGF treatment for nearly 6 years. Results Last Time I Meet RLM was less than 1 week
revealed that the ARMD was significantly before her death in her house. Despite she was
delayed with no undesired side effects. These rather tired, before leaving she asked me: “Luigi,
findings were published in 2009 (Lambiase et al. please remain a while more just to tell me some-
2009a). thing new about NGF.” Overall, for me, the sci-
entific and human experience working for nearly
45 years close to a Nobel winner, described as the
2.10 NGF and Cutaneous Wounds queen of modern neuroscience, brought a signifi-
cant contribution to both the basic and the clinical
Cutaneous wounds are known to elicit a series of saga of NGF.
topical cellular responses that include clotting,
inflammatory infiltration, epithelialization, and
the formation of granulation tissue, including References
new blood vessel, followed by tissue remodeling
and wound contraction. Based on the above and Aloe L (2004a) Rita Levi-Montalcini: the discovery of
other previous studies, we decided to investigate nerve growth factor and modern neurobiology. Trends
the effect of the topical application of purified Cell Biol USA 14(7):395–399
Aloe L (2004b) Nerve growth factor, human skin ulcers
NGF in human cutaneous ulcers induced by dia- and vascularization. Our experience. Prog Brain Res
betes, vasculitis, and rheumatoid arthritis. The 146:515–522. USA
results showed the efficacy of NGF was due to Aloe L, Chaldakov GN (2013) Homage to Rita Levi-
its promoting activity on keratinocyte prolifera- Montalcini, the queen of modern neuroscience. Cell
Biol Int USA 37(8):761–765
tion and vascular neoangiogenesis, suggesting Aloe L, Levi-Montalcini R (1972a) In vitro analysis of the
that topical application of NGF might represent frontal and ingluvial ganglia from nymphal specimens
2 The First and Last Time I Met Rita Levi-Montalcini: From the Insect. . . 17

of the cockroach Periplaneta Americana. Brain Res Giardino L, Giuliani A, Battaglia A, Carfagna N, Aloe L,
USA 44(1):147–163 Calza L (2002) Neuroprotection and aging of the cho-
Aloe L, Levi-Montalcini R (1972b) Interrelation and linergic system: a role for the ergoline derivative
dynamic activity of visceral muscle and nerve cells nicergoline (Sermion). Neuroscience USA 109
from insect embryos in long term cultures. J Neurobiol (3):487–497
USA 3(1):3–23 Hefti F, Will B (1987) Nerve growth factor is a
Aloe L, Levi-Montalcini R (1977) Mast cells increase in neurotrophic factor for forebrain cholinergic neurons;
tissues of neonatal rats injected with NGF. Brain Res implications for Alzheimer’s disease. J Neural Transm
USA 133(2):358–366 Suppl USA 24:309–315
Aloe L, Alleva E, Böhm A, Levi-Montalcini R (1986) Lambiase A, Aloe L (1996) Nerve growth factor delays
Aggressive behavior induces release of nerve growth retinal degeneration in C3H mice. Graefes arch Clin
factor from mouse salivary gland into the bloodstream. Exp Ophthalmol USA 234(1):96–100
Proc Natl Acad Sci USA 83(16):6184–6187 Lambiase A, Rama P, Bonini S, Caprioglio G, Aloe L
Calza L, Giardino L, Giuliani A, Aloe L, Levi-Montalcini (1998) Topical treatment with nerve growth factor for
R (2001) Nerve growth factor control of neuronal corneal neurotrophic ulcers. N Engl J Med. USA 338
expression of angiogenetic and vasoactive factors. (17):1174–1180
Proc Natl Acad Sci USA 98(7):4160–4165 Lambiase A, Aloe L, Centofanti M, Parisi V, Báo SN,
Calza L, Giuliani A, Fernandez M, Pirondi S, D’Intino G, Mantelli F, Colafrancesco V, Manni GL, Bucci MG,
Aloe L, Giardino L (2003) Neural stem cells and cho- Bonini S, Levi-Montalcini R (2009a) Experimental
linergic neurons: regulation by immunolesion and and clinical evidence of neuroprotection by nerve
treatment with mitogens, retinoic acid, and nerve growth factor eye drops: Implications for glaucoma.
growth factor. Proc Natl Acad Sci USA 100 Proc Natl Acad Sci USA 106(32):13469–13474
(12):7325–7330 Lambiase A, Coassin M, And AL (2009b) Eye drops
Chaldakov G (2011) The metabotrophic NGF and BDNF: improve visual acuity and electron functional activity
an emerging concept. Arch Ital Biol Italy 149 in age-related macular degeneration. Ann Ist super
(2):257–263 Sanita’ Italy 45:439–442
Chaldakov GN, Stankulov IS, Fiore M, Ghenev PI, Aloe L Landi F, Aloe L, Russo A, Cesari M, Onder G, Bonini S,
(2001) Nerve growth factor levels and mast cell distri- Carbonin PU, Bernabei R (2003) Topical treatment of
bution in human coronary atherosclerosis. Atheroscle- pressure ulcers with nerve growth factor: a randomized
rosis USA 159:57–66 clinical trial. Ann Intern Med Italy 139(8):635–641
Coassin M, Lambiase A, Sposato V, Micera A, Bonini S, Levi-Montalcini R (1952) Effects of mouse tumor trans-
Aloe L (2008) Retinal p75 and bax overexpression is plantation on the nervous system. Ann New York Acad
associated with retinal ganglion cells apoptosis in a rat Sci USA 55:330–343
model of glaucoma. Graefes Arch Clin Exp Levi-Montalcini R (1964) Growth control of nerve cells by
Ophthalmol USA 246(12):1743–1749 a protein and its antiserum. Science USA 143
Cohen S (1960) Purification of a nerve growth factor (3602):105–115
promoting protein from a mouse salivary gland and Levi-Montalcini R, Booker B (1960) Excessive growth of
its neurocytoxic antiserum. Proc Natl Acad Sci USA the sympathetic ganglia evoked by a protein isolated
46(3):302–311 from the mouse salivary glands. Proc Natl Acad Sci
Colafrancesco V, Parisi V, Sposato V, Rossi S, Russo MA, USA 46(3):373–384
Coassin M, Lambiase A, Aloe L (2011) Ocular appli- Levi-Montalcini R, Hamburger V (1951) Selective growth
cation of nerve growth factor protects degenerating stimulating effects of mouse sarcoma on the sensory
retinal ganglion cells in a rat model of glaucoma. J and sympathetic nervous system of the chick embryos.
Glaucoma 20(2):100–108 J Exp Zool USA 116(2):321–362
Fischer W, Wictorin K, Björklund A, Williams LR, Levi-Montalcini R, Aloe L, Mugnaini E, Oesch F,
Varon S, Gage FH (1987) Amelioration of cholinergic Thoenen H (1975) Nerve growth factor induces vol-
neuron atrophy and spatial memory impairment in ume increase and enhances tyrosine hydroxylase syn-
aged rats by nerve growth factor. Nature USA 329 thesis in chemically axotomized sympathetic ganglia
(6134):65–68 of newborn rats. Proc Natl Acad Sci USA 72
Generini S, Tuveri MA, Matucci Cerinic M, Mastinu F, (2):595–599
Manni L, Aloe L (2004) Topical application of nerve Seiler M, Schwab ME (1984) Specific retrograde transport
growth factor in human diabetic foot ulcers. A study of of nerve growth factor (NGF) from neocortex to
three cases. Exp Clin Endocrinol Diabetes USA 112 nucleus basalis in the rat. Brain Res USA 300(1):33–39
(9):542–544 Spillantini MG, Aloe L, Alleva E, De Simone R,
Goedert M, Levi-Montalcini R (1989) Nerve growth
18 L. Aloe

factor mRNA and protein increase in hypothalamus in Tuszynski MH, Sang H, Yoshida K, Gage FH (1991)
a mouse model of aggression. Proc Natl Acad Sci USA Recombinant human nerve growth factor infusions
86(21):8555–8559 prevent cholinergic neuronal degeneration in the adult
Torcia M, Bracci-Laudiero L, Lucibello M, Nencioni L, primate brain. Ann Neurol USA 30(5):625–636
Labardi D, Rubartelli A, Cozzolino F, Aloe L, Garaci E Tuveri M, Generini S, Matucci-Cerinic M, Aloe L (2000)
(1996) Nerve growth factor is an autocrine survival NGF, a useful tool in the treatment of chronic
factor for memory b lymphocytes. Cell USA 85 vasculitic ulcers in rheumatoid arthritis. Lancet USA
(3):345–356 356(9243):1739–1740
NGF Signaling in Endosomes
3
Naoya Yamashita

Abstract and biological functions of these specialized


During the development of the nervous sys- endosomes called “signaling endosomes.”
tem, neurons respond to diffusible cues
secreted by target cells. Because such target-
derived factors regulate development, matura-
tion, and maintenance of axons as well as 3.1 Retrograde NGF Signaling
somatodendritic compartments, signals Through Endosomes
initiated at distal axons must be retrogradely
transmitted toward cell bodies. Neurotrophins, During development, newborn neurons migrate to
including the nerve growth factor (NGF), pro- their proper locations and then extend their axons
vide one of the best-known examples of target- and dendrites into correct target regions to form
derived growth factors. The cell biological synapses with suitable partners. The neurons that
processes of endocytosis and retrograde traf- fail to innervate these targets are eliminated by a
ficking of their Trk receptors from growth genetically coded mechanism. Target innervation
cones to cell bodies are key mechanisms by of axons influences axon development and matu-
which target-derived neurotrophins influence ration as well as almost all aspects of the pro-
neurons. Evidence accumulated over the past cesses involved in neuronal development.
several decades has begun to uncover the Because neurons have long axons that extend
molecular mechanisms of formation, transport, even more than several centimeters, the signals
from target-derived factors must be retrogradely
propagated to the cell body and/or dendrites.
Therefore, long-distance retrograde signaling
from distal axons is one of the fundamental
features for regulating the formation, mainte-
nance, and function of neuronal circuits
(Harrington and Ginty 2013; Cosker and Segal
2014; Scott-Solomon and Kuruvilla 2018). Yet,
the molecular mechanisms of this retrograde sig-
nal propagation remain to be elucidated in full.
N. Yamashita (*) Neurotrophins are often produced in postsyn-
Department of Pharmacology, Juntendo University School aptic cells and act as target-derived factors to
of Medicine, Tokyo, Japan initiate signals at axonal growth cones (Glebova
e-mail: ny-yamashita@juntendo.ac.jp

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 19


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_3
20 N. Yamashita

and Ginty 2005). Early studies using radiolabeled 3.2 Endocytosis


125
I-NGF showed that in vivo injection into the of Neurotrophin-Receptor
iris and salivary glands—both of which are target Complexes in Growth Cones
organs for sympathetic neurons—resulted in the
accumulation of labeled NGF in the neuronal cell Like other receptor tyrosine kinases, the Trk
bodies that provide innervation to these structures receptors dimerize in response to a ligand (Jing
(Hendry et al. 1974; Stoeckel et al. 1975). Simi- et al. 1992). The dimeric receptors phosphorylate
larly, the retrograde accumulation of 125I-brain- one another in trans to activate a downstream
derived neurotrophic factor (BDNF), cascade, including phosphatidylinositol 3-kinase
125
I-neurotrophin 3 (NT-3), and 125I-NT-4 (also (PI3 kinase)-, Ras/MAPK-, and phospholipase
known as NT-5) in the cell bodies of oculomotor C-γ (PLCγ)-mediated pathways (Huang and
neurons were observed when those neurotrophins Reichardt 2003). In general, ligand-receptor
were applied to the extraocular eye muscles complexes are internalized by different endocytic
(Steljes et al. 1999), indicating that the target- pathways, and, after internalization, some
derived neurotrophins were retrogradely receptors are recycled back to the plasma mem-
transported along axons to mediate retrograde brane, while others are sorted together with their
signaling. Accumulating evidence further showed ligands for degradation in late endosomes and
that neurotrophins and their high-affinity Trk lysosomes. The internalization of ligand-receptor
receptor complexes were endocytosed into signaling complexes by endocytosis was, there-
growth cones and then retrogradely transported fore, considered for many years to be crucial for
to somatodendritic compartments for regulating the termination of signaling. However, it is now
neuronal growth, differentiation, survival, and clear that there is a high degree of compartmen-
synapse formation (Fig. 3.1). Because these talization of signaling needed to control and mod-
endosomes contain active neurotrophin signaling ulate the cell response. The NGF-bound TrkA
components—including several downstream receptors are internalized into signaling
molecules—they can transmit retrograde signal- endosomes that have been found to include
ing with high accuracy and persistency components of their downstream molecules
(Harrington and Ginty 2013; Cosker and Segal (Grimes et al. 1996, 1997; Howe et al. 2001;
2014; Yamashita and Kuruvilla 2016). Wu et al. 2001). Pharmacological inhibition of
The endosomes that provide retrograde signals Trk kinase activity and ectopic expression of
through this endosome-based transportation are kinase-dead Trk receptor mutant blocks ligand-
called “signaling endosomes,” a term that was receptor endocytosis and subsequent retrograde
proposed by W.C. Mobley and colleagues transport (Reynolds et al. 1998; Ye et al. 2003;
(Beattie et al. 1996). Initially, it was thought that Heerssen et al. 2004). Inhibition of TrkA kinase
these specialized endosomes communicated with activity in distal axons, but not in middle axons,
the nucleus to regulate the gene expression. How- of sympathetic neurons blocks retrograde trans-
ever, we now know that these endosomes also port of 125I-NGF (Ye et al. 2003; Zweifel et al.
influence neurons through transcription- 2005). Although the exact molecular mechanisms
independent mechanisms. Over several decades, are still unknown, it is therefore likely that kinase
the accumulating evidence—the majority of activity of TrkA mediates the formation of signal-
which is discovered in NGF-TrkA signaling— ing endosomes, but it is not required for retro-
began to uncover the molecular mechanisms of grade transport of these endosomes.
formation, transport, and biological functions of TrkA kinase activity is necessary for the asso-
signaling endosomes. ciation of clathrin with the plasma membrane in
3 NGF Signaling in Endosomes 21

Fig. 3.1 Nerve growth factor (NGF) secreted from target endosomes, called “signaling endosomes,” transmit retro-
organs activates TrkA receptors localized in distal axons. grade NGF signaling to regulate neuronal growth, differ-
In addition to activating the local signaling in growth entiation, survival, and synapse formation. Neurons that
cones, the active NGF-TrkA complexes are endocytosed fail to innervate their targets die because of the lack of
and then undergo retrograde axonal transport to survival signals. P phosphate. All figures in this chapter
somatodendritic compartments. TrkA-harboring are created with Biorender.com

PC12 cells (Beattie et al. 2000). In addition, required for the formation of signaling
clathrin-coated vesicles isolated from endosomes. At present, there is some debate as
NGF-treated PC12 cells contain NGF-TrkA to which endocytic route is the predominant mode
complexes and effectors of MAPK cascade, one for the formation of signaling endosomes. How-
of the downstream pathways of NGF-TrkA sig- ever, because NGF signaling endosomes have
naling (Howe et al. 2001). Therefore, clathrin- several biological functions and exist as several
dependent endocytosis may be at least one of distinct types of vesicles (see below), several
the major routes for the formation of signaling types of endocytosis may be involved, which is
endosomes. Several lines of evidence support the probably related to the sorting of endosomes for
idea that NGF-TrkA complexes are also regulating diverse biological functions.
endocytosed by clathrin-independent Both clathrin- and Pincher-mediated
macropinocytosis. Pincher, an NGF-upregulated endocytoses could be regulated by the endocytic
GTPase, is involved in this process. The GTPase, dynamin, because NGF-TrkA signaling
overexpression of Pincher enhances NGF/TrkA adjusts the activity of neuron-specific splicing
internalization both in PC12 cells and in sympa- isoform, dynamin 1. NGF stimulation induces
thetic neurons (Shao et al. 2002; Valdez et al. the tyrosine phosphorylation of TrkA at Tyr794
2005). In contrast, inhibiting the activity of (in rats, Tyr784 in human TrkA), which activates
Pincher suppresses internalization as well as the calcium-responsive phosphatase, calcineurin,
NGF retrograde signaling (Shao et al. 2002). by recruiting PLCγ to TrkA. Calcineurin
Thus, Pincher-mediated macropinocytosis is dephosphorylates PxIxIT motif-containing
22 N. Yamashita

dynamin 1 isoforms to promote TrkA endocyto- neurons, NGF and NT-3 bind to and activate
sis, which is required for NGF retrograde signal- TrkA, but only NGF supports survival because
ing through signaling endosomes (Bodmer et al. it induces the TrkA internalization and retrograde
2011). Once internalized, the NGF-TrkA transport (Kuruvilla et al. 2004). NT-3 binds to
endosomes must disassemble an F-actin mesh- TrkA with low affinity compared with NGF and
work to promote their entry into the axon for is therefore thought to dissociate from TrkA
retrograde transport. This process is mediated by within endosomes at low pH, resulting in
the activation of Rac1, which leads to actin depo- endosomes that are incapable of recruiting and
lymerization through the recruitment of cofilin to activating Rac1 and cofilin, a prerequisite step
the NGF-TrkA signaling endosomes (Fig. 3.2) for retrograde transport (Harrington et al. 2011).
(Harrington et al. 2011). A recent study showed
that the Rac1 is locally translated in growth cones
in response to NGF followed by prenylation of 3.3 Retrograde Axonal Transport
axonally translated Rac1. Because prenylation is of Signaling Endosomes
essential for retrograde transport of signaling
endosomes, prenylation coupled with the local As with other types of transported vesicles, the
translation of Rac1 in growth cones may mediate retrograde transport of neurotrophin-Trk
actin breakdown by neurotrophin signaling endosomes is driven by the dynein-microtubule
endosomes (Scott-Solomon and Kuruvilla transport system (Fig. 3.2). Microtubule depo-
2020). Although the exact contribution is lymerization inhibits the retrograde appearance
unknown, another TrkA effector, PI3 kinase, is of an active neurotrophin-Trk complex in the
also involved in the NGF retrograde signaling cell bodies following neurotrophin stimulation
through signaling endosomes. The inhibition of on distal axons (Watson et al. 1999). The disrup-
PI3 kinase prevents the accumulation of tion of the dynein-dynactin complex consistently
NGF-TrkA endosomes in the cell bodies of sym- attenuates the retrograde transport of
pathetic neurons, but it does not block internali- neurotrophin endosomes (Wu et al. 2007). More-
zation (Kuruvilla et al. 2000; Zweifel et al. 2005; over, the inhibition of dynein-dependent retro-
Yamashita and Kuruvilla 2016). Therefore, the grade transport also blocks survival responses to
PI3 kinase pathway probably regulates post- the target-derived neurotrophins (Heerssen et al.
endocytic sorting in distal axons. 2004). Thus, the dynein-dependent retrograde
The retrograde transport of signaling transport of signaling endosomes is indeed
endosomes may not be the only fate of required for the neurotrophin retrograde
neurotrophin-Trk receptor complexes that are signaling.
internalized into growth cones, because only a The type of vesicle used by the NGF signaling
minor population (approximately 2–2.5%) of endosomes during retrograde transport is one of
activated Trk receptors undergo retrograde trans- the most important issues to be resolved. It has
port (Ure and Campenot 1997; Tsui-Pierchala and long been argued that the neurotrophin signaling
Ginty 1999). The neurotrophin endosomes are endosomes are transported as a multivesicular
also recycled back to the growth cone membrane body (MVB), a Rab7-positive late endosome, or
or fused with lysosomes for degradation. a Rab5-positive early endosome (Bhattacharyya
Although the sorting mechanisms are currently et al. 2002; Delcroix et al. 2003; Deinhardt et al.
unknown, molecular components of individual 2006). Recent work by Ye et al. (2018) provided
endosomes may be a critical determinant. For new insight into this issue. By using FLAG-
example, Nedd4-2, an E3 ubiquitin ligase, binds tagged TrkA knock-in mice, they specifically
to TrkA and promotes its ubiquitination, a step labeled endogenous FLAG-TrkA localized on
that is involved in the sorting of neurotrophin distal axons. They found that although TrkA+
endosomes for degradation (Arevalo et al. endosomes in distal axons were mainly Rab5+,
2006). Moreover, in developing sympathetic the majority of distal axon-derived TrkA+
3 NGF Signaling in Endosomes 23

Fig. 3.2 The NGF-TrkA


complexes are endocytosed
into growth cones through a
PLCγ-calcineurin-dynamin
1 pathway. Once
internalized, the NGF-TrkA
endosomes disassemble an
F-actin meshwork through
Rac1 and cofilin to promote
entry into the axon for
retrograde transport. The
neurotrophin signaling
endosomes interact with the
dynein-dynactin complex,
thereby being transported
retrogradely to the
somatodendritic
compartments

endosomes observed in cell bodies were MVBs. independent mode of interaction with a dynein
They further showed that the TrkA+ MVBs were complex (Yano et al. 2001; Ha et al. 2008; Zhou
also Rab7-positive and these endosomes et al. 2012). In addition, other researchers showed
associated the dynein complex with the dynein the retrograde transport of Rab11 (a marker for
adaptor, RILP, utilizing Rab7. MVBs were recycling endosome)-positive signaling
shown to be attractive structures because they endosomes derived from the axonal TrkA of
were immune to lysosomal degradation and thus cultured sympathetic neurons from FLAG-TrkA
provided persistent signaling during retrograde knock-in mice (Barford et al. 2018). As described
transport. However, the axon-derived TrkA+ above, signaling endosomes are formed into
MVBs in cell bodies should be broken down to growth cones through several different types of
permit signaling from individual endosomes as endocytosis. Ultrastructural analyses of the sciatic
the topology of TrkA in MVB is resistant to nerve segment consistently showed that retro-
relaying signals to cytoplasmic effectors. Interest- gradely transported TrkA+ endosomes were
ingly, once reaching the cell bodies, the TrkA+ observed as several structurally distinct types of
MVBs evolved into single-membrane vesicles, vesicles ranging from 50 to 200 nanometers
which may therefore be signaling competent (nm) in diameter and included coated and
(Ye et al. 2018). uncoated vesicles as well as multivesicular
Whether or not an MVB is the sole organelle structures (Bhattacharyya et al. 2002). Therefore,
responsible for the retrograde transport of signaling endosomes may be heterogenetic
neurotrophin signaling endosomes is still organelles and thus may have many biological
unknown. Trk receptors also interact with dynein functions.
intermediate chains, suggesting a Rab7-
24 N. Yamashita

3.4 Biological Functions post-ganglionic sympathetic neurons without


of Retrograde Signaling affecting pre-ganglionic axon innervation,
Endosomes thereby suggesting the role of NGF in the matu-
ration of post-ganglionic sympathetic dendrites
During development, the neurons that compete (Sharma et al. 2010). Although how NGF signal-
for limited amounts of target-derived factors can ing endosomes influence dendritic arborization is
survive and form a neuronal circuit (Fig. 3.1) currently unknown, recent studies have begun to
(Glebova and Ginty 2005). The neurotrophin sig- uncover the role of NGF signaling endosomes in
naling endosomes surely contribute to this well- postsynaptic maturation. NGF stimulation on
accepted process. Upon arrival into the cell bod- sympathetic axons enhances the formation of
ies, signaling endosomes activate transcription postsynaptic receptor complexes, which are not
factors in the nucleus, including cyclic adenosine inhibited by de novo protein synthesis. Instead,
monophosphate response-element binding pro- distal axon-derived TrkA+ endosomes that have
tein (CREB), myocyte enhancer factor 2D been transported to dendrites induce the cluster-
(MEF2D), and c-fos utilizing MAPK and PI3K ing of postsynaptic components such as
signaling pathways, thereby inducing the gene membrane-associated guanylate kinases
expression required for neuronal survival (Riccio (MAGUKs), Shank, guanylate kinase-associated
et al. 1997; Watson et al. 1999; Pazyra-Murphy proteins (GKAPs), and nicotinic acetylcholine
et al. 2009). One gene responsible for a retrograde receptors (nAChRs) in dendrites (Sharma et al.
neuronal survival response is Bcl-w, an anti- 2010). At least about 35% of the axon-derived
apoptotic Bcl-2 family member, which is TrkA in dendrites is phosphorylated TrkA
upregulated by MEF2D through the MAPK path- (an active form of TrkA). Furthermore, local inhi-
way (Fig. 3.3a) (Pazyra-Murphy et al. 2009). In bition of TrkA kinase activity in dendrites
addition, NGF signaling endosomes also promote suppresses NGF-dependent postsynaptic cluster-
neuronal survival by enhancing their own signal- ing (Lehigh et al. 2017). These data indicate that
ing. In sympathetic neurons, NGF induces tran- the axon-derived TrkA signaling endosomes
scriptional upregulation of TrkA, which enhances found in dendrites are still signaling competent
NGF sensitivity and the duration of survival and regulate synaptic assembly and maintenance
signals in neurons responding to NGF through a transcription-independent mechanism
(Fig. 3.3a) (Deppmann et al. 2008). NGF further (Fig. 3.3c).
induces the expression of Coronin-1 which The retrograde transport of signaling
extends the duration of endosomal signaling. endosomes further controls the specificity of
Coronin-1 interacts with TrkA+ signaling anterograde transport. In addition to a direct
endosomes, which then mediate the local secretory pathway, TrkA is also targeted to
recycling of TrkA+ signaling endosomes and pre- growth cones through transcytosis; the naïve
vent them from fusing with lysosomes in cell TrkA receptors are initially localized on cell
bodies (Fig. 3.3a and b) (Suo et al. 2014). body surfaces followed by endocytosis and anter-
The target-derived signals from distal axons ograde transport to the growth cone surface
also play a crucial role in dendrite development (Ascano et al. 2009). This mode of TrkA axonal
and maturation (Fig. 3.3c). In sympathetic targeting is enhanced by NGF signaling
neurons, the target size influences the size and endosomes. The retrogradely transported axon-
complexity of the dendritic arbors (Voyvodic derived TrkA-harboring endosomes are
1989; Andrews et al. 1996). The administration exocytosed on the soma surface, which then
of NGF in vivo consistently enhances the den- induces the phosphorylation and endocytosis of
dritic arborization of sympathetic neurons (Snider unliganded naïve TrkA receptors through the
1988; Ruit et al. 1990). In addition, NGF is interaction of somatic and axonal TrkA receptors
required for the synaptic assembly of pre- and on the cell body membrane. Before anterograde
transport, phosphorylated naïve TrkA receptors
3 NGF Signaling in Endosomes 25

Fig. 3.3 Once reaching the cell bodies, the neurotrophin and pre-existing postsynaptic density components, includ-
signaling endosomes perform multiple biological ing MAGUK, GKAP, and Shank (c). TrkA+ endosomes
functions. The signaling endosomes activate several tran- further enhance their own trafficking. Axon-derived TrkA
scription programs in the nucleus (a). Coronin-1, one of interacts with naïve TrkA on the soma surface and subse-
the target genes for NGF signaling endosomes, interacts quently elicits phosphorylation and endocytosis of naïve
with TrkA+ endosomes to extend the duration of unliganded TrkA. These naïve TrkA receptors are then
endosomal signaling by promoting local recycling of dephosphorylated by PTP1B followed by anterograde
TrkA+ signaling endosomes or preventing TrkA+ transport to growth cones through the transcytotic pathway
endosomes to fuse with lysosomes in cell bodies (b). The (d). ER endoplasmic reticulum, CREB cyclic adenosine
active TrkA+ signaling endosomes are also transported to monophosphate response-element binding protein,
the dendrite where they regulate the clustering of nAChR MEF2D myocyte enhancer factor 2D

are dephosphorylated by an endoplasmic period of time and a tremendous amount of


reticulum-resident protein tyrosine phosphatase energy (Chowdary et al. 2012). Therefore, the
1B (PTP1B) (Fig. 3.3d). The inhibition of NGF-enhanced transcytosis machinery would be
PTP1B in cell bodies suppresses transcytosis an economical way to deliver TrkA to growth
and induces the degradation of Trk receptors cones because it can deliver an appropriate
that have been endocytosed from soma surfaces. amount of TrkA to the neurons that compete for
Thus, PTP1B acts as a gatekeeper that ensures the a limited amount of target-derived NGF. Retro-
targeting of inactive receptors (dephosphorylated grade transport of signaling endosomes is an
receptors) to growth cones to engage with the attractive way of retrograde signaling because it
ligand (Yamashita et al. 2017). Long-distance can specifically deliver the signaling components
axonal transport of endosomes takes an extended themselves where they are needed. Because
26 N. Yamashita

retrograde signaling endosomes regulate the spec- cell bodies by the dynein-dependent retrograde
ificity of anterograde transport, it is possible that transport. The axon-derived Sema3A signaling
the NGF-enhanced transcytosis further regulates complexes in cell bodies recruit the AMPA recep-
the trafficking of additional membrane proteins tor subunit GluA2 through interaction with
necessary for NGF functioning on axons. Further PlexinA4. These signaling complexes then
studies are needed to elucidate the molecular enhance the dendritic localization of GluA2,
identity of transcytosed vesicles. thereby regulating dendritic development
(Yamashita et al. 2014). Sema3A and NGF sig-
naling endosomes share some signaling
3.5 Generality of Retrograde components through the PlexinA4-TrkA interac-
Signaling Through Endosomes tion. The kinase activity and dynein-interacting
activity of TrkA are required for the retrograde
Although some alternative models have been pro- transport of Sema3A signaling endosomes,
posed, the model of signaling endosomes that whereas retrograde transport of PlexinA4 is not
transmit retrograde signaling is well accepted. involved in retrograde NGF signaling (Yamashita
Several lines of evidence suggest that this model et al. 2016; Yamane et al. 2017). These findings
may be a general mechanism that target-derived suggest that the molecular components of
growth factors utilize to transmit their retrograde Sema3A and NGF signaling endosomes do not
signals. Stimulation with ciliary neurotrophic fac- completely overlap, which indicates distinct sig-
tor (CNTF) or leukemia inhibitory factor (LIF) on naling pathways for Sema3A and NGF
distal axons of sympathetic neurons activates (Yamashita 2019). The target-derived Sema3A
STAT3 in the cell bodies, which is microtubule also induces cell death signaling in developing
dependent (O’Brien and Nathanson 2007). Simi- sympathetic neurons through retrograde trans-
larly, the retrograde transport of distal axon- port. In this case, PlexinA3 (but not PlexinA4)
derived bone morphogenetic protein (BMP) sig- is involved in the effect (Wehner et al. 2016),
naling complexes is required for the activation of which may imply that the functions of signaling
SMAD in the cell bodies (Smith et al. 2012; endosomes are regulated by organizing their
Walker et al. 2012). In addition, retrograde trans- molecular components.
port of glial cell line-derived neurotrophic factor
(GDNF) in motor neurons is considered to be
involved in the activation of Pea3 (Leitner et al. 3.6 Conclusions
1999; Haase et al. 2002). Until now, however,
physiological functions and detailed molecular From a classical view in which retrograde
mechanisms of these target-derived signals have neurotrophin signaling endosomes mediate
not been elucidated. target-derived survival signals in neurons, the
Recent studies have demonstrated the scenario has broadened to incorporate many
functions and molecular mechanisms of the retro- other biological functions that depend on endocy-
grade transport-dependent semaphorin 3A tosis of neurotrophin-Trk complex from the
(Sema3A) signal. Sema3A, a repulsive axon growth cone surface. Recent live cell imaging
guidance molecule, is also involved in the regula- experiments demonstrate that, although TrkA
tion of dendritic development and maturation in endosomes in axons move exclusively in a retro-
the cerebral cortex and hippocampus (Morita grade manner, axon-derived TrkA endosomes
et al. 2006; Yamashita et al. 2007; Nakamura exhibit various behaviors in somatodendritic
et al. 2009). In cultured hippocampal neurons, compartments. Although the majority of somatic
the site of Sema3A activity is restricted to the TrkA endosomes are localized to the perinuclear
axonal growth cone. Sema3A and its receptor region, others are localized peripherally and then
complex, including PlexinA4, are internalized undergo exo- and endocytosis and/or the move to
into the growth cone and are then transported to the dendrites (Lehigh et al. 2017; Ye et al. 2018).
3 NGF Signaling in Endosomes 27

In addition, some signaling endosomes undergo Arevalo JC, Waite J, Rajagopal R, Beyna M, Chen ZY,
fission and fusion events in somatodendritic Lee FS, Chao MV (2006) Cell survival through Trk
neurotrophin receptors is differentially regulated by
compartments (Barford et al. 2018). Thus, axon- ubiquitination. Neuron 50:549–559
derived neurotrophin signaling endosomes are Ascano M, Richmond A, Borden P, Kuruvilla R (2009)
well diversified in somatodendritic Axonal targeting of Trk receptors via transcytosis
compartments, and thus they may exert many regulates sensitivity to neurotrophin responses. J
Neurosci 29:11674–11685
biological functions. However, we still do not Barford K, Keeler A, McMahon L, McDaniel K, Yap CC,
know when, where, or how the destination and Deppmann CD, Winckler B (2018) Transcytosis of
function of the individual signaling endosome are TrkA leads to diversification of dendritic signaling
determined. In this regard, future studies are endosomes. Sci Rep 8:4715
BasuRay S, Mukherjee S, Romero E, Wilson MC,
needed to clarify how the functions of signaling Wandinger-Ness A (2010) Rab7 mutants associated
endosomes are related to the type of endocytosis, with Charcot-Marie-tooth disease exhibit enhanced
the mechanisms of dynein-dependent transport, NGF-stimulated signaling. PLoS One 5:e15351
and the molecular components of individual Beattie EC, Zhou J, Grimes ML, Bunnett NW, Howe CL,
Mobley WC (1996) A signaling endosome hypothesis
endosomes. to explain NGF actions: potential implications for
Neurons are unique cells with long axons, and neurodegeneration. Cold Spring Harb Symp Quant
the long-distance communication between axons Biol 61:389–406
and somatodendritic compartments is one of the Beattie EC, Howe CL, Wilde A, Brodsky FM, Mobley
WC (2000) NGF signals through TrkA to increase
important cell biological activities that neurons clathrin at the plasma membrane and enhance
perform throughout life without regeneration. clathrin-mediated membrane trafficking. J Neurosci
The study of signaling endosomes helps clarify 20:7325–7333
the mechanism by which signals initiated at the Bhattacharyya A, Watson FL, Pomeroy SL, Zhang YZ,
Stiles CD, Segal RA (2002) High-resolution imaging
distal axon propagate within the cell. This mech- demonstrates dynein-based vesicular transport of
anism is considered to be one of the foundations activated Trk receptors. J Neurobiol 51:302–312
for maintaining the neuronal survival and func- Bodmer D, Ascano M, Kuruvilla R (2011) Isoform-
tion. Because defects in signaling endosomes are specific dephosphorylation of dynamin1 by calcineurin
couples neurotrophin receptor endocytosis to axonal
known to be associated with various neurological growth. Neuron 70:1085–1099
disorders (BasuRay et al. 2010; Poon et al. 2011; Chowdary PD, Che DL, Cui B (2012) Neurotrophin sig-
Poon et al. 2013; Patel et al. 2015), understanding naling via long-distance axonal transport. Annu Rev
the formation, transport, and function of signaling Phys Chem 63:571–594
Cosker KE, Segal RA (2014) Neuronal signaling through
endosomes at the molecular level is necessary for endocytosis. Cold Spring Harb Perspect Biol 6(2):
understanding the larger issues surrounding the a020669
neuronal circuits in health and disease. Deinhardt K, Salinas S, Verastegui C, Watson R, Worth D,
Hanrahan S, Bucci C, Schiavo G (2006) Rab5 and
Rab7 control endocytic sorting along the axonal retro-
Acknowledgments The author’s work was supported by grade transport pathway. Neuron 52:293–305
the Naito Foundation, the Ichiro Kanehara Foundation, the Delcroix JD, Valletta JS, Wu C, Hunt SJ, Kowal AS,
Daiichi Sankyo Foundation of Life Science, MEXT/JSPS Mobley WC (2003) NGF signaling in sensory neurons:
KAKENHI (20K07742), and the Institute for Diseases of evidence that early endosomes carry NGF retrograde
Old Age, Juntendo University Graduate School of signals. Neuron 39:69–84
Medicine. Deppmann CD, Mihalas S, Sharma N, Lonze BE,
Niebur E, Ginty DD (2008) A model for neuronal
competition during development. Science
320:369–373
References Glebova NO, Ginty DD (2005) Growth and survival
signals controlling sympathetic nervous system devel-
Andrews TJ, Thrasivoulou C, Nesbit W, Cowen T (1996) opment. Annu Rev Neurosci 28:191–222
Target-specific differences in the dendritic morphology Grimes ML, Zhou J, Beattie EC, Yuen EC, Hall DE,
and neuropeptide content of neurons in the rat SCG Valletta JS, Topp KS, LaVail JH, Bunnett NW,
during development and aging. J Comp Neurol Mobley WC (1996) Endocytosis of activated TrkA:
368:33–44
28 N. Yamashita

evidence that nerve growth factor induces formation of Morita A, Yamashita N, Sasaki Y, Uchida Y, Nakajima O,
signaling endosomes. J Neurosci 16:7950–7964 Nakamura F, Yagi T, Taniguchi M, Usui H, Katoh-
Grimes ML, Beattie E, Mobley WC (1997) A signaling Semba R, Takei K, Goshima Y (2006) Regulation of
organelle containing the nerve growth factor-activated dendritic branching and spine maturation by
receptor tyrosine kinase, TrkA. Proc Natl Acad Sci semaphorin3A-Fyn signaling. J Neurosci
USA 94:9909–9914 26:2971–2980
Ha J, Lo KW, Myers KR, Carr TM, Humsi MK, Rasoul Nakamura F, Ugajin K, Yamashita N, Okada T, Uchida Y,
BA, Segal RA, Pfister KK (2008) A neuron-specific Taniguchi M, Ohshima T, Goshima Y (2009)
cytoplasmic dynein isoform preferentially transports Increased proximal bifurcation of CA1 pyramidal api-
TrkB signaling endosomes. J Cell Biol 181:1027–1039 cal dendrites in sema3A mutant mice. J Comp Neurol
Haase G, Dessaud E, Garces A, de Bovis B, Birling M, 516:360–375
Filippi P, Schmalbruch H, Arber S, deLapeyriere O O’Brien JJ, Nathanson NM (2007) Retrograde activation
(2002) GDNF acts through PEA3 to regulate cell of STAT3 by leukemia inhibitory factor in sympathetic
body positioning and muscle innervation of specific neurons. J Neurochem 103:288–302
motor neuron pools. Neuron 35:893–905 Patel A, Yamashita N, Ascano M, Bodmer D, Boehm E,
Harrington AW, Ginty DD (2013) Long-distance retro- Bodkin-Clarke C, Ryu YK, Kuruvilla R (2015)
grade neurotrophic factor signalling in neurons. Nat RCAN1 links impaired neurotrophin trafficking to
Rev Neurosci 14:177–187 aberrant development of the sympathetic nervous sys-
Harrington AW, St Hillaire C, Zweifel LS, Glebova NO, tem in down syndrome. Nat Commun 6:10119
Philippidou P, Halegoua S, Ginty DD (2011) Recruit- Pazyra-Murphy MF, Hans A, Courchesne SL, Karch C,
ment of actin modifiers to TrkA endosomes governs Cosker KE, Heerssen HM, Watson FL, Kim T,
retrograde NGF signaling and survival. Cell Greenberg ME, Segal RA (2009) A retrograde neuro-
146:421–434 nal survival response: target-derived neurotrophins
Heerssen HM, Pazyra MF, Segal RA (2004) Dynein regulate MEF2D and bcl-w. J Neurosci 29:6700–6709
motors transport activated Trks to promote survival of Poon WW, Blurton-Jones M, Tu CH, Feinberg LM,
target-dependent neurons. Nat Neurosci 7:596–604 Chabrier MA, Harris JW, Jeon NL, Cotman CW
Hendry IA, Stockel K, Thoenen H, Iversen LL (1974) The (2011) Beta-amyloid impairs axonal BDNF retrograde
retrograde axonal transport of nerve growth factor. trafficking. Neurobiol Aging 32:821–833
Brain Res 68:103–121 Poon WW, Carlos AJ, Aguilar BL, Berchtold NC,
Howe CL, Valletta JS, Rusnak AS, Mobley WC (2001) Kawano CK, Zograbyan V, Yaopruke T,
NGF signaling from clathrin-coated vesicles: evidence Shelanski M, Cotman CW (2013) Beta-amyloid
that signaling endosomes serve as a platform for the (Abeta) oligomers impair brain-derived neurotrophic
Ras-MAPK pathway. Neuron 32:801–814 factor retrograde trafficking by down-regulating
Huang EJ, Reichardt LF (2003) Trk receptors: roles in ubiquitin C-terminal hydrolase, UCH-L1. J Biol
neuronal signal transduction. Annu Rev Biochem Chem 288:16937–16948
72:609–642 Reynolds AJ, Bartlett SE, Hendry IA (1998) Signalling
Jing S, Tapley P, Barbacid M (1992) Nerve growth factor events regulating the retrograde axonal transport of
mediates signal transduction through trk homodimer 125I-beta nerve growth factor in vivo. Brain Res
receptors. Neuron 9:1067–1079 798:67–74
Kuruvilla R, Ye H, Ginty DD (2000) Spatially and func- Riccio A, Pierchala BA, Ciarallo CL, Ginty DD (1997) An
tionally distinct roles of the PI3-K effector pathway NGF-TrkA-mediated retrograde signal to transcription
during NGF signaling in sympathetic neurons. Neuron factor CREB in sympathetic neurons. Science
27:499–512 277:1097–1100
Kuruvilla R, Zweifel LS, Glebova NO, Lonze BE, Ruit KG, Osborne PA, Schmidt RE, Johnson EM Jr,
Valdez G, Ye H, Ginty DD (2004) A neurotrophin Snider WD (1990) Nerve growth factor regulates sym-
signaling cascade coordinates sympathetic neuron pathetic ganglion cell morphology and survival in the
development through differential control of TrkA traf- adult mouse. J Neurosci 10:2412–2419
ficking and retrograde signaling. Cell 118:243–255 Scott-Solomon E, Kuruvilla R (2018) Mechanisms of
Lehigh KM, West KM, Ginty DD (2017) Retrogradely neurotrophin trafficking via Trk receptors. Mol Cell
transported TrkA endosomes signal locally within Neurosci 91:25–33
dendrites to maintain sympathetic neuron synapses. Scott-Solomon E, Kuruvilla R (2020) Prenylation of
Cell Rep 19:86–100 axonally translated Rac1 controls NGF-dependent
Leitner ML, Molliver DC, Osborne PA, Vejsada R, axon growth. Dev Cell 53(691–705):e697
Golden JP, Lampe PA, Kato AC, Milbrandt J, Johnson Shao Y, Akmentin W, Toledo-Aral JJ, Rosenbaum J,
EM Jr (1999) Analysis of the retrograde transport of Valdez G, Cabot JB, Hilbush BS, Halegoua S (2002)
glial cell line-derived neurotrophic factor (GDNF), Pincher, a pinocytic chaperone for nerve growth factor/
neurturin, and persephin suggests that in vivo signaling TrkA signaling endosomes. J Cell Biol 157:679–691
for the GDNF family is GFRalpha coreceptor-specific. Sharma N, Deppmann CD, Harrington AW, St Hillaire C,
J Neurosci 19:9322–9331 Chen ZY, Lee FS, Ginty DD (2010) Long-distance
3 NGF Signaling in Endosomes 29

control of synapse assembly by target-derived NGF. functional dynein-microtubule network is required for
Neuron 67:422–434 NGF signaling through the Rap1/MAPK pathway.
Smith RB, Machamer JB, Kim NC, Hays TS, Marques G Traffic 8:1503–1520
(2012) Relay of retrograde synaptogenic signals Yamane M, Yamashita N, Hida T, Kamiya Y,
through axonal transport of BMP receptors. J Cell Sci Nakamura F, Kolattukudy P, Goshima Y (2017) A
125:3752–3764 functional coupling between CRMP1 and Nav1.7 for
Snider WD (1988) Nerve growth factor enhances dendritic retrograde propagation of semaphorin3A signaling. J
arborization of sympathetic ganglion cells in develop- Cell Sci 130:1393–1403
ing mammals. J Neurosci 8:2628–2634 Yamashita N (2019) Retrograde signaling via axonal
Steljes TP, Kinoshita Y, Wheeler EF, Oppenheim RW, transport through signaling endosomes. J Pharmacol
von Bartheld CS (1999) Neurotrophic factor regulation Sci 141:91–96
of developing avian oculomotor neurons: differential Yamashita N, Kuruvilla R (2016) Neurotrophin signaling
effects of BDNF and GDNF. J Neurobiol 41:295–315 endosomes: biogenesis, regulation, and functions. Curr
Stoeckel K, Schwab M, Thoenen H (1975) Specificity of Opin Neurobiol 39:139–145
retrograde transport of nerve growth factor (NGF) in Yamashita N, Morita A, Uchida Y, Nakamura F, Usui H,
sensory neurons: a biochemical and morphological Ohshima T, Taniguchi M, Honnorat J, Thomasset N,
study. Brain Res 89:1–14 Takei K, Takahashi T, Kolattukudy P, Goshima Y
Suo D, Park J, Harrington AW, Zweifel LS, Mihalas S, (2007) Regulation of spine development by
Deppmann CD (2014) Coronin-1 is a neurotrophin semaphorin3A through cyclin-dependent kinase
endosomal effector that is required for developmental 5 phosphorylation of collapsin response mediator pro-
competition for survival. Nat Neurosci 17:36–45 tein 1. J Neurosci 27:12546–12554
Tsui-Pierchala BA, Ginty DD (1999) Characterization of Yamashita N, Usui H, Nakamura F, Chen S, Sasaki Y,
an NGF-P-TrkA retrograde-signaling complex and Hida T, Suto F, Taniguchi M, Takei K, Goshima Y
age-dependent regulation of TrkA phosphorylation in (2014) Plexin-A4-dependent retrograde semaphorin
sympathetic neurons. J Neurosci 19:8207–8218 3A signalling regulates the dendritic localization of
Ure DR, Campenot RB (1997) Retrograde transport and GluA2-containing AMPA receptors. Nat Commun
steady-state distribution of 125I-nerve growth factor in 5:3424
rat sympathetic neurons in compartmented cultures. J Yamashita N, Yamane M, Suto F, Goshima Y (2016)
Neurosci 17:1282–1290 TrkA mediates retrograde semaphorin 3A signaling
Valdez G, Akmentin W, Philippidou P, Kuruvilla R, Ginty through plexin A4 to regulate dendritic branching. J
DD, Halegoua S (2005) Pincher-mediated Cell Sci 129:1802–1814
macroendocytosis underlies retrograde signaling by Yamashita N, Joshi R, Zhang S, Zhang ZY, Kuruvilla R
neurotrophin receptors. J Neurosci 25:5236–5247 (2017) Phospho-regulation of soma-to-axon
Voyvodic JT (1989) Peripheral target regulation of den- transcytosis of neurotrophin receptors. Dev Cell
dritic geometry in the rat superior cervical ganglion. J 42:626–639.e625
Neurosci 9:1997–2010 Yano H, Lee FS, Kong H, Chuang J, Arevalo J, Perez P,
Walker BA, Ji SJ, Jaffrey SR (2012) Intra-axonal transla- Sung C, Chao MV (2001) Association of Trk
tion of RhoA promotes axon growth inhibition by neurotrophin receptors with components of the cyto-
CSPG. J Neurosci 32:14442–14447 plasmic dynein motor. J Neurosci 21:RC125
Watson FL, Heerssen HM, Moheban DB, Lin MZ, Ye H, Kuruvilla R, Zweifel LS, Ginty DD (2003) Evi-
Sauvageot CM, Bhattacharyya A, Pomeroy SL, Segal dence in support of signaling endosome-based retro-
RA (1999) Rapid nuclear responses to target-derived grade survival of sympathetic neurons. Neuron
neurotrophins require retrograde transport of ligand- 39:57–68
receptor complex. J Neurosci 19:7889–7900 Ye M, Lehigh KM, Ginty DD (2018) Multivesicular bod-
Wehner AB, Abdesselem H, Dickendesher TL, Imai F, ies mediate long-range retrograde NGF-TrkA signal-
Yoshida Y, Giger RJ, Pierchala BA (2016) ing. elife 7:e33012
Semaphorin 3A is a retrograde cell death signal in Zhou B, Cai Q, Xie Y, Sheng ZH (2012) Snapin recruits
developing sympathetic neurons. Development dynein to BDNF-TrkB signaling endosomes for retro-
143:1560–1570 grade axonal transport and is essential for dendrite
Wu C, Lai CF, Mobley WC (2001) Nerve growth factor growth of cortical neurons. Cell Rep 2:42–51
activates persistent Rap1 signaling in endosomes. J Zweifel LS, Kuruvilla R, Ginty DD (2005) Functions and
Neurosci 21:5406–5416 mechanisms of retrograde neurotrophin signalling. Nat
Wu C, Ramirez A, Cui B, Ding J, Delcroix JD, Valletta JS, Rev Neurosci 6:615–625
Liu JJ, Yang Y, Chu S, Mobley WC (2007) A
The NGF Metabolic Pathway: New
Opportunities for Biomarker Research 4
and Drug Target Discovery
NGF Pathway Biomarkers and Drug Targets

Rowan Pentz and M. Florencia Iulita

Abstract disease-modifying nature of the cholinergic


Recent research has demonstrated that degen- deficit in AD and can incorporate other key
eration of the basal forebrain cholinergic sys- factors in basal forebrain cholinergic degener-
tem, far from being a mere downstream ation, including NGF receptor changes and
mediator of Alzheimer’s disease retrograde transport deficits in AD. This chap-
(AD) symptoms, may play a disease- ter will focus on the potential of NGF meta-
aggravating role in the continuum of AD bolic pathway biomarkers in AD as well as
pathology. The search for novel biomarkers therapeutic targets to correct NGF metabolic
of the cholinergic deficit in AD and novel deficits, aiding the development of novel
therapeutic targets for the sustenance of the pro-cholinergic therapeutics.
basal forebrain cholinergic system has there-
fore taken on more urgency. A novel model
that explains the preferential vulnerability of
basal forebrain cholinergic neurons in AD as 4.1 Introduction
the result of pathological alterations to nerve
growth factor (NGF) metabolism offers an It could be said that 2021 marks the beginning of
integrated investigative platform for the devel- a new era for Alzheimer’s disease (AD) patients:
opment of such biomarkers and therapeutics. accurate blood-based biomarkers based on
By positing a reciprocal trophic interaction phosphorylated tau isoforms (pTau-181 and
between the basal forebrain and its target pTau-217) are at the forefront in diagnostics
tissues, this model can also explain the (Janelidze et al. 2020; Karikari et al. 2020;
Palmqvist et al. 2020; Thijssen et al. 2020), neu-
R. Pentz roimaging tracers are being refined, and the anti-
Department of Neurology and Neurosurgery, McGill amyloid monoclonal antibody aducanumab is
University, Montreal, Canada
waiting for the FDA's decision on its application
e-mail: rowan.pentz@mail.mcgill.ca
by June 2021, which if approved, would be the
M. F. Iulita (*)
first disease-modifying treatment for AD. How-
Sant Pau Memory Unit, Department of Neurology,
Hospital de la Santa Creu i Sant Pau, Biomedical Research ever, despite these promising developments, the
Institute Sant Pau, Universitat Autònoma de Barcelona, reality remains that the majority of AD patients
Barcelona, Spain still get diagnosed when they develop cognitive
Center of Biomedical Investigation Network for impairment and still receive anticholinesterase
Neurodegenerative Diseases (CIBERNED), Madrid, Spain medication, dying approximately after
e-mail: miulita@santpau.cat

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 31


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_4
32 R. Pentz and M. F. Iulita

8–10 years, just like they would have in the (Debeir et al. 1999b; Allard et al. 2012b; Allard
previous century. et al. 2018b).
It is now widely recognized that Alzheimer’s The relevance of the BFCN system in AD is
dementia and even mild cognitive impairment best evidenced by the fact that four of the five
(MCI) represent relatively late stages in the AD licenced drugs to treat AD are acetylcholinester-
pathological process and ones that may entail an ase inhibitors (AChEIs), which prevent the break-
irreversible loss of the microstructural integrity of down of acetylcholine and thereby enhance
the brain. Furthermore, it is increasingly cholinergic neurotransmission. AChEIs are
recognized that AD is a heterogenous disorder widely applied drugs for AD and routinely pro-
for which no single biomarker or therapeutic vide a 6-month–2-year reprieve from cognitive
may be effective. Therefore, the effective man- decline—though after these drugs cease to be
agement of AD will likely require a panel of effective, patients return to their original patho-
easily accessible and cost-effective biomarkers logical trajectory (Giacobini and Becker 2007;
that can be widely implemented, as well as Pepeu and Giovannini 2010; Birks and Harvey
disease-modifying treatments that prevent, 2018). Importantly, the efficacy of AChEIs
reduce, or delay cognitive decline better than depends on some partial degree of cholinergic
current symptomatic medications (McDade and integrity (Richter et al. 2018) and correlates to
Bateman 2017). inhibition of AChE (Giacobini et al. 2002),
In this chapter, we will summarize the evi- demonstrating that the cholinergic system is a
dence of a novel and promising source of poten- validated therapeutic target in AD.
tial biomarker candidates and drug targets As AChEIs were considered to be “merely”
involving the nerve growth factor (NGF) meta- symptomatic drugs, interest in cholinergic
bolic pathway. As discussed at length in Chap. 8 therapies waned. However, in the effort to priori-
by A.C. Cuello, the discovery of the NGF meta- tize the development of disease-modifying
bolic pathway has unified paradoxical findings to treatments targeting core aspects of AD neuropa-
account for the cholinergic deficit in AD (Cuello thology, perhaps an opportunity to build on
et al. 2007; Iulita and Cuello 2014; Cuello et al. validated targets was missed. Indeed,
2019), serving as a foundation and framework for symptomatic treatments for diseases of old age
the discovery of novel therapeutic targets and can substantially recapitulate the benefits of
biomarker candidates. disease-modifying therapies. It has been
estimated that a drug (e.g. a novel AChEI) that
could provide 3 rather than the existing 2 years of
4.2 Targeting the Cholinergic symptomatic relief would produce an ~10%
System in AD: The Value reduction in the global prevalence of AD and a
of Symptomatic Treatments cholinergic therapy that extended that symptom-
atic benefit to 5 years would produce an ~40%
Cholinergic neurons that innervate the cerebral reduction in prevalence (Zissimopoulos et al.
cortex and hippocampus are essential for many 2015)—this is a viable notion, as L-DOPA
aspects of cognition and have their cell bodies in provides precisely such a 5-year reprieve for
the basal forebrain. Even though during develop- patients with typical Parkinson’s disease
ment all neurons depend on growth factors and presentations. Indeed, projections in the context
neurotrophins for survival, basal forebrain cholin- of the Canadian healthcare system have indicated
ergic neurons (BFCNs) are the only ones that that a drug offering 5 years of symptomatic relief
retain this close dependency in the adult brain would cause cases of AD and associated
(Hefti 1986; Hefti et al. 1986; Cuello 1996). healthcare costs to increase only by 10% rather
Specifically, these neurons depend on the contin- than by 98% over the next 50 years (Manuel et al.
uous supply of NGF for the maintenance of their 2016). Given that there are legitimate concerns
functional phenotype and synaptic integrity about the ability of modern healthcare systems to
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 33

meet the looming challenge of the AD epidemic decades as preclinical AD pathology advances.
(El-Hayek et al. 2019), the value of better Their atrophy is evident well prior to the onset
treatments for AD becomes readily apparent. of cognitive impairments (Grothe et al. 2014),
Although AChEIs are considered symptomatic and their degeneration predicts the atrophy of
drugs, there is evidence to suggest that they may the entorhinal cortex and cerebral cortex
also have disease-modifying properties. From a according to the topography of cholinergic inner-
mechanistic standpoint, earlier reports had vation (Schmitz et al. 2016, 2018).
indicated that physiological cholinergic signal- New insight into the causes of cholinergic
ling (particularly through M1 and M3 muscarinic degeneration in AD has come from the discovery
receptors) reduces amyloidosis (Nitsch et al. of the NGF metabolic pathway, which is
1992), lymphocyte proliferation, cytokine pro- described extensively in Chap. 8. Briefly, the
duction (Nizri et al. 2006), vasculopathy NGF precursor proNGF is secreted from neurons
(Claassen and Jansen 2006), and glutamate in response to cholinergic stimulation, is
excitotoxicity (Shen et al. 2010), leading to processed extracellularly to produce the mature
neuroprotection (Fisher 2008). Thus, the loss of peptide, and is subsequently degraded; these steps
this neuroprotective signalling, as a result of are accomplished by a set of co-released
BFCN degeneration and denervation of choliner- zymogens and convertases (Bruno and Cuello
gic input to target tissues (Bartus et al. 1982), 2006). Activated plasmin cleaves proNGF to
could help produce the neuropathology of AD mature NGF (mNGF) in the synaptic space. This
(Hampel et al. 2018). plasmin is derived from cleavage of its inactive
Recent evidence has converged to support this zymogen, plasminogen, by tissue plasminogen
model (Risacher et al. 2016; Hampel et al. 2018). activator (tPA), which is regulated by
Indeed, magnetic resonance imaging (MRI) stud- neuroserpin. The mNGF that is not taken up by
ies have shown that AChEIs lead to reductions in the presynaptic BFCN is degraded by matrix
the rate of atrophy of the hippocampus, cerebral metalloproteinase-9 (MMP-9) or MMP-3.
cortex, and BFCN in patients with suspected pro- MMP-9 and MMP-3 are derived from cleavage
dromal AD (Dubois et al. 2015; Cavedo et al. of their preprotein precursors, in a manner
2016; Cavedo et al. 2017). The validity of these regulated by tissue inhibitor of
findings is further strengthened by preclinical metalloproteinase-1 (TIMP1) (Bruno and Cuello
studies with novel pro-cholinergic drugs showing 2006).
lasting, disease-modifying effects of cholinergic The relevance of the NGF metabolic pathway
therapy in AD rodent models (Fig. 4.1) (Caccamo in modulating the availability of mNGF was
et al. 2006; Hall et al. 2018). Given that M1 demonstrated by experiments showing that the
receptors are relatively spared in AD (Svensson intracortical infusion of neuroserpin increased
et al. 1992; Overk et al. 2010), finding novel ways proNGF and that the inhibition of MMP-9
of activating these receptors (with minimal increased mNGF in naïve rats (Bruno and Cuello
peripheral side effects) remains an area of active 2006). These findings suggested that the NGF
research. pathway could indeed modulate cholinergic
tone, as the steady-state number of cortical cho-
linergic synapses is determined by NGF availabil-
4.3 NGF Dysmetabolism ity (Debeir et al. 1998, 1999a). The link between
and the Degeneration the NGF pathway and the status of the cholinergic
of the Cholinergic System projection system was later demonstrated by
in AD: A Self-Propagating experiments in which NGF maturation was
Feedback Loop? disrupted with long-term intracortical infusions
of α2-antiplasmin in naïve rats, leading to
Like the core AD pathological hallmarks, damage reduced density and integrity of cortical choliner-
to BFCNs accumulates silently over years or gic synapses (Allard et al. 2012a) as well as
34

Fig. 4.1 A novel selective allosteric M1 muscarinic agonist (AF710B) attenuates performance on the Novel Object Recognition Test and Morris Water Maze Test, as
cognitive deficits and AD pathology in McGill-R-Thy1-APP transgenic rats. At well as reduced amyloid plaque density, normalized synaptophysin levels, and reduced
13 months of age (post-plaque stage), rats homozygous for mutant human APP bearing microgliosis. Reproduced with permission from John Wiley and Sons, Hall et al.,
the Swedish and Indiana mutations were treated with oral AF710B or vehicle for Alzheimer’s & Dementia 2018
5 months. After a 1-month washout, AF710B-treated rats demonstrated better
R. Pentz and M. F. Iulita
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 35

smaller soma size of BFCNs selectively (Allard (Mufson et al. 2019), it is worth noting that
et al. 2018a). These reductions in NGF metabo- TrkA mRNA is induced by mNGF signalling
lism/cholinergic tone also led to impaired (Holtzman et al. 1992; Gibbs and Pfaff 1994;
learning and memory on a variety of behavioural Venero et al. 1994; Figueiredo et al. 1995) and
tasks (Allard et al. 2012a). TrkA changes could be downstream of changes to
The inclusion of NGF metabolism into a NGF availability, possibility exacerbating these
model of cholinergic degeneration in AD can alterations. It is evident how this cycle could
further explain how such changes may become a become self-propagating in light of the
self-propagating feedback loop driving AD accumulating amyloid pathology, evolving
pathology (Fig. 4.2). Amyloidosis has been inflammation, and NGF dysmetabolism charac-
demonstrated to lead to NGF dysmetabolism, as teristic of AD, driving cholinergic and cognitive
the infusion of amyloid oligomers alone is suffi- deficits (Fig. 4.2).
cient to trigger AD-like NGF dysmetabolism in
naïve rats (Bruno et al. 2009a), as transgenic rats
expressing mutant human APP exhibit NGF
4.4 The NGF Pathway as a Platform
dysmetabolism (Iulita et al. 2017), and as brain
for the Search of New
amyloidosis correlates to various measures of
Biomarkers
NGF dysmetabolism in human AD (Bruno and
Cuello 2006; Bruno et al. 2009a; Pentz et al.
Biomarkers for the incipient stage of AD have
2020). Mediation of this effect by inflammation
advanced significantly over the past decades, and
is suggested by studies demonstrating that Aβ
it is now possible to identify people in early
oligomer-induced NGF dysmetabolism is
stages of the disease with relatively high cer-
restored to a trophic state by the application of
tainty, through imaging and CSF measures of
an anti-inflammatory (Bruno et al. 2009a), the
core AD pathological markers and, more recently,
well-established associations between Aβ pathol-
via ultrasensitive plasma assays (Blennow and
ogy and inflammation (Welikovitch et al. 2020),
Zetterberg 2018; Jack 2020). Despite these posi-
and the key role that NGF pathway proteins
tive advances, there is also an increasing recogni-
(including MMPs and the plasmin-activating sys-
tion that AD is a heterogenous condition
tem) play in a variety of inflammatory pathways
comprising several distinct clinical and patholog-
(Parks et al. 2004; Mehra et al. 2016).
ical subtypes (Murray et al. 2011), and meaning-
The resulting impairment of proNGF matura-
ful results may therefore not come from a single
tion and excessive mNGF degradation will then
biomarker.
lead to the atrophy of BFCNs and the loss of
In this regard, the NGF metabolic pathway
cortical cholinergic tone, as demonstrated causa-
offers an integrative exploratory platform to iden-
tively in experimental models (Allard et al.
tify new biomarkers of AD-related cholinergic
2012a, 2018a) and supported correlatively in
degeneration. Indeed, as cholinergic function/
human AD by a demonstration of the association
pathology is tightly tied to cognition (Drachman
between VAChT depletion and proNGF accumu-
and Leavitt 1974; Hasselmo and Sarter 2011) and
lation (Pentz et al. 2020). These deficits will then
differs across AD subtypes (Machado et al.
result in increased amyloidosis and/or inflamma-
2020), a biomarker that tracks this process could
tion by the mechanisms described above, and
have utility for sub-diagnosis and prognosis. As
excitotoxicity could play a role as well, as it is
the efficacy of AChEIs depends on partially intact
reduced by cholinergic signalling (Shen et al.
cholinergic tone (Richter et al. 2018), biomarkers
2010) and associated with acute increases in
associated with NGF metabolism could also be
proNGF (Le 2011).
used to inform about the efficacy of cholinergic
While a downregulation in TrkA levels has
therapies.
been implicated in AD cholinergic pathology
36 R. Pentz and M. F. Iulita

Fig. 4.2 Hypothetical basocortical feedback loop driving downregulation of NGF receptor expression and dimin-
Aβ pathology and NGF dysmetabolism in AD. Aβ pathol- ished cholinergic trophic support resulting in reduced cho-
ogy is sufficient to unleash an early pro-inflammatory linergic tone. Because of reduced cholinergic
process and to impair NGF metabolism by facilitating neurotransmission, the generation of toxic Aβ peptides
proNGF accumulation and reduced mNGF availability. and inflammation will be favoured, contributing to the
A compromised NGF metabolism would lead to self-propagation of this loop

alzheimer-down-unit/) (Fortea et al. 2018b,


4.5 Tracking NGF Metabolism
2020).
in AD Biofluids
The first study on the characterization of NGF
metabolic pathway proteins in plasma samples
Initial studies investigating NGF pathway
from a population-based cohort of individuals
markers in body fluids focused on individuals
with DS from Catania, Italy, found significant
with Down syndrome (DS). Because of the tri-
differences in DS with respect to euploid controls:
somy of chromosome 21, which harbours the
increased proNGF and increased MMP-9,
APP gene, individuals with DS are considered a
MMP-3, and MMP-1 in people with DS and
genetically determined high-risk population for
established dementia (DSAD), as well as in
AD. As they inevitably develop advanced AD
those who were asymptomatic (DS) (Iulita et al.
pathology by midlife (Head and Lott 2004;
2016). No changes were observed for tPA and
Head et al. 2012; Lott and Head 2019), studies
neuroserpin. A longitudinal assessment further
in DS represent a unique opportunity to investi-
showed that an increase in proNGF over a
gate biomarker trajectories particularly in the
1-year interval predicted prospective cognitive
early, silent stages of AD. This has been reflected
deterioration over 2 years (Iulita et al. 2016).
in the launch of several new projects assessing
A subsequent study, assessing a different
AD biomarkers, such as the Alzheimer’s
population-based cohort of people with DS from
Biomarkers Consortium-Down Syndrome
Barcelona, Spain, recapitulated these findings on
(ABC-DS, https://www.nia.nih.gov/research/
NGF markers in plasma and further extended
abc-ds) (Handen et al. 2020) and the Down
them to comparisons in CSF (Pentz et al.
Alzheimer Barcelona Neuroimaging Initiative
2021a). Highly similar profiles of NGF
(DABNI, https://santpaumemoryunit.com/
dysmetabolism were found in plasma, while
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 37

profiles in CSF bore yet more similarity to the with clinical dementia ratings (CDRs) of 0.5 com-
status of the NGF metabolic pathway in AD pared to people with CDR 0 (Counts et al. 2016),
(Pentz et al. 2020) and DSAD brains (Iulita and the difference was higher when combined
et al. 2014). Furthermore, CSF analysis permitted with Aβ42 levels, indicating the changes in
the differentiation of different profiles in DS and proNGF precede the onset of dementia and are
DSAD: DS was characterized by associated with preclinical pathology. CSF
increased proNGF (27 kDa and 50 kDa forms), proNGF correlated to several cognitive test scores
increased neuroserpin, increased MMP-3, and (Counts et al. 2016), recapitulating similar
increased MMP-1. DSAD in CSF recapitulated associations observed in brain tissue (Peng et al.
these changes and was associated with signifi- 2015; Pentz et al. 2020). However, it must be
cantly further increased 50 kDa proNGF, noted that this analysis was done in CSF taken
increased MMP-9, and normalized MMP-1. intracerebroventricularly, post-mortem, not spinal
ROC analysis showed that CSF MMP-9 and the CSF taken from living patients. In line with this
50 kDa form of proNGF differentiated DS from account, a previous study had identified increased
DSAD with good diagnostic accuracy NGF in AD CSF using methods that did not
(AUC ¼ 0.86–0.87). This study also revealed distinguish proNGF from mNGF (Massaro et al.
that NGF pathway proteins correlated to core 1994), and, given that that proNGF predominates
biomarkers of AD, with particularly strong over mNGF in CSF, this could be counted as
associations observed between MMP-3 and total confirmation.
tau as well as between proNGF (50 kDa) and the
Aβ42/40 ratio. Together, these results provide a
strong rationale for the idea that components of 4.5.2 tPA and Neuroserpin
the AD pathological process may be traceable by
signatures of NGF pathway proteins. Lower tPA levels have been reported in AD CSF,
Below we will summarize and discuss previ- where it correlates positively to cognitive scores
ous studies assessing NGF pathway proteins indi- (Hanzel et al. 2014a)—a relationship that was
vidually in biofluids as biomarkers of AD: also demonstrated in human frontal cortex from
cognitively healthy aged individuals (Pentz et al.
2020). However, no differences in tPA levels in
4.5.1 ProNGF CSF between MCI, AD, and controls were
reported in other studies (Martorana et al. 2012;
Other studies have examined NGF/proNGF in Marksteiner et al. 2014), in line with our findings
body fluids independent from its metabolic path- in a DS cohort (Pentz et al. 2021a). tPA may be
way. However, most proNGF and mNGF ELISA sensitive to storage conditions or to inter-assay
systems do not effectively distinguish between variance, or the results may be confounded by
the two (Malerba et al. 2016)—a critical draw- another association—for example, tPA has been
back in a condition where such markers are suggested as a biomarker of geriatric depression
regulated in opposite directions. Furthermore, (Shi et al. 2010), and prodromal (and clinically
the independent and important contribution of manifest) AD patients, as well as people with DS,
proNGF in the brain and in AD was not often manifest depression symptoms.
recognized until 2001 (Fahnestock et al. 2001), CSF neuroserpin has been reliably found to be
and as such, in biomarker studies prior to that upregulated in AD CSF (Nielsen et al. 2007;
date, it was not independently assessed (Hock Hanzel et al. 2014a), in accordance with the
et al. 2000). results in DSAD CSF from the Barcelona cohort.
In the sole other study to explicitly consider Interestingly, one study found that neuroserpin
CSF proNGF in the context of AD, CSF proNGF levels could identify people with AD from
levels were markedly higher in amnesic MCI and controls and from patients with other neurodegen-
AD patients. They were also elevated in persons erative conditions, suggesting that a neuroserpin
38 R. Pentz and M. F. Iulita

upregulation might be a specific feature of AD et al. 2009; Lim et al. 2011; Tuna et al. 2018;
(Nielsen et al. 2007). Iulita et al. 2019), while others have described
increases (Lorenzl et al. 2003; Cai et al. 2007;
Lorenzl et al. 2008; Whelan et al. 2019), as in our
4.5.3 Matrix Metalloproteinase-3 study in DS, or reductions (Horstmann et al.
and Metalloproteinase-9 2010). Similar discrepancies have been obtained
when MMP-9 was considered in CSF, with a
CSF MMP-3 has been observed to be upregulated plurality of studies identifying increases (Lorenzl
in sporadic AD compared to MCI and healthy et al. 2003; Cai et al. 2007; Lorenzl et al. 2008;
controls (Hanzel et al. 2014b), in accordance Stomrud et al. 2010; Whelan et al. 2019) but other
with the increase observed in DS/DSAD. CSF studies finding no change (Adair et al. 2004;
MMP-3 was also shown to correlate to total tau Hanzel et al. 2014a) or decreases (Mroczko
and p-Tau-181 levels, as well as cognitive decline et al. 2014) with respect to controls. Several
in the context of AD (Hanzel et al. 2014b). factors could underlie these discrepancies.
Plasma MMP-3 was likewise demonstrated to be MMP-9 is scarcely expressed in CSF (Horstmann
upregulated in AD and to associate with cognitive et al. 2010), and even temperatures of 80  C do
outcomes in a retrospective cohort analysis (Iulita not fully protect it from degradation, rendering it
et al. 2019). Higher MMP-3 in AD plasma and highly susceptible to differences in storage
correlations to cognitive status were also conditions (Rouy et al. 2005). The preparation
demonstrated by a second study (Lin et al. of plasma, which commonly contains protease
2006), while a third, using both CSF and plasma, inhibitors and chelators such as EDTA, may con-
showed that MMP-3 was increased in AD found zymographic studies (which are used to
plasma, and a trend was observable in AD CSF assess MMP activity), as MMPs are Zn+2-depen-
(Horstmann et al. 2010). dent enzymes. Other storage conditions (as well
MMP-3 is also higher in CSF from cognitively as inhibitors/peptidase levels) could also cause
healthy individuals classified as “at risk of AD” activity-based and antigen-based MMP assays to
by biomarker scores (Stomrud et al. 2010). This differ. Additionally, different comorbidities
study also confirmed the associations with between the study populations, some of which
p-Tau-181 and total tau (Stomrud et al. 2010) are very common in DS syndrome, should also
observed by Hanzel and colleagues (Hanzel be considered.
et al. 2014b) and in our work in DS (Pentz et al.
2021b). Diverging results were found in a study
that showed lower MMP-3 alongside reduced 4.5.4 Correspondence Between NGF
CSF Aβ42 in people classified as having Metabolism in Biofluids
neuroinflammation/neuroinfection while finding and the CNS
no association between CSF tau and MMP-3
(Mlekusch and Humpel 2009). Although the How do the described biomarker results accord
reasons behind these discrepant findings are not with the status of the NGF metabolic pathway in
clear, CSF was frozen 3 days after collection in brains with AD pathology? ProNGF protein,
this study, which might have affected MMP-3 MMP-9 activity, and neuroserpin protein have
stability. Overall, there is general agreement that been demonstrated to be elevated in brains from
plasma and CSF MMP-3 are increased in the AD people with DSAD, concurrent with reduced tPA
pathological process and that biofluid MMP-3 is mRNA (Iulita et al. 2014), as reviewed in (Iulita
associated with p-Tau-181 and total tau rather and Cuello 2016). These changes, apart from tPA,
than other core biomarkers of AD. accord with findings in CSF in the DS population;
Assessments of plasma MMP-9 in AD have however, plasma did not reflect the changes to
yielded divergent results. Several studies have MMP-9 or neuroserpin seen in the brain (Iulita
demonstrated no change in AD (Martín-Aragón et al. 2014; Pentz et al. 2021a). Interestingly,
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 39

Table 4.1 Summary of changes in NGF pathway markers found in biofluids and the brain
Plasma CSF Brain Comment
proNGF " " " Diagnostic accuracy to distinguish AD in DS is superior in
CSF vs. plasma; 50 kDa form appears more sensitive to diagnosis
than 27 kDa form
tPA # or no # or no # tPA may be susceptible to storage conditions or to inter-assay
change change variance. Results may be confounded by another association,
e.g. depression
Neuroserpin No " " Results consistent across studies. Increased in AD vs. controls
change and vs. other dementias
MMP-3 " " " Consistent results across studies; biofluid MMP-3 is associated with
p-Tau-181 and total tau; MMP-3 levels differ by sex
MMP-9 "# or no "# or no " MMP-9 may be sensitive to degradation and susceptible to storage
change change conditions. EDTA tubes for plasma collection may impact MMP
activity as they are Zn+2-dependent enzymes
mNGF Data not Data not # ELISA cannot differentiate mNGF and proNGF. No accounts of
available available mNGF measured by Western blot in biofluids. With special
extraction protocols, it is possible to measure it in brain homogenates
AD Alzheimer’s disease, CSF cerebrospinal fluid, DS Down syndrome, mNGF mature NGF, MMP matrix
metalloproteinase, proNGF precursor of nerve growth factor, tPA tissue plasminogen activator

increased levels of proNGF, neuroserpin, that each of those people would have a 75%
unchanged tPA, MMP-9, and MMP-3 also corre- chance of never manifesting the condition
spond to the status of the NGF metabolic pathway (Brookmeyer and Abdalla 2018). As such, there
in sporadic AD brains, though in one study tPA would be three unnecessary AD treatments for
was found to be downregulated (Bruno et al. every successful one—an extreme cost when
2009a). These findings demonstrate that NGF considering that these potentially expen-
dysmetabolism is highly similar in DS and spo- sive treatments might have to be applied for
radic AD brains and that biofluids—particularly decades in a large swath of the elderly population.
CSF—reflect this cerebral NGF dysmetabolism These models suggest that biomarkers of preclin-
well (Pentz et al. 2021a). Table 4.1 offers a com- ical AD need to be refined by inclusion of other
parative summary between biofluids and brains. markers beyond amyloid and tau that are also
surrogates of key neurodegenerative processes
in this complex disease. Given the strong associ-
ation between NGF metabolism, cholinergic tone,
4.6 Diagnostic Value of NGF
and cognition, we suggest that the NGF metabolic
Pathway Biomarkers
pathway could be a source of such biomarkers.
Could NGF pathway proteins assist in the
Why does the AD field continue to fight for better
identifications of individuals with incipient
or additional biomarkers? Although AD can now
dementia? As described above, the ROC curve
be diagnosed by biomarker expression at preclin-
values determined for CSF proNGF and CSF
ical stages (Dubois et al. 2014; McDade and
MMP-9 in the DS population were good (0.86
Bateman 2017; Jack et al. 2018), these measures
and 0.87), though they were outperformed by the
are imperfect as they fail to acknowledge the
diagnostic accuracy of Aβ42 or the Aβ42/40 ratio
heterogeneity inherent in AD (Murray et al.
(Fortea et al. 2018a). As such, NGF pathway
2011) and offer limited prognostic value at such
markers alone will not likely serve as a primary
early stages. Indeed, one recent model suggested
means of diagnosing AD. However, NGF meta-
that 30% of Americans over the age of 50 would
bolic pathway proteins could add value and fur-
be diagnosed with preclinical AD under interna-
ther specificity to traditional biomarkers (i.e. as
tional diagnostic criteria (IWG-2/NIA-AA) but
40 R. Pentz and M. F. Iulita

part of a panel), particularly to identify subtypes 4.7 NGF as a Therapeutic Target


of AD with prominent cholinergic lesions in AD
(Machado et al. 2020). Indeed, a recent study
has demonstrated that neuronal density was low- AChEIs, while valuable, only transiently boost
est and tau pathology was highest in the nucleus the output of BFCNs. Could NGF be an effective
basalis of Meynert (NbM)—part of the BFC sys- intervention to sustain BFCN output and cholin-
tem—in the hippocampal-sparing subtype of spo- ergic tone more substantially in AD? Initial
radic AD, that typical AD was intermediate with results from a single patient who received intra-
regard to both tau pathology and NbM cell count, ventricular NGF showed increased nicotinic
and that limbic-predominant AD showed the low- receptor expression and cerebral perfusion, with
est NbM tau pathology and the highest NbM positive indications on some—though not all—
neuronal density, as well as the lowest NbM cognitive tests and particularly good results in a
plaque counts (Al-Shaikh et al. 2020). A second test of verbal memory (Olson et al. 1992). Similar
study identified limbic-sparing AD as being positive results (verbal fluency excepted) had pre-
associated with the greatest longitudinal loss of viously been obtained with the direct infusion of
BFCN volume over several visits, while a “mini- NGF into the brains of crab-eating macaques
mal atrophy subtype” predictably displayed the (Tuszynski et al. 1990; Tuszynski et al. 1991).
least BFCN volume loss (Machado et al. 2020). However, further studies demonstrated that this
When these patients were treated with the approach was nonviable in humans, as it led to
encapsulated cell devices overexpressing NGF chronic pain and weight loss, presumably by the
(discussed in greater detail below), reduced atro- cross-sprouting of spinal nerve afferents and
phy of the precuneus and hippocampus was hypothalamic plasticity (Jönhagen et al. 1998;
observed only in the hippocampal-sparing and McKelvey et al. 2013). The authors of these stud-
typical AD subtypes (Machado et al. 2020). ies concluded they provided substantial proof of
While further work will be required, these initial concept for NGF therapy in AD but that direct
studies provide evidence that the cholinergic def- infusion would not be viable, indicating that other
icit may differ across subtypes of AD and methods of altering NGF-mediated trophic sup-
strongly suggest that NGF pathway-derived port would be required (Jönhagen 2000).
biomarkers could identify certain of these Gene therapy approaches can drive the
subtypes with different degree of cholinergic overexpression of mNGF in target tissues without
degeneration. Follow-up studies should incorpo- nociceptive or constitutional side effects
rate other novel biomarkers of cholinergic tone in (Tuszynski et al. 1990; Hao et al. 2000). In a
AD, including the proposed “cholinergic index”, successful phase I trial, this approach trended
a ratio of ChAT to AChE levels in CSF, which towards reducing decline on the Mini-Mental Sta-
demonstrated the ability to measure treatment tus Examination and Alzheimer’s Disease
response to AChEIs in a recent study (Karami Assessment Scale-Cognitive subcomponent and
et al. 2019). Similar measures of treatment increased cortical glucose metabolism in eight
response could provide a second potential signifi- individuals with AD (Tuszynski et al. 2005).
cant role for NGF pathway biomarkers in AD, Follow-up trials attempted to use advances in
and, as AChEIs depend on some degree of cho- adeno-associated virus technology to deliver
linergic tone to exert their effect (Richter et al. human mNGF directly to the NbM. This con-
2018), identifying patients likely to respond to struct, entitled CERE-110, enhanced cholinergic
AChEIs or other cholinergic therapies could pro- trophism in rodent models (Bishop et al. 2008)
vide a third. and AD patients (Rafii et al. 2014; Tuszynski
et al. 2015), with no signs of deleterious side
effects. Unfortunately, a larger clinical trial in
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 41

49 AD patients showed no effects on its competence but does not induce a pain response
endpoints, which included cognitive testing, (Capsoni et al. 2011). This mutant NGF has been
FDG-PET, and longitudinal hippocampal primarily delivered intranasally, a protocol that
volumetry (Rafii et al. 2018). However, recent has been shown to alleviate plaque load and cog-
results have suggested that this may have been nitive decline in APP/PSEN1 mutant mice
mediated by technical rather than conceptual (Capsoni et al. 2012). However, these effects do
limitations, as a post-mortem analysis of the not appear to be mediated by cholinergic
brains of 15 patients revealed that not one of trophism, and the associated researchers now pro-
their CERE-110 injections was correctly pose “painless” intranasal NGF as modulator of
localized to the basal forebrain (Castle et al. inflammation (Capsoni et al. 2017; Cattaneo and
2020). This, in addition to the fact that the study Capsoni 2019). Although more remains to be
was underpowered to detect efficacy and applied learned about this protein, mutant NGF is an
only to patients with advanced AD (Rafii et al. exciting possibility in the future of pro-trophic
2018), suggests that this approach, while inevita- therapeutics.
bly challenging and invasive, may still hold The above approaches, though varied in many
promise. ways, share not only their target but the fact that
While the virus-mediated modification of the increased NGF they elicit is exogenously
neurons to overexpress NGF has been abandoned, derived and localized widely over a given area,
a similar approach in which fibroblasts are col- most of which lies outside cholinergic synapses.
lected from a patient, modified to overexpress This entails several disadvantages. Firstly, NGF
mNGF, and implanted into the basal forebrain can have effects on off-target neurons and even
within an encapsulation device is being actively glia; in fact, many cells that are not usually
pursued (Eriksdotter-Jönhagen et al. 2012; NGF-competent express the p75NTR in AD
Eyjolfsdottir et al. 2016). There are positive (Mufson and Kordower 1992; McKelvey et al.
indications for the utility of this approach, as 2013; Cattaneo and Capsoni 2019), so it is not
small studies have demonstrated positive effects clear what the sum effect of exogenous NGF
on cognition, brain atrophy, and some (but not would be on the AD pathological process. Sec-
all) fluid AD biomarkers (Ferreira et al. 2015; ondly, these treatments would be applied in the
Karami et al. 2015). Intriguingly, this treatment context of significantly elevated NGF-degrading
also rectified CSF ChAT and AChE levels, which metalloproteinases (Bruno and Cuello 2006;
are proposed biomarkers of cholinergic integrity. Iulita et al. 2014; Pentz et al. 2020)—therefore it
Despite these promising findings, there are is not clear that the amount reaching cholinergic
reasons to be cautious. Many of the positive synapses (if any) would suffice to overcome this
changes were only observed in patients that excessive degradation. Thirdly, many of these
received a post hoc classification as “responders” approaches are highly invasive and technically
based on 2 point reduction on the Mini-Mental challenging, requiring injections or insertions
State Examination (MMSE) (Ferreira et al. 2015; into the brain parenchyma or ventricles.
Karami et al. 2015), and a recent report suggests Targeting other proteins that participate in the
that CSF factors may impair the long-term effi- metabolism of NGF has the potential to overcome
cacy of the implants (Eriksdotter et al. 2018). all three of these following concerns. In the first,
A third approach takes advantage of the dis- NGF would still be solely produced at cholinergic
covery that a condition called “type IV hereditary synapses; in the second, the excessive degrada-
sensory and autonomic neuropathy”, in which tion of mNGF could itself be a target for therapy;
patients have a significant nociceptive deficit, and in the third, if a target specific to cholinergic
results from a R100W point mutation in the synapses can be identified—as discussed
NGF gene (Capsoni et al. 2011). This mutant below—a drug could be applied systemically.
NGF could permit the resurrection of direct This type of therapy could have the potential to
infusion approaches, as it retains full trophic produce a long-lasting rescue of BFCNs and
42 R. Pentz and M. F. Iulita

cortical cholinergic tone. Such a drug could also potential of tPA therapy in correcting NGF
be used adjunctly to enhance the effect of dysmetabolism, several considerations should be
AChEIs. weighed. Some studies in cellular cultures have
suggested that tPA might induce or mediate AD
neuropathology, particularly amyloidosis and the
4.8 Targeting NGF Metabolism inflammatory activation of microglia (Siao and
Tsirka 2002; Medina et al. 2005), suggesting
A therapeutic that intervenes to restore trophic that tPA is a complex target. It must further be
support to BFCNs by normalizing NGF metabo- recalled that tPA therapy in cases of acute ische-
lism could do so by two mechanisms: by restor- mic stroke is limited to a small window of 4.5 h
ing/enhancing the normal conversion of proNGF after the stroke occurs, as tPA applied later or for
to mNGF and by diminishing the degradation of longer increases the risk of intracerebral
mNGF. Both are valid approaches, as both haemorrhage and subsequent mortality. Therapies
alterations occur in preclinical stages of AD and that increase tPA activity in AD, while a
correlate to AD neuropathology and cognitive promising means of elevating proNGF matura-
decline at these stages (Pentz et al. 2020), though tion, will therefore require careful dosing and
correlations to markers of cholinergic degenera- thorough analyses of potentially multifaceted
tion have only been observed for the maturation effects on wider AD pathology.
system. Below we will discuss various Neuroserpin, in contrast, is a tPA inhibitor that
approaches that could be used to target NGF has been demonstrated to be co-released with
metabolism in the context of prior preclinical proNGF in response to cholinergic stimulation
studies. and which has been repeatedly linked to NGF
metabolism through correlational and pharmaco-
logical studies (Bruno and Cuello 2006; Pentz et
4.8.1 Restoring Normal Conversion al. 2020). Neuroserpin has a restricted expression
of proNGF to mNGF pattern compared to other NGF pathway proteins,
with particularly high expression in the CNS,
How could proNGF maturation be promoted? particularly the isocortex and hippocampal for-
tPA, which mediates the formation of plasmin, mation (Osterwalder et al. 1996; Krueger et al.
is an already FDA-approved therapeutic for the 1997). As neuroserpin levels are elevated in
treatment of stroke (Group 1995) and has the MCI/AD brains, their normalization at these
potential to enhance proNGF maturation. stages would presumably have even more limited
Transgenesis experiments have demonstrated side effects as compared to a reduction in activity
that tg2576 mice deficient for the tPA gene from normal levels.
(PLAT /+) have increased cerebral amyloidosis Preclinical evidence suggests that neuroserpin
and mortality compared to PLAT +/+ littermates inhibition could be efficacious in AD. The partial
(Oh et al. 2014). In addition, preclinical studies knockdown of neuroserpin reduced amyloidosis
showed that chronic systemic administration of and enhanced cognition in the J20 mouse model
tPA to APPswe/PS1 transgenic mice reduced of AD (Fabbro et al. 2011), and indeed,
amyloidosis and enhanced cognition (ElAli et al. neuroserpin is known to have significant roles in
2016), which is also in line with its role in normal hippocampal plasticity (Hermann et al.
facilitating neurovascular coupling (Park et al. 2020). However, in vitro aggregation studies
2008). Furthermore, studies in which central and in vivo studies in transgenic drosophila flies
tPA inhibitors such as neuroserpin have been have shown that neuroserpin has the capacity to
knocked down or inhibited with small molecules bind Aβ42, thereby reducing its toxicity to
also demonstrated positive effects on amyloidosis cultured embryonic neurons (Kinghorn et al.
and cognition (Fabbro et al. 2011; Liu et al. 2011; 2006). Moreover, neuroserpin has unique phar-
Akhter et al. 2018). To further explore the macological properties that make it difficult to
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 43

target (Miranda and Lomas 2006), and there are MMP-3/MMP-9 inhibition on cholinergic integ-
consequently no known inhibitors. Therefore, rity, administration of an MMP-2/MMP-9 inhibi-
novel drugs and careful analyses of side effects tor (CAS 193807-58-8) has been shown to
will be required to effectively engage this target. increase mNGF levels and cortical cholinergic
tone after 2 weeks of continuous infusion (Allard
et al. 2012a). However, it should also be consid-
4.8.2 Restoring Normal mNGF ered that MMP-9 has a wide variety of targets
Degradation beyond NGF, including amyloid peptides
(Backstrom et al. 1996; Yan et al. 2006).
While less mNGF is produced than usual in AD, MMP-3, likewise, has a wide range of physiolog-
that which is produced is exposed to excessive ical roles (Van Hove et al. 2012), and its levels
levels of the metalloproteinases that degrade it differ between men and women (Iulita et al. 2016;
(Bruno et al. 2009a, 2009b; Iulita et al. 2014; Iulita et al. 2019; Pentz et al. 2020). Therefore,
Pentz et al. 2020), and inhibiting these while the preclinical evidence for MMP inhibi-
metalloproteinases is an equally viable approach tion in AD is promising, its inhibition could have
from pro-trophic, pro-cholinergic interventions in a wide range of potential side and sex-specific
AD. Additionally, it has recently been discovered effects that should be carefully considered.
that MMP-3, in addition to MMP-9, is competent New interest in potential disease-modifying
to degrade mNGF both in vitro and in vivo and is roles for cholinergic therapeutics has
elevated at preclinical disease stages and that reinvigorated this field (Hampel et al. 2018;
these elevated MMP-3 levels correlate to early Cuello et al. 2019). Of all the therapeutic avenues
decline on the MMSE in the AD continuum under development, targeting NGF metabolism
(Pentz et al. 2021b). Furthermore, both MMP-9 appears to have the greatest potential to elevate
and MMP-3 correlate to levels of mNGF in pre- NGF levels at physiological sites with minimally
clinical stages of AD, suggesting that they may be invasive procedures while addressing what is
relevant for adjusting the steady state of NGF likely to be the root cause of cholinergic degener-
metabolism. ation in AD. However, identifying potential drug
Preclinical work has previously demonstrated candidates is challenging, based on the highly
some efficacy for MMP-9 inhibition in AD. The varied localizations and activities that NGF path-
application of 100μg/kg of the MMP-9 inhibitor way proteins display, as well as the lack of spe-
GM6001, delivered intraparenchymally into the cific inhibitors for certain proteins, thus
cerebral cortices of young wild-type rats, caused underscoring an opportunity to develop novel
NGF levels to increase—in some cases, mature drugs with the capability of restoring normal
NGF could only be detected in the brain when NGF metabolism.
MMP-9 was inhibited (Bruno and Cuello 2006).
Recently, a study showed that injections of the
MMP-3-specific inhibitor UK356618 into the 4.9 Conclusion
hippocampi of young wild-type rats provoked a
marked increase in mNGF levels. Since this drug The discovery of the NGF metabolic pathway and
is highly selective (I.C. ¼ 5.9 nM), it could there- its essential role in the pathogenesis of choliner-
fore be applied at very low doses (Pentz et al. gic dysfunction in AD has opened new scientific
2021b). Additionally, minocycline, a broad- and therapeutic possibilities. ProNGF and NGF
spectrum MMP inhibitor and anti-inflammatory, pathway markers have demonstrated efficacy as
has previously been used to normalize the NGF indicators of the AD pathological process and
dysmetabolism induced by the intrahippocampal interventions targeting NGF pathway proteins
injection of amyloid oligomers to young naïve have shown promise in preclinical studies.
rats (Bruno et al. 2009a). Although the previously While specific compounds and assays remain to
discussed studies did not assess the effect of be developed, this new framework should offer
44 R. Pentz and M. F. Iulita

novel avenues for the clinical management Blennow K, Zetterberg H (2018) Biomarkers for
of AD. Alzheimer’s disease: current status and prospects for
the future. J Intern Med 284:643–663
Brookmeyer R, Abdalla N (2018) Estimation of lifetime
Acknowledgements The authors wish to thank and dedi- risks of Alzheimer’s disease dementia using
cate this chapter to Professor A. Claudio Cuello, for his biomarkers for preclinical disease. Alzheimers Dement
continued mentorship and support and for sharing with us 14:981–988
Rita Levi Montalcini’s inspirational legacy. Rowan Pentz Bruno MA, Cuello AC (2006) Activity-dependent release
is the recipient of a Returning Student Fellowship from the of precursor nerve growth factor, conversion to mature
McGill Integrated Program in Neuroscience and CIHR nerve growth factor, and its degradation by a protease
Doctoral Award no. 201610GSD-385496-2466860. cascade. Proc Natl Acad Sci 103:6735–6740
M. Florencia Iulita would like to acknowledge financial Bruno MA et al (2009a) Amyloid β-induced nerve growth
support from a Postdoctoral Research Fellowship from the factor dysmetabolism in Alzheimer disease. J
Jérôme Lejeune and Sisley D’Ornano Foundations and Neuropathol Exp Neurol 68:857–869
from a pilot exploratory grant (no. 1941) from the Jérôme Bruno MA et al (2009b) Increased matrix
Lejeune Foundation. Metalloproteinase-9 activity in mild cognitive
impairment. J Neuropathol Exp Neurol 68:1309
Caccamo A et al (2006) M1 receptors play a central role in
modulating AD-like pathology in transgenic mice.
References Neuron 49:671–682
Cai Z-Y et al (2007) Serum level of MMP-2, MMP-9 and
Adair JC et al (2004) Measurement of gelatinase B ox-LDL in Alzheimer’s disease with hyperlipoidemia.
(MMP-9) in the cerebrospinal fluid of patients with J Med Coll PLA 22:352–356
vascular dementia and Alzheimer disease. Stroke 35: Capsoni S et al (2011) Taking pain out of NGF: a “pain-
e159–e162 less” NGF mutant, linked to hereditary sensory auto-
Akhter H et al (2018) A small molecule inhibitor of plas- nomic neuropathy type V, with full neurotrophic
minogen activator inhibitor-1 reduces brain amyloid-β activity. PloS One 6:e17321
load and improves memory in an animal model of Capsoni S et al (2012) Intranasal “painless” human nerve
alzheimer’s disease. J Alzheimers Dis 64:447–457 growth factors slows amyloid neurodegeneration and
Allard S et al (2012a) Impact of the NGF maturation and prevents memory deficits in app X PS1 mice. PLoS
degradation pathway on the cortical cholinergic system One 7:e37555
phenotype. J Neurosci 32:2002–2012 Capsoni S et al (2017) The chemokine CXCL12 mediates
Allard S et al (2012b) Impact of the NGF maturation and the anti-amyloidogenic action of painless human nerve
degradation pathway on the cortical cholinergic system growth factor. Brain 140:201–217
phenotype. J Neurosci 32:2002–2012 Castle MJ et al (2020) Postmortem analysis in a clinical
Allard S et al (2018a) Compromise of cortical proNGF trial of AAV2-NGF gene therapy for Alzheimer’s dis-
maturation causes selective retrograde atrophy in cho- ease identifies a need for improved vector delivery.
linergic nucleus basalis neurons. Neurobiol Aging Hum Gene Ther 31:415–422
67:10–20 Cattaneo A, Capsoni S (2019) Painless nerve growth fac-
Allard S et al (2018b) Compromise of cortical proNGF tor: a TrkA biased agonist mediating a broad
maturation causes selective retrograde atrophy in cho- neuroprotection via its actions on microglia cells.
linergic nucleus basalis neurons. Neurobiol Aging Pharmacol Res 139:17–25
67:10–20 Cavedo E et al (2016) Reduced regional cortical thickness
Al-Shaikh FSH et al (2020) Selective vulnerability of the rate of change in donepezil-treated subjects with
nucleus basalis of Meynert among neuropathologic suspected prodromal Alzheimer’s disease. J Clin Psy-
subtypes of Alzheimer disease. JAMA Neurol chiatry 77(12):e1631–e1638
77:225–233 Cavedo E et al (2017) Reduced basal forebrain atrophy
Backstrom JR et al (1996) Matrix metalloproteinase-9 progression in a randomized donepezil trial in prodro-
(MMP-9) is synthesized in neurons of the human hip- mal Alzheimer’s disease. Sci Rep 7:11706
pocampus and is capable of degrading the amyloid-β Claassen JA, Jansen RW (2006) Cholinergically mediated
peptide (1–40). J Neurosci 16:7910–7919 augmentation of cerebral perfusion in alzheimer’s dis-
Bartus RT et al (1982) The cholinergic hypothesis of ease and related cognitive disorders: the cholinergic–
geriatric memory dysfunction. Science 217:408–414 vascular hypothesis. J Gerontol Ser A Biol Med Sci
Birks JS, Harvey RJ (2018) Donepezil for dementia due to 61:267–271
Alzheimer’s disease. Cochrane Database Syst Rev 6: Counts S et al (2016) Cerebrospinal fluid proNGF: a
CD001190 putative biomarker for early Alzheimer’s disease.
Bishop KM et al (2008) Therapeutic potential of CERE- Curr Alzheimer Res 13:800–808
110 (AAV2-NGF): targeted, stable, and sustained NGF Cuello AC (1996) Effects of trophic factors on the CNS
delivery and trophic activity on rodent basal forebrain cholinergic phenotype. Prog Brain Res 109:347–358
cholinergic neurons. Exp Neurol 211:574–584
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 45

Cuello A et al (2007) NGF-cholinergic dependency in releasing nerve growth factor. J Alzheimers Dis
brain aging, MCI and Alzheimer’s disease. Curr 43:1059–1072
Alzheimer Res 4:351–358 Figueiredo B et al (1995) Differential expression of
Cuello AC et al (2019) The brain NGF metabolic pathway p140trk, p75NGFR and growth-associated phospho-
in health and in Alzheimer’s pathology. Front Neurosci protein-43 genes in nucleus basalis magnocellularis,
13:62 thalamus and adjacent cortex following neocortical
Debeir T et al (1998) TrkA antagonists decrease infarction and nerve growth factor treatment. Neuro-
NGF-induced ChAT activity in vitro and modulate science 68:29–45
cholinergic synaptic number in vivo. J Physiol Paris Fisher A (2008) Cholinergic treatments with emphasis on
92:205–208 m1 muscarinic agonists as potential disease-modifying
Debeir T et al (1999a) A nerve growth factor mimetic agents for Alzheimer’s disease. Neurotherapeutics
TrkA antagonist causes withdrawal of cortical cholin- 5:433–442
ergic boutons in the adult rat. Proc Natl Acad Sci Fortea J et al (2018a) Plasma and CSF biomarkers for the
96:4067–4072 diagnosis of Alzheimer’s disease in adults with down
Debeir T et al (1999b) A nerve growth factor mimetic syndrome: a cross-sectional study. Lancet Neurol
TrkA antagonist causes withdrawal of cortical cholin- 17:860–869
ergic boutons in the adult rat. Proc Natl Acad Sci U S A Fortea J et al (2018b) Plasma and CSF biomarkers for the
96:4067–4072 diagnosis of Alzheimer’s disease in adults with down
Drachman DA, Leavitt J (1974) Human memory and the syndrome: a cross-sectional study. Lancet Neurol
cholinergic system. A relationship to aging? Arch 17:860–869
Neurol 30:113–121 Fortea J et al (2020) Clinical and biomarker changes of
Dubois B et al (2014) Advancing research diagnostic Alzheimer’s disease in adults with down syndrome: a
criteria for Alzheimer’s disease: the IWG-2 criteria. cross-sectional study. Lancet 395:1988–1997
Lancet Neurol 13:614–629 Giacobini E, Becker RE (2007) One hundred years after
Dubois B et al (2015) Donepezil decreases annual rate of the discovery of Alzheimer’s disease. A turning point
hippocampal atrophy in suspected prodromal for therapy? J Alzheimers Dis 12:37–52
Alzheimer’s disease. Alzheimers Dement Giacobini E et al (2002) Inhibition of acetyl- and butyryl-
11:1041–1049 cholinesterase in the cerebrospinal fluid of patients
ElAli A et al (2016) Tissue-plasminogen activator with Alzheimer’s disease by rivastigmine: correlation
attenuates Alzheimer’s disease-related pathology with cognitive benefit. J Neural Transm
development in APPswe/PS1 mice. Neuropsychophar- 109:1053–1065
macology 41:1297 Gibbs RB, Pfaff D (1994) In situ hybridization detection of
El-Hayek YH et al (2019) Tip of the iceberg: assessing the trkA mRNA in brain: distribution, colocalization with
global socioeconomic costs of Alzheimer’s disease and p75NGFR and up-regulation by nerve growth factor. J
related dementias and strategic implications for Comp Neurol 341:324–339
stakeholders. J Alzheimers Dis 70:323–341 Grothe MJ et al (2014) Basal forebrain atrophy and corti-
Eriksdotter M et al (2018) Cerebrospinal fluid from cal amyloid deposition in nondemented elderly
Alzheimer patients affects cell-mediated nerve growth subjects. Alzheimers Dement 10:S344–S353
factor production and cell survival in vitro. Exp Cell Group, N. I. o. N. D. S. r.-P. S. S (1995) Tissue plasmino-
Res 371:175–184 gen activator for acute ischemic stroke. N Engl J Med
Eriksdotter-Jönhagen M et al (2012) Encapsulated cell 333:1581–1588
biodelivery of nerve growth factor to the basal fore- Hall H et al (2018) AF710B, an M1/sigma-1 receptor
brain in patients with Alzheimer’s disease. Dement agonist with long-lasting disease-modifying properties
Geriatr Cogn Disord 33:18–28 in a transgenic rat model of Alzheimer’s disease.
Eyjolfsdottir H et al (2016) Targeted delivery of nerve Alzheimers Dement 14:811–823
growth factor to the cholinergic basal forebrain of Hampel H et al (2018) The cholinergic system in the
Alzheimer’s disease patients: application of a second- pathophysiology and treatment of Alzheimer’s disease.
generation encapsulated cell biodelivery device. Brain 141:1917–1933
Alzheimers Res Ther 8:30 Handen BL et al (2020) The Alzheimer’s biomarker
Fabbro S et al (2011) Amyloid-beta levels are significantly consortium-down syndrome: rationale and methodol-
reduced and spatial memory defects are rescued in a ogy. Alzheimers Dement (Amst) 12:e12065
novel neuroserpin-deficient Alzheimer’s disease trans- Hanzel CE et al (2014a) Analysis of matrix metallo-
genic mouse model. J Neurochem 118:928–938 proteases and the plasminogen system in mild cogni-
Fahnestock M et al (2001) The precursor pro-nerve growth tive impairment and Alzheimer’s disease cerebrospinal
factor is the predominant form of nerve growth factor fluid. J Alzheimers Dis 40:667–678
in brain and is increased in Alzheimer’s disease. Mol Hanzel CE et al (2014b) Neuronal driven pre-plaque
Cell Neurosci 18:210–220 inflammation in a transgenic rat model of Alzheimer’s
Ferreira D et al (2015) Brain changes in Alzheimer’s disease. Neurobiol Aging 35:2249–2262
disease patients with implanted encapsulated cells
46 R. Pentz and M. F. Iulita

Hao J-X et al (2000) Intracerebroventricular infusion of diagnosis, neuropathology and longitudinal progres-
nerve growth factor induces pain-like response in rats. sion to Alzheimer’s dementia. Nat Med 26:379–386
Neurosci Lett 286:208–212 Jönhagen ME (2000) Nerve growth factor treatment in
Hasselmo ME, Sarter M (2011) Modes and models of dementia. Alzheimer Dis Assoc Disord 14:S31–S38
forebrain cholinergic neuromodulation of cognition. Jönhagen ME et al (1998) Intracerebroventricular infusion
Neuropsychopharmacology 36:52–73 of nerve growth factor in three patients with
Head E, Lott IT (2004) Down syndrome and beta-amyloid Alzheimer’s disease. Dement Geriatr Cogn Disord
deposition. Curr Opin Neurol 17:95–100 9:246–257
Head E et al (2012) Aging and down syndrome. Curr Karami A et al (2015) Changes in CSF cholinergic
Gerontol Geriatr Res 2012:412536 biomarkers in response to cell therapy with NGF in
Hefti F (1986) Nerve growth factor promotes survival of patients with Alzheimer’s disease. Alzheimers Dement
septal cholinergic neurons after fimbrial transections. J 11:1316–1328
Neurosci 6:2155–2162 Karami A et al (2019) CSF cholinergic index, a new
Hefti F et al (1986) Localization of nerve growth factor biomeasure of treatment effect in patients with
receptors in cholinergic neurons of the human basal Alzheimer’s disease. Frontiers in molecular. Neurosci-
forebrain. Neurosci Lett 69:37–41 ence 12:239
Hermann M et al (2020) Deficits in developmental Karikari TK et al (2020) Blood phosphorylated tau 181 as
neurogenesis and dendritic spine maturation in mice a biomarker for Alzheimer’s disease: a diagnostic per-
lacking the serine protease inhibitor neuroserpin. Mol formance and prediction modelling study using data
Cell Neurosci 102:103420 from four prospective cohorts. Lancet Neurol
Hock C et al (2000) Increased CSF levels of nerve growth 19:422–433
factor in patients with Alzheimer’s disease. Neurology Kinghorn KJ et al (2006) Neuroserpin binds Aβ and is a
54:2009–2011 neuroprotective component of amyloid plaques in
Holtzman DM et al (1992) p140trk mRNA marks Alzheimer disease. J Biol Chem 281:29268–29277
NGF-responsive forebrain neurons: evidence that trk Krueger SR et al (1997) Expression of neuroserpin, an
gene expression is induced by NGF. Neuron inhibitor of tissue plasminogen activator, in the devel-
9:465–478 oping and adult nervous system of the mouse. J
Horstmann S et al (2010) Matrix metalloproteinases in Neurosci 17:8984–8996
peripheral blood and cerebrospinal fluid in patients Le AP (2011) Regulation of proNGF processing and its
with Alzheimer’s disease. Int Psychogeriatr effects on p75NTR-mediated cell death following sei-
22:966–972 zure. Rutgers University-Graduate School-Newark
Iulita MF, Cuello AC (2014) Nerve growth factor meta- Lim NK-H et al (2011) Investigation of matrix metallopro-
bolic dysfunction in Alzheimer’s disease and down teinases, MMP-2 and MMP-9, in plasma reveals a
syndrome. Trends Pharmacol Sci 35:338–348 decrease of MMP-2 in Alzheimer’s disease. J
Iulita MF, Cuello AC (2016) The NGF metabolic pathway Alzheimers Dis 26:779–786
in the CNS and its Dysregulation in down syndrome Lin Z et al (2006) The plasma matrix metalloproteinase-3
and Alzheimer’s disease. Curr Alzheimer Res increased in patients with Alzheimer’s disease. Chin J
13:53–67 Psychiatry 39:209
Iulita MF et al (2014) Nerve growth factor metabolic Liu R-M et al (2011) Knockout of plasminogen activator
dysfunction in Down’s syndrome brains. Brain 137 inhibitor 1 gene reduces amyloid beta peptide burden
(Pt 3):860–872. https://doi.org/10.1093/brain/awt372 in a mouse model of Alzheimer’s disease. Neurobiol
Iulita MF et al (2016) An inflammatory and trophic dis- Aging 32:1079–1089
connect biomarker profile revealed in down syndrome Lorenzl S et al (2003) Increased plasma levels of matrix
plasma: relation to cognitive decline and longitudinal metalloproteinase-9 in patients with Alzheimer’s dis-
evaluation. Alzheimers Dement 12(11):1132–1148 ease. Neurochem Int 43:191–196
Iulita MF et al (2017) Differential deregulation of NGF Lorenzl S et al (2008) Profiles of matrix metallopro-
and BDNF neurotrophins in a transgenic rat model of teinases and their inhibitors in plasma of patients with
Alzheimer’s disease. Neurobiol Dis 108:307–323 dementia. Int Psychogeriatr 20:67–76
Iulita MF et al (2019) Identification and preliminary vali- Lott IT, Head E (2019) Dementia in down syndrome:
dation of a plasma profile associated with cognitive unique insights for Alzheimer disease research. Nat
decline in dementia and at-risk individuals: a retrospec- Rev Neurol 15:135–147
tive cohort analysis. J Alzheimers Dis 67:327–341 Machado A et al (2020) The cholinergic system in
Jack CR Jr et al (2018) NIA-AA research framework: subtypes of Alzheimer’s disease: an in vivo longitudi-
toward a biological definition of Alzheimer’s disease. nal MRI study. Alzheimers Res Ther 12:1–11
Alzheimers Dement 14:535–562 Malerba F et al (2016) NGF and proNGF reciprocal inter-
Jack CR Jr (2020) The transformative potential of plasma ference in immunoassays: open questions, criticalities,
phosphorylated tau. Lancet Neurol 19:373–374 and ways forward. Front Mol Neurosci 9:63
Janelidze S et al (2020) Plasma P-tau181 in Alzheimer’s Manuel DG et al (2016) Alzheimer’s and other dementias
disease: relationship to other biomarkers, differential in Canada, 2011 to 2031: a microsimulation population
4 The NGF Metabolic Pathway: New Opportunities for Biomarker Research and. . . 47

health modeling (POHEM) study of projected preva- Olson L et al (1992) Nerve growth factor affects
lence, health burden, health services, and caregiving 11 C-nicotine binding, blood flow, EEG, and verbal
use. Popul Health Metr 14:37 episodic memory in an Alzheimer patient (case report).
Marksteiner J et al (2014) Analysis of 27 vascular-related J Neural Transm–Parkinson’s disease and dementia
proteins reveals that NT-proBNP is a potential bio- section 4:79–95
marker for Alzheimer’s disease and mild cognitive Osterwalder T et al (1996) Neuroserpin, an axonally
impairment: a pilot-study. Exp Gerontol 50:114–121 secreted serine protease inhibitor. EMBO J
Martín-Aragón S et al (2009) Metalloproteinase’s activity 15:2944–2953
and oxidative stress in mild cognitive impairment and Overk CR et al (2010) Cortical M1 receptor concentration
Alzheimer’s disease. Neurochem Res 34:373 increases without a concomitant change in function in
Martorana A et al (2012) Plasmin system of Alzheimer’s Alzheimer’s disease. J Chem Neuroanat 40:63–70
disease patients: CSF analysis. J Neural Transm Palmqvist S et al (2020) Discriminative accuracy of
119:763–769 plasma Phospho-tau217 for Alzheimer disease vs
Massaro A et al (1994) Nerve growth factor (NGF) in other neurodegenerative disorders. JAMA
cerebrospinal fluid (CSF) from patients with various 324:772–781
neurological disorders. Ital J Neurol Sci 15:105–108 Park L et al (2008) Key role of tissue plasminogen activa-
McDade E, Bateman RJ (2017) Stop Alzheimer’s before it tor in neurovascular coupling. Proc Natl Acad Sci
starts. Nature 547:153 105:1073–1078
McKelvey L et al (2013) Nerve growth factor-mediated Parks WC et al (2004) Matrix metalloproteinases as
regulation of pain signalling and proposed new inter- modulators of inflammation and innate immunity. Nat
vention strategies in clinical pain management. J Rev Immunol 4:617
Neurochem 124:276–289 Peng M et al (2015) Plasma gelsolin and matrix
Medina MG et al (2005) Tissue plasminogen activator metalloproteinase 3 as potential biomarkers for
mediates amyloid-induced neurotoxicity via Erk1/ Alzheimer disease. Neurosci Lett 595:116–121
2 activation. EMBO J 24:1706–1716 Pentz R et al (2020) The human brain NGF metabolic
Mehra A et al (2016) The plasminogen activation system pathway is impaired in the pre-clinical and clinical
in neuroinflammation. Biochim Biophys Acta (BBA)- continuum of Alzheimers disease. Mol Psych 1–15
molecular basis of disease 1862:395–402 Pentz R et al (2021a) Nerve growth factor (NGF) pathway
Miranda E, Lomas D (2006) Neuroserpin: a serpin to think biomarkers in Down syndrome prior to and after the
about. Cell Mol Life Sci CMLS 63:709–722 onset of clinical Alzheimer’s disease: a paired CSF and
Mlekusch R, Humpel C (2009) Matrix metalloproteinases- plasma study. Alzheimers Dement 17(4):605–617
2 and-3 are reduced in cerebrospinal fluid with low Pentz R et al (2021b) A new role for matrix
beta-amyloid1–42 levels. Neurosci Lett 466:135–138 metalloproteinase-3 in the NGF metabolic pathway:
Mroczko B et al (2014) Concentrations of matrix proteolysis of mature NGF and sex-specific differences
metalloproteinases and their tissue inhibitors in the in the continuum of Alzheimer’s pathology. Neurobiol
cerebrospinal fluid of patients with Alzheimer’s dis- Dis 148:105150
ease. J Alzheimers Dis 40:351–357 Pepeu G, Giovannini MG (2010) Cholinesterase inhibitors
Mufson EJ, Kordower JH (1992) Cortical neurons express and memory. Chem Biol Interact 187:403–408
nerve growth factor receptors in advanced age and Rafii MS et al (2014) A phase 1 study of stereotactic gene
Alzheimer disease. Proc Natl Acad Sci 89:569–573 delivery of AAV2-NGF for Alzheimer’s disease.
Mufson EJ et al (2019) Nerve growth factor pathobiology Alzheimers Dement 10:571–581
during the progression of Alzheimer’s disease. Front Rafii MS et al (2018) Adeno-associated viral vector (sero-
Neurosci 13:533 type 2)–nerve growth factor for patients with alzheimer
Murray ME et al (2011) Neuropathologically defined disease: a randomized clinical trial. JAMA Neurol
subtypes of Alzheimer’s disease with distinct clinical 75:834–841
characteristics: a retrospective study. Lancet Neurol Richter N et al (2018) Effect of cholinergic treatment
10:785–796 depends on cholinergic integrity in early Alzheimer’s
Nielsen HM et al (2007) Plasma and CSF serpins in disease. Brain 141:903–915
Alzheimer disease and dementia with Lewy bodies. Risacher SL et al (2016) Association between anticholin-
Neurology 69:1569–1579 ergic medication use and cognition, brain metabolism,
Nitsch RM et al (1992) Release of Alzheimer amyloid and brain atrophy in cognitively normal older adults.
precursor derivatives stimulated by activation of mus- JAMA Neurol 73:721–732
carinic acetylcholine receptors. Science 258:304–307 Rouy D et al (2005) Plasma storage at 80 C does not
Nizri E et al (2006) Anti-inflammatory properties of cho- protect matrix metalloproteinase-9 from degradation.
linergic up-regulation: a new role for acetylcholinester- Anal Biochem 338:294–298
ase inhibitors. Neuropharmacology 50:540–547 Schmitz TW et al (2016) Basal forebrain degeneration
Oh SB et al (2014) Tissue plasminogen activator arrests precedes and predicts the cortical spread of
Alzheimer’s disease pathogenesis. Neurobiol Aging Alzheimer’s pathology. Nat Commun 7:13249
35:511–519
48 R. Pentz and M. F. Iulita

Schmitz TW et al (2018) Longitudinal Alzheimer’s degen- Tuszynski MH et al (1991) Recombinant human nerve
eration reflects the spatial topography of cholinergic growth factor infusions prevent cholinergic neuronal
basal forebrain projections. Cell Rep 24:38–46 degeneration in the adult primate brain. Ann Neurol:
Shen H et al (2010) Neuroprotection by donepezil against Official Journal of the American Neurological Associ-
glutamate excitotoxicity involves stimulation of α7 ation and the Child Neurology Society 30:625–636
nicotinic receptors and internalization of NMDA Tuszynski MH et al (2005) A phase 1 clinical trial of nerve
receptors. Br J Pharmacol 161:127–139 growth factor gene therapy for Alzheimer disease. Nat
Shi Y et al (2010) Plasma BDNF and tPA are associated Med 11:551–555
with late-onset geriatric depression. Psychiatry Clin Tuszynski MH et al (2015) Nerve growth factor gene
Neurosci 64:249–254 therapy: activation of neuronal responses in Alzheimer
Siao C-J, Tsirka SE (2002) Tissue plasminogen activator disease. JAMA Neurol 72:1139–1147
mediates microglial activation via its finger domain Van Hove I et al (2012) Matrix metalloproteinase-3 in the
through annexin II. J Neurosci 22:3352–3358 central nervous system: a look on the bright side. J
Stomrud E et al (2010) Alterations of matrix metallopro- Neurochem 123:203–216
teinases in the healthy elderly with increased risk of Venero J et al (1994) Expression of neurotrophin and trk
prodromal Alzheimer’s disease. Alzheimer’s Res Ther receptor genes in adult rats with fimbria transections:
2:20 effect of intraventricular nerve growth factor and brain-
Svensson AL et al (1992) Characterization of muscarinic derived neurotrophic factor administration. Neurosci-
receptor subtypes in Alzheimer and control brain corti- ence 59:797–815
ces by selective muscarinic antagonists. Brain Res Welikovitch LA et al (2020) Early intraneuronal amyloid
596:142–148 triggers neuron-derived inflammatory signaling in APP
Thijssen EH et al (2020) Diagnostic value of plasma transgenic rats and human brain. Proc Natl Acad Sci
phosphorylated tau181 in Alzheimer’s disease and 117:6844–6854
frontotemporal lobar degeneration. Nat Med Whelan CD et al (2019) Multiplex proteomics identifies
26:387–397 novel CSF and plasma biomarkers of early
Tuna G et al (2018) Evaluation of matrix Alzheimer’s disease. Acta Neuropathol Commun
Metalloproteinase-2 (MMP-2) and-9 (MMP-9) and 7:1–14
their tissue inhibitors (TIMP-1 and TIMP-2) in plasma Yan P et al (2006) Matrix metalloproteinase-9 degrades
from patients with neurodegenerative dementia. J amyloid-β fibrils in vitro and compact plaques in situ. J
Alzheimers Dis 66:1265–1273 Biol Chem 281:24566–24574
Tuszynski MH et al (1990) Nerve growth factor infusion in Zissimopoulos J et al (2015) The value of delaying
the primate brain reduces lesion-induced cholinergic Alzheimer’s disease onset. Forum Health Econ Policy
neuronal degeneration. J Neurosci 10:3604–3614 18:25–39. De Gruyter
Part II
Development and Regeneration
NGF and Endogenous Regeneration:
From Embryology Toward Therapies 5
Vito Antonio Baldassarro, Luca Lorenzini, Andrea Bighinati,
Alessandro Giuliani, Giuseppe Alastra, Micaela Pannella,
Mercedes Fernandez, Luciana Giardino, and Laura Calzà

Abstract through the regulation of the stem cell niches.


The self-repair ability of tissues and organs in In fact, this growth factor is very promising for
case of injury and disease is a fundamental its key role in the development and multiplic-
biological mechanism and an important thera- ity of the cellular targets.
peutic target. The tissue plasticity and the pres- In this chapter, we have traveled across the
ence of adult stem cell niches open a new path recent history of NGF pleiotropic role in ecto-
in the development of pharmacological and dermal tissue generation and repair, from
non-pharmacological treatments finalized to embryonic development to skin wound
improve the intrinsic regeneration. healing, axonal regrowth, and remyelination.
In this context, nerve growth factor (NGF) The better understanding of both the
is widely studied for its capability of driving biological mechanisms underlying regenera-
endogenous regeneration of ectoderm-derived tion and the physiological role of NGF in
tissues, directly acting on the cell targets and development and injury response will open
new therapeutic strategies, driven by the

V. A. Baldassarro · G. Alastra
Health Science and Technologies Interdepartmental
Center for Industrial Research (HST-ICIR), University of
L. Giardino
Bologna, Ozzano Emilia, Bologna, Italy
Department of Veterinary Medical Sciences, University of
e-mail: vito.baldassarro2@unibo.it; giuseppe.
Bologna, Bologna, Italy
alastra2@unibo.it
Health Science and Technologies Interdepartmental
L. Lorenzini
Center for Industrial Research (HST-ICIR), University of
Health Science and Technologies Interdepartmental
Bologna, Bologna, Italy
Center for Industrial Research (HST-ICIR), University of
Bologna, Ozzano Emilia, Bologna, Italy IRET Foundation, Bologna, Italy
e-mail: luciana.giardino@unibo.it
Department of Veterinary Medical Sciences, University of
Bologna, Ozzano Emilia, Bologna, Italy L. Calzà (*)
e-mail: luca.lorenzini8@unibo.it Health Science and Technologies Interdepartmental
Center for Industrial Research (HST-ICIR), University of
A. Bighinati · A. Giuliani · M. Fernandez
Bologna, Ozzano Emilia, Bologna, Italy
Department of Veterinary Medical Sciences, University of
Bologna, Ozzano Emilia, Bologna, Italy Department of Pharmacy and Biotechnology, University
e-mail: andrea.bighinati@unibo.it; a.giuliani@unibo.it; of Bologna, Bologna, Italy
mercedes.fernandez@unibo.it
Montecatone Rehabilitation Institute, Montecatone,
M. Pannella Bologna, Italy
IRET Foundation, Ozzano Emilia, Bologna, Italy e-mail: laura.calza@unibo.it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 51


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_5
52 V. A. Baldassarro et al.

potential applications of this growth factor as is also defined “in vivo regenerative medicine” or
an agent for improving endogenous regenera- “autotherapies” (Lumelsky et al. 2018).
tion processes. The role of growth and neurotrophic factors
during development makes these biomolecules
natural candidates for improving endogenous
regeneration. NGF in particular is very attractive
5.1 Introduction from this point of view, being a pleiotropic
molecule acting on many different cell types,
“Endogenous regeneration” refers to the ability of both during development and in adulthood
tissues and organs to self-repair in the event of (Aloe and Calzà 2004). In this short review,
injury or disease. This capability varies signifi- we will focus on data supporting the potential
cantly between the different animal species and applications of NGF as an agent for improving
districts of the body; tailed amphibians, for exam- endogenous regeneration of ectoderm-derived
ple, can completely regenerate parts of the body, tissues, summarizing the basic and translational
such as limbs, tails, jaws, eyes, and a variety of findings supporting the development of NGF as
internal structures. In mammals, tissues with a a drug.
high cell turnover such as the skin may effectively
regenerate in the event of extensive lesions, while
the nervous tissue, such as that of the central 5.2 NGF and Embryonic
nervous system (CNS), has a very limited regen- Development
erative capability, confined to the myelin sheath.
In both cases, this capability may be severely NGF and its receptors appear very early during
impaired by disease or excessive lesion area. development (reviewed by Bracci-Laudiero and
Since the discovery of stem cell niches in most De Stefano 2016). The NGF mRNA is found in
adult mammalian tissues and organs, which con- oocytes at different maturation stages and is
tribute to the respective and tissue-specific cell involved in follicle and oocyte maturation.
turnover rate (Bagnara 2020), the self-repair capa- p75NTR and TrkA appear in mouse embryos at
bility of adult tissue has been the focus of increas- the blastocyst stage, being confined to the inner
ing interest. In some tissues, such as the skin, the cell mass and absent in the trophoblast. NGF
proliferation rate of niche stem and precursor receptor expression then differentiates into spe-
cells increases in the event of injury, while asym- cific areas during gastrulation and neurulation,
metric division provides precursors, thus increas- suggesting that NGF plays a role in germinal
ing cell turnover, and ultimately leading to tissue layer differentiation and early body shape specifi-
repair. In other tissues, such as that of the CNS, cation. NGF neutralization experiments in
stem cells in the neurogenic niches react weakly chickens indicate that NGF absence alters body
to injuries, an activation which has no substantial axis rotation as well as notochord and somite
effects on lesion or pathology evolution (Sun formation and evolution.
2016). On the contrary, oligodendrocyte precur- During neurulation, p75NTR expression in neu-
sor cells disseminated throughout the white and ral crest cells correlates with the beginning of cell
gray matter, or newly generated from the migration (Wislet et al. 2018). The different
glycogenic niches, may efficiently repair the mye- isoforms of Trk receptors then appear during neu-
lin, restoring appropriate axonal function (Bruce ral crest cell migration and differentiation
et al. 2010). (Martin-Zanca et al. 1990).
The subsequent hypothesis was that organ and Both mouse (Moscatelli et al. 2009) and
tissue plasticity, and endogenous regeneration as a human embryonic stem cells (Pyle et al. 2006)
result, could be improved by pharmacological and express NGF receptors, and differentiation stud-
non-pharmacological treatments able to direct the ies of 3D embryonic stem cell cultures indicate
stem cell niches (Wells and Watt 2018). This area that exposure to NGF plus retinoic acid
5 NGF and Endogenous Regeneration: From Embryology Toward Therapies 53

(RA) favors the ectodermal and mesodermal toward differentiation, gradually losing their
lineages (Inanç et al. 2008). pluripotency to become specific germinative
We studied the expression of NGF and its layer committed cells.
receptors in embryonic stem cells derived from Figure 5.1 shows the mRNA expression pro-
rat blastocytes (RESCs), using a cell line isolated file of NGF, trkA, and p75NTR under these differ-
in our laboratory (Fernández et al. 2011) ent in vitro conditions. We observed that all
(Fig. 5.1). These cells can be maintained in sev- mRNAs are expressed in the aforementioned cul-
eral ways: first as clusters (CL), growing in the ture conditions, with a significant increase of
presence of LIF (leukemia inhibitory factor) and p75NTR expression level in EBs, around
bFGF (basic-fibroblast growth factor) in ultra-low 10 times higher than in CL (**p ¼ 0.0077) and
attachment plates; second as a mixed population all the other studied conditions (SC, CL mixed
of single cells (SC) and CL, growing in the pres- population, **p ¼ 0.0093, p ¼ 0.0091). Interest-
ence of LIF and bFGF, in attaching and floating ingly, the trkA expression was significantly lower
conditions, respectively; third as embryoid bodies (around 0.5-fold) in single cells only compared to
(EB), whose formation can be induced by grow- the reference group (**p ¼ 0.0084).
ing the cells obtained from cluster splitting NGF also regulates organogenesis and tissue
(RESC-CL) in ultra-low attachment plates with- maturation in several districts, acting through
out LIF but in the presence of bFGF (Pannella both sensory innervation and angiogenesis (Kim
et al. 2018). These EBs recapitulate the first steps et al. 2013), as in the case of endochondral bone,

A B C D

E NGF mRNA F TrkA mRNA G P75 mRNA


2 2 15 **
R e la tiv e e x p re s s io n

**
n o rm a liz e d to C L

**

**
10

1 1
5

0 0 0
c l s c l s L c l s
c B
L

s
L

s c B s c B C
C

E
C

E E
L

mixed population mixed population mixed population

Floating + - + +

LIF + + + -
bFGF + + + +

Fig. 5.1 NGF, trkA, and p75 gene expression in RESCs. independent experiments performed in duplicate are
Representative images of RESCs grown as CLs (a), mixed included in the graphs. Statistical analysis was performed
population of SCs and CLs (b, c), and EBs (d) in the using one-way ANOVA and Tukey’s multiple comparison
specified culture conditions and mediums containing the test. Significant results were fixed at p < 0.05.
mitogens indicated. Graphs show relative mRNA expres- Abbreviations: bFGF, basic fibroblast growth factor; CL,
sion of NGF (e) and its receptors, trkA (f) and p75 (g), clusters; EBs, embryoid bodies; LIF, leukemia inhibitory
normalized to CL. Mean values SEM from three factor; SC, single cells
54 V. A. Baldassarro et al.

but also acts directly on non-neural cells, such as particular, the development of thinly myelinated
during odontogenesis (Mitsiadis and Pagella Aδ or unmyelinated C-fibers that innervate the
2016). skin is NGF-dependent (Indo 2012), and half of
Finally, classical neutralization experiments the nociceptive sensory neurons innervating the
have indicated that several neural populations mature skin are dependent upon an appropriate
and pathways in the peripheral nervous system NGF supply (Snider and McMahon 1998).
(PNS), including the sensory and sympathetic Neurotrophins, including NGF, play a complex
neurons, and selective neural populations in the role in the hair follicle cycle (Botchkarev et al.
CNS, such as the cholinergic neurons of the basal 2004), and the low-affinity neurotrophin receptor
forebrain, require NGF for appropriate axonal is a key regulator of the melanocytic cell lineage
growth, cell differentiation, cell migration, and (Kasemeier-Kulesa and Kulesa 2018). During
promotion and control of nerve regeneration development, cell types other than
(Levi-Montalcini 1997). The sympathetic keratinocytes—for example, melanocytes—also
neurons of the superior cervical ganglion are the require NGF, which derives from the neural
best-studied example of the role of NGF in crest: p75NTR is in fact a marker for neural crest
controlling pro-survival vs. pro-apoptotic cells and is thus used for cell sorting experiments.
pathways during development. These cells Recently, p75NTR - (CD271) positivity has been
require NGF at the time of target innervation, associated with an aggressive clinical subtype of
and in vitro experiments indicate that NGF with- melanoma which metastasizes to the brain, being
drawal activates the mitochondrial (intrinsic) indicated as a mediator of phenotype switching
pathway of apoptosis involving caspases, Bcl-2 which suppresses melanoma cell proliferation,
(B-cell lymphoma 2) family proteins, and XIAP while concomitantly promoting metastasis forma-
(X-linked inhibitor of apoptosis protein) tion in vivo (Kasemeier-Kulesa and Kulesa
(Kristiansen and Ham 2014). 2018).
However, the role of NGF in mature neural The breakdown of skin integrity, when it is
phenotype maintenance within the adult nervous incorrectly and rapidly repaired, results in a sub-
system, in cell reaction to lesion as either survival stantial physiological imbalance and renders a
or repair, and as a pro-apoptotic factor, is still patient vulnerable to a number of pathological
disputed, being dependent on: (1) the involved conditions, such as infection, fluid loss, and elec-
receptor subtype, (2) the synthesis pathway from trolyte imbalance (Reinke and Sorg 2012),
the pro-NGF, and (3) the target cell type. underlining the critical importance of a complete
and efficient healing of cutaneous wounds. The
wound healing process is characterized by four
5.3 NGF and Epithelial Wound sequential and overlapping phases: hemostasis
Healing and early inflammation (0–several hours after
injury), inflammation (1–3 days), proliferation
The skin is the largest organ in the body, covering (4–21 days), and maturation and remodeling
its entire external surface, and forms the physical (21 days–1 year) (Reinke and Sorg 2012). Dereg-
barrier between the host and the external environ- ulation of any of these steps results in impaired
ment. The skin protects the entire body against healing, potentially leading to chronic ulcers (Sen
numerous insults, prevents excessive body water et al. 2009).
loss, and plays a role in immune surveillance and Several in vitro and in vivo studies have
sensory detection. shown that NGF affects several cell types
The epithelial component of the skin derives involved in skin wound healing. TrkA and/or
from the embryonic surface ectoderm, whereas p75NTR expression is not limited to sensory
the underlying derma derives from the mesoderm. nerve endings but also includes inflammatory
During development, NGF plays a major role in cells (neutrophils, macrophages, mast cells, and
the development of selective skin components: in lymphocytes), keratinocytes, fibroblasts, and
5 NGF and Endogenous Regeneration: From Embryology Toward Therapies 55

endothelial cells. NGF is also secreted by full-thickness excision model. One limiting side
fibroblasts (Matsuda et al. 1991), keratinocytes effect of the therapeutic use of NGF is local
(Tron et al. 1990), mast cells (Leon et al. 1994), hyperalgesia to mechanical and electrical stimuli
and T cells (Ehrhard et al. 1993), and has angio- lasting for weeks, due to nociceptor sensitization
genic properties, via nitric oxide synthase (NOS) (Obreja et al. 2018).
and vascular endothelial growth factor (VEGF) The use of NGF as therapeutic agent has been
(Calzà et al. 2001). approved for lesions of the corneal epithelium
We recently demonstrated that human (Sacchetti et al. 2020), and recombinant human
fibroblasts (BJ cells) and keratinocytes (HEK NGF has been an approved drug (Oxervate®)
cells), as well as human endothelial cells since 2017 as eye drops for neurotrophic keratitis.
(HUVECs), express both high- (TrkA) and
low-affinity (p75NTR) receptors for NGF, and
key physiological properties of these cells, such 5.4 NGF and Axonal Regrowth
as proliferation, migration, and angio-tube forma-
tion, are affected by NGF exposure in both phys- NGF exerts multiple effects on NGF-sensitive
iological and pathological (hyperglycemia) neural pathway formation, including axonal elon-
conditions (Gostynska et al. 2019). gation and guidance (Guthrie 2007). Moreover,
The underlying molecular mechanism, NGF binding at TrkA or p75NTR receptors
activated by TrkA, includes the PI3K/Akt and activates either pro-survival or pro-apoptotic
ERK/MAPK signaling pathways and the down- pathways. During development, nervous system
stream mTOR. Notably, several NGF-mediated post-synaptic cells are responsible for the major-
effects during development, such as endothelial ity of NGF release, thus directing the appropriate
cell invasion and cord formation (Park et al. target innervation. The binding of NGF to TrkA
2007), angiogenesis in bone (Yu et al. 2019), as on sympathetic and sensory axon terminals, for
well as neuroprotection (Elsherbiny et al. 2019), example, leads to receptor dimerization and inter-
activate these pathways. Moreover, an nalization; then the complex NGF/TkA is retro-
impairment of the AKT-mTOR pathway has gradely transported to the neuronal cell body,
been indicated as a possible cause of wound where it activates molecular pathways for neuro-
healing impairment in diabetic mice (Jere et al. nal survival and differentiation (Johnson et al.
2019). 1980). Neuronal cell survival is mediated by the
A role of endogenous NGF in wound healing phosphorylation and activation of phosphatidy-
has been suggested by several studies in animal linositol 3-kinase (PI-3K) and MAPK which
models, human pathologies, and in vitro models. leads to Akt activation, the final result of which
In particular, the diabetic skin is characterized by is the inhibition of pro-apoptotic genes (Bim and
a reduced epidermal innervation, considered to be FasL) in the developing neuron (Reichardt 2006).
a main cause of diabetic “small-fiber neuropathy” The role of p75NTR in nervous system devel-
(Ebenezer and Polydefkis 2014), leading to opment is complex, promoting either survival and
chronic ulcers (Laverdet et al. 2015). Notably, a growth or apoptosis and degeneration, depending
reduction in neurotrophic support to PNS neurons on the expression of its coreceptors and ligands.
has been indicated as responsible for peripheral p75NTR-TrkA complexes elicit pro-survival and
neuropathy (Sima 2003). axonal growth signaling in response to NGF,
Topical application of NGF has therefore been while neurotrophin binding to p75NTR alone, or
considered as a possible therapy for skin wound the binding of proNGF, can trigger cell death and
healing in chronic ulcers (Tuveri et al. 2000; axonal degeneration. Moreover, p75NTR intracel-
Generini et al. 2004), and several in vivo studies lular trafficking is not yet fully understood
report the positive effect on fibroblast migration, (Edward Hickman et al. 2018).
re-epithelialization and capillary lumen forma- In addition to the neurotrophic effect mediated
tion, and an increase in wound contraction in the by the action on pro-survival genes, NGF
56 V. A. Baldassarro et al.

promotes neurite and axonal elongation, the The PNS and the CNS differ substantially in
biological effect from which this neurotrophin their ability to respond and regenerate axons after
takes its name, as derived from the historical damage. In both districts, the “sprouting” of a
experiments by Levi-Montalcini and Hamburger. stump occurs following axon transection, but
The original experiments described the sympa- this sprouting is followed by axonal elongation
thetic innervation of solid tumors secreting this in the PNS only, while this attempt aborts in the
molecule, when ectopically implanted in chick CNS (Cafferty et al. 2010). The main reason for
embryos (Levi-Montalcini and Hamburger this different behavior, as proven by Aguayo and
1951). In this system, NGF acts as collaborators’ pioneering work in bridging spinal
chemoattractant for neurites, and it has been cord lesions with peripheral nerve grafts, seems to
demonstrated that the elongation of neuron pro- be the non-permissive microenvironment of the
cesses mediated by NGF depends on neurite CNS compared to the PNS (Richardson et al.
arborization rather than retrograde axonal trans- 1980; David and Aguayo 1981; Benfey and
port for its action. In fact, as demonstrated in an Aguayo 1982; Richardson and Riopelle 1984).
in vitro model of sympathetic neuron cultures, This microenvironment is determined by several
NGF application to the cell body compartment factors, including inflammatory stimuli (glial acti-
does not promote neurite extension retrogradely vation and scarring in the CNS versus Wallerian
or anterogradely, while NGF application to the degeneration in the PNS), myelin and cellular
neurite compartment promotes neurite elonga- debris from the core of the injury, extracellular
tion, depending on concentration gradient matrix composition, availability of growth-
(Campenot 1994). promoting proteins, and different remyelinating
At molecular level, TrkA activation in neuro- cells (oligodendrocytes and astrocytes in the CNS
nal cells leads to an increase in β-actin concentra- versus Schwann cells and macrophages in the
tion and its polymerization in the growth cones, PNS) (Mietto et al. 2015).
ultimately resulting in axonal elongation via In the CNS, maintenance of the basal cholin-
PI-3K and PLC-γ activation and subsequent sta- ergic system phenotype is NGF-dependent, but
bilization by de-phosphorylation of the actin- spontaneous axonal regrowth does not occur
stabilizing complex ADF/Cofilin (Meberg et al. even in the event of extreme axonal degeneration,
1998). The activation of p75NTR in the develop- as in the case of experimental lesions or in
ing nervous system also leads to growth cone Alzheimer’s disease (Mesulam et al. 2019). In
stabilization and axon elongation through inacti- the PNS, damage to the adult nerves induces a
vation of RhoA, which normally phosphorylates rapid and robust increase in the synthesis of
and destabilizes ADF/Cofilin via the ROCK cas- neurotrophins by the surrounding milieu near
cade (Yamashita et al. 1999). the lesion. NGF synthesis, for example, increases
The role of NGF on the target neural popula- in the sensory neurons in the dorsal root ganglia
tion in the mature nervous system is much more (DRG), thus reactivating the cellular mechanisms
disputed, and differs in the CNS and PNS. It is active during development, and switching their
noteworthy that p75NTR levels are reduced in phenotype from “transmitting mode” to
neuronal cells during adulthood, and activation “regenerating/regrowth mode” (Richner et al.
of this receptor by NGF has been shown to 2014).
cause neuronal death in motor neurons, in partic- Due to the pleiotropic nature of this molecule,
ular through activation of the JNK and NF-κB however, NGF synthesis also increases in
pathway (Wiese et al. 1999), suggesting a switch non-neuronal cells, such as the Schwann cells
between pro- and anti-neurotrophic NGF signal- and perineural fibroblasts surrounding the stumps
ing via p75NTR in adulthood. In addition, NGF following axon lesion (Matsuoka et al. 1991), and
levels are reduced in adult neurons once nervous acts on axons of the afferent sensory neurons in
system development is complete (Terenghi the proximal stump via TrkA. NGF is retro-
1999). gradely transported to the cell body, where it
5 NGF and Endogenous Regeneration: From Embryology Toward Therapies 57

mediates a trophic effect, enhancing the levels of (OL) wrapping multiple axons, in a process
the pro-survival gene GAP-43 (Melville et al. which reaches its peak at 2–4 week postnatal
1989; Wiese et al. 1992; Verge 1996). NGF also age. During the development of the PNS, SCPs
stimulates Schwann cell migration and recruit- are generated from the neural crest and then
ment in the distal stump of the transected nerve migrate along with growing axons to form the
(Taniuchi et al. 1988). peripheral nerves, where the whole cell body
On the contrary, deafferentation of adult sym- wraps around the nude axon. Notably, the tran-
pathetic neurons does not modify the expression scriptional networks that drive OPC and SCP
level of NGF or TrkA (DeCouto et al. 2003), differentiation, such as central and peripheral
while axotomy of the cervical vagus nerve results myelination and remyelination, are substantially
in increased expression of mRNAs for NGF in different (Sock and Wegner 2019).
non-neuronal cells at both the proximal and distal A role of the neurotrophins in regulating
segments of the transected nerve segment (Lee myelination has been suggested and
et al. 2001). demonstrated by a number of studies, but still
The indirect effects of NGF on axonal remains a matter of investigation. It has, in fact,
regrowth support the pharmacological use of been suggested that NGF acts as a potent regula-
this neurotrophin in non-NGF-dependent tor of the axonal signals controlling the
pathways, including the use of innovative myelination of TrkA-expressing DRG neurons.
approaches based on biomaterials. Taking the Unexpectedly, these NGF-regulated axonal
results obtained from conventional grafts loaded signals have opposite effects on the myelination
with NGF, such as nerve bridges (Derby et al. of the peripheral and central branch of the
1993), or vein grafts (Gravvanis et al. 2004) as a pseudounipolar neurons in DRGs, promoting
starting point, more recent approaches have peripheral myelination by Schwann cells (SCs)
extended the potential use of NGF to treating but reducing central myelination by OLs (Chan
spinal cord injury. For example, the incorporation et al. 2004).
of NGF into an electrospun scaffold (Colello et al. This discrepant effect is still a topic of debate,
2016) or nanoparticles (Xu et al. 2019) has led to since little is known about the role of NGF and
improved axonal regrowth in spinal cord injury, other neurotrophins in the regulation of CNS
possibly regulating the inflammation and angio- myelination, with most of the data deriving from
genesis associated with contusion damage. in vitro models (Xiao et al. 2009). Whereas NGF
is ineffective in promoting myelination in axon-
OL DRG co-cultures, while promoting
5.5 NGF and Remyelination myelination in axon-SC DRG (Chan et al.
2004), the neurotrophin increases the expression
Myelination is the process whereby the axons are of the genes encoding for myelin-related proteins
wrapped in a multilayer lipid-enriched membrane in OL by acting on both p75NTR and TrkA
known as myelin sheath, the thickness of which receptors (Du et al. 2003, 2006). Moreover,
depends upon axon diameter, to guarantee the since NGF is also able to interact directly with
appropriate axonal potential conduction. the myelin oligodendrocyte glycoprotein (MOG),
Myelination occurs during development in both modulating axon growth and survival, this novel
CNS and PNS during late gestation and the early mechanism may mediate communication
post-natal period and is carried out by two distinct between growing axons and myelination (von
cell types, oligodendrocyte precursor cells Büdingen et al. 2015).
(OPCs) and Schwann cells precursors (SCP). In the PNS, converging results indicate that
In the CNS, three different sequential waves NGF regulates both the migration and differenti-
(E12.5, E15.5, E18.5) in mice generate the OPCs, ation of SCPs. The study of Ntrk1 (the gene
which spread throughout all brain and spinal cord encoding for TrkA) expression in the sciatic
regions, with a single mature oligodendrocyte nerve during development shows its high
58 V. A. Baldassarro et al.

expression at E17, before decreasing at E19 and sclerosis (MS), an increase in tissue NGF concen-
disappearing postnatally. p75NTR also increases at tration has been described (Calzà et al. 1997;
E19 and decreases at the postnatal stages (Piirsoo Micera et al. 1998; Acosta et al. 2013), such as
et al. 2010). This dynamic expression is also an increase of p75NTR positive cells in the
involved in SCP/SC differentiation and subventricular zone (SVZ) of the lateral ventricles
myelination, supporting the bi-directional signal- (Calzà et al. 1997). These cells, recognized as
ing occurring between developing axons and SCs. OPCs (Oderfeld-Nowak et al. 2009), migrate to
In vitro data also support the role of NGF in SC the corpus callosum (Calzà et al. 1998), p75NTR
differentiation, the Ngf gene being constantly being a main regulator of the migration process
over-expressed at all stages compared to during remyelination also (Anton et al. 1994;
differentiated cells. Ntrk1 expression is also Bentley and Lee 2000). In addition, OLs
higher than during proliferation and the growth- expressing p75NTR have also been found in MS
arrest state, and p75NTR expression is induced by patients (Chang et al. 2000). The importance of
differentiation (Bentley and Lee 2000). the NGF low-affinity receptor in OPC biology is
In both the CNS and the PNS, numerous cell also confirmed by the exacerbation of symptoms
characteristics and signaling molecules, deriving in MS animal models lacking p75NTR (Copray
from the demyelination process, or from glial and et al. 2004), in which an altered composition of
neuronal cell signals, orchestrate the complex the inflammatory cellular infiltrates is described
machinery of myelin repair in adulthood. In the (Küst et al. 2006). On the other hand, a protective
CNS, a significant number of the OPCs generated effect on OLs has been shown by a synthetic
during development do not differentiate but ligand for TrkA in the cuprizone demyelination
remain quiescent, producing the pool of adult model (Bonetto et al. 2017).
precursors identified by the expression of specific The general view of the possible role of NGF
markers (e.g., NG2 and PDGFRα). Resident and in OPC/OL cell fate, as derived from in vitro
newly formed OPCs, derived from neural stem experiments, has been proposed by Casaccia-
cells (NSCs), are activated by demyelinating Bonefill and Chao (Casaccia-Bonnefil et al.
insults, and by signals produced by demyelinated 2000). In brief, the binding of neurotrophins to
axons, resident astrocytes, and microglia, and Trk receptor tyrosine kinases initiates signaling
may efficiently repair the myelin, thus restoring cascades that promote cell survival and differen-
saltatory conduction. Indeed, remyelination offers tiation. In contrast, p75NTR modulates the suscep-
a unique example of endogenous regeneration in tibility to death of fully differentiated, but not
a system with poor regeneration capabilities, such OPCs, in specific conditions. NGF has no toxic
as the CNS (Crawford et al. 2013). This effects on trkA or p75NTR pig-derived OPCs
remyelination process shares some aspects and (Althaus and Klöppner 2006), such as OL
mechanisms with developmental myelination, as isolated from the rat cerebrum and expressing
supported by the recapitulation hypothesis (Fancy p75NTR (Starkey et al. 2001); in fact NGF
et al. 2011). However, it is now clear that devel- overexpression in human NSCs increases OL dif-
opmental myelination and remyelination present ferentiation (Marei et al. 2013). Lastly, NGF
important differences (Miron et al. 2011; exposure protects OLs from TNFa-induced toxic-
Baldassarro et al. 2019a, b); there are also impor- ity, not only in resident precursors, but also in
tant regional differences in the remyelination OPCs generated in the SVZ (Takano et al. 2000;
capabilities of the adult CNS (Horiuchi et al. Petratos et al. 2004). Overall, the final picture of
2017; Kuhn et al. 2019). A large body of data p75NTR and Trks expression throughout OPC dif-
on the possible role of NGF in myelin protection ferentiation is still disputed, depending on the
and/or repair in the CNS comes from animal animal species, cell system, and animal model
models of inflammatory/demyelinating diseases. used for in vitro and in vivo experiments (Althaus
In experimental allergic encephalomyelitis, the et al. 2008; Guo et al. 2013).
most widely used animal model for multiple
5 NGF and Endogenous Regeneration: From Embryology Toward Therapies 59

SCs, like OLs, are postmitotic and fully Anton ES, Weskamp G, Reichardt LF, Matthew WD
differentiated cells. Peripheral demyelination, (1994) Nerve growth factor and its low-affinity recep-
tor promote Schwann cell migration. Proc Natl Acad
however, leads to a rapid response of these cells, Sci U S A 91:2795–2799. https://doi.org/10.1073/
which de-differentiate, re-enter the cell cycle, and pnas.91.7.2795
activate the repair process (Jessen et al. 2015). In Bagnara GP (2020) Stem cells. Societa Editrice Esculapi
addition, SC lineage is characterized by Baldassarro VA, Krężel W, Fernández M, Schuhbaur B,
Giardino L, Calzà L (2019a) The role of nuclear
differences throughout the different phases of receptors in the differentiation of oligodendrocyte pre-
development and differentiation, but heterogene- cursor cells derived from fetal and adult neural stem
ity is lower compared to OLs (Jessen et al. 2015). cells. Stem Cell Res 37:101443. https://doi.org/10.
There is much evidence to suggest that NGF 1016/j.scr.2019.101443
Baldassarro VA, Marchesini A, Giardino L, Calzà L
and other neurotrophins play different roles in (2019b) Differential effects of glucose deprivation on
these complex contexts (Althaus et al. 2008; the survival of fetal versus adult neural stem cells-
Webber and Zochodne 2010). p75NTR plays a derived oligodendrocyte precursor cells. Glia. https://
role in the regulation of SCP migration to the doi.org/10.1002/glia.23750
Benfey M, Aguayo AJ (1982) Extensive elongation of
PNS (Anton et al. 1994). Throughout p75NTR axons from rat brain into peripheral nerve grafts.
activation, NGF treatment following peripheral Nature 296:150–152. https://doi.org/10.1038/
nerve lesions promotes SC autophagy, enhancing 296150a0
the clearance of myelin debris and accelerating Bentley CA, Lee KF (2000) p75 Is important for axon
growth and Schwann cell migration during develop-
recovery from injury (Li et al. 2020). NGF also ment. J Neurosci 20:7706–7715. https://doi.org/10.
acts as chemo-attractant and chemo-kinetic cue 1523/jneurosci.20-20-07706.2000
for both SCs and SCPs (Maniwa et al. 2003; Bonetto G, Charalampopoulos I, Gravanis A, Karagogeos
Cornejo et al. 2010). D (2017) The novel synthetic microneurotrophin
BNN27 protects mature oligodendrocytes against
The data briefly summarized above show a cuprizone-induced death, through the NGF receptor
clear portrait of the pro-myelinating role of NGF TrkA. Glia 65:1376–1394. https://doi.org/10.1002/
in PNS, while still uncertain is the effect on glia.23170
OPCs/OLs, and, therefore, on central Botchkarev VA, Botchkareva N V., Peters EMJ, Paus R
(2004) Epithelial growth control by neurotrophins:
myelination. In both districts, moreover, it is leads and lessons from the hair follicle. In: Progress
still unclear whether NGF acts directly on in brain research. Elsevier, Amsterdam, pp 493–513
myelinating cells, or indirectly through axons or Bracci-Laudiero L, De Stefano ME (2016) NGF in early
the satellite cells (fibroblasts, inflammatory cells, embryogenesis, differentiation, and pathology in the
nervous and immune systems. In: Current topics in
glial cells other than myelinating, etc.). behavioral neurosciences. Springer, Cham, pp
125–152
Bruce CC, Zhao C, Franklin RJM (2010) Remyelination—
An effective means of neuroprotection. Horm Behav
References 57:56–62. https://doi.org/10.1016/j.yhbeh.2009.06.
004
Acosta CMR, Cortes C, MacPhee H, Namaka MP (2013) Cafferty WBJ, Duffy P, Huebner E, Strittmatter SM
Exploring the role of nerve growth factor in multiple (2010) MAG and OMgp synergize with Nogo-A to
sclerosis: implications in myelin repair. CNS Neurol restrict axonal growth and neurological recovery after
Disord Drug Targets 12:1242–1256 spinal cord trauma. J Neurosci 30:6825–6837. https://
Aloe L, Calzà L (2004) NGF and related molecules in doi.org/10.1523/JNEUROSCI.6239-09.2010
health and disease: preface. In: Progress in brain Calzà L, Giardino L, Pozza M, Micera A, Aloe L (1997)
research, p 544 Time-course changes of nerve growth factor,
Althaus HH, Klöppner S (2006) Mature pig oligoden- corticotropin-releasing hormone, and nitric oxide
drocytes rapidly process human recombinant synthase isoforms and their possible role in the devel-
pro-nerve growth factor and do not undergo cell opment of inflammatory response in experimental
death. J Neurochem 98:506–517. https://doi.org/10. allergic encephalomyelitis. Proc Natl Acad Sci U S A
1111/j.1471-4159.2006.03891.x 94:3368–3373. https://doi.org/10.1073/pnas.94.7.3368
Althaus HH, Klöppner S, Klopfleisch S, Schmitz M (2008) Calzà L, Giardino L, Pozza M, Bettelli C, Micera A, Aloe
Oligodendroglial cells and neurotrophins: a poly- L (1998) Proliferation and phenotype regulation in the
phonic cantata in major and minor. J Mol Neurosci subventricular zone during experimental allergic
35:65–79. https://doi.org/10.1007/s12031-008-9053-y
60 V. A. Baldassarro et al.

encephalomyelitis: in vivo evidence of a role for nerve 119:73–82. https://doi.org/10.1016/j.molbrainres.


growth factor. Proc Natl Acad Sci U S A 2003.08.015
95:3209–3214. https://doi.org/10.1073/pnas.95.6.3209 Derby A, Engleman VW, Frierdich GE, Neises G, Rapp
Calzà L, Giardino L, Giuliani A, Aloe L, Levi-Montalcini SR, Roufa DG (1993) Nerve growth factor facilitates
R (2001) Nerve growth factor control of neuronal regeneration across nerve gaps: morphological and
expression of angiogenetic and vasoactive factors. behavioral studies in rat sciatic nerve. Exp Neurol
Proc Natl Acad Sci U S A 98:4160–4165. https://doi. 119:176–191. https://doi.org/10.1006/exnr.1993.1019
org/10.1073/pnas.051626998 Du Y, Fischer TZ, Lee LN, Lercher LD, Dreyfus CF
Campenot RB (1994) NGF and the local control of nerve (2003) Regionally specific effects of BDNF on
terminal growth. J Neurobiol 25:599–611. https://doi. oligodendrocytes. Dev Neurosci 25:116–126. https://
org/10.1002/neu.480250603 doi.org/10.1159/000072261
Casaccia-Bonnefil P, Gu C, Chao MV (2000) Du Y, Fischer TZ, Clinton-Luke P, Lercher LD, Dreyfus
Neurotrophins in cell survival/death decisions. Adv CF (2006) Distinct effects of p75 in mediating actions
Exp Med Biol 468:275–282. https://doi.org/10.1007/ of neurotrophins on basal forebrain oligodendrocytes.
978-1-4615-4685-6_22 Mol Cell Neurosci 31:366–375. https://doi.org/10.
Chan JR, Watkins TA, Cosgaya JM, Zhang C, Chen L, 1016/j.mcn.2005.11.001
Reichardt LF, Shooter EM, Barres BA (2004) NGF Ebenezer G, Polydefkis M (2014) Epidermal innervation
controls axonal receptivity to myelination by Schwann in diabetes. In: Handbook of clinical neurology.
cells or oligodendrocytes. Neuron 43:183–191. https:// Elsevier, pp 261–274
doi.org/10.1016/j.neuron.2004.06.024 Edward Hickman F, Stanley EM, Carter BD (2018)
Chang A, Nishiyama A, Peterson J, Prineas J, Trapp BD Neurotrophin responsiveness of sympathetic neurons
(2000) NG2-positive oligodendrocyte progenitor cells is regulated by rapid mobilization of the p75 receptor
in adult human brain and multiple sclerosis lesions. J to the cell surface through trka activation of arf6. J
Neurosci 20:6404–6412. https://doi.org/10.1523/ Neurosci 38:5606–5619. https://doi.org/10.1523/
jneurosci.20-17-06404.2000 JNEUROSCI.0788-16.2018
Colello RJ, Chow WN, Bigbee JW, Lin C, Dalton D, Ehrhard PB, Erb P, Graumann U, Otten U (1993) Expression
Brown D, Jha BS, Mathern BE, Lee KD, Simpson of nerve growth factor and nerve growth factor receptor
DG (2016) The incorporation of growth factor and tyrosine kinase Trk in activated CD4-positive T-cell
chondroitinase ABC into an electrospun scaffold to clones. Proc Natl Acad Sci U S A 90:10984–10988.
promote axon regrowth following spinal cord injury. https://doi.org/10.1073/pnas.90.23.10984
J Tissue Eng Regen Med 10:656–668. https://doi.org/ Elsherbiny NM, Abdel-Mottaleb Y, Elkazaz AY, Atef H,
10.1002/term.1805 Lashine RM, Youssef AM, Ezzat W, El-Ghaiesh SH,
Copray S, Küst B, Emmer B, Lin MY, Liem R, Amor S, Elshaer RE, El-Shafey M, Zaitone SA (2019) Carba-
De Vries H, Floris S, Boddeke E (2004) Deficient p75 mazepine alleviates retinal and optic nerve neural
low-affinity neurotrophin receptor expression degeneration in diabetic mice via nerve growth
exacerbates experimental allergic encephalomyelitis factor-induced PI3K/Akt/mTOR activation. Front
in C57/BL6 mice. J Neuroimmunol 148:41–53. Neurosci 13. https://doi.org/10.3389/fnins.2019.01089
https://doi.org/10.1016/j.jneuroim.2003.11.008 Fancy SPJ, Chan JR, Baranzini SE, Franklin RJM,
Cornejo M, Nambi D, Walheim C, Somerville M, Rowitch DH (2011) Myelin regeneration: a recapitula-
Walker J, Kim L, Ollison L, Diamante G, tion of development? Annu Rev Neurosci 34:21–43.
Vyawahare S, De Bellard ME (2010) Effect of https://doi.org/10.1146/annurev-neuro-061010-
NRG1, GDNF, EGF and NGF in the migration of a 113629
Schwann cell precursor line. Neurochem Res Fernández M, Pirondi S, Chen BL, Del Vecchio G,
35:1643–1651. https://doi.org/10.1007/s11064-010- Alessandri M, Farnedi A, Pession A, Feki A, Jaconi
0225-0 MEE, Calzà L (2011) Isolation of rat embryonic stem-
Crawford AH, Chambers C, Franklin RJM (2013) like cells: a tool for stem cell research and drug discov-
Remyelination: the true regeneration of the central ery. Dev Dyn 240:2482–2494. https://doi.org/10.1002/
nervous system. J Comp Pathol 149:242–254. https:// dvdy.22761
doi.org/10.1016/j.jcpa.2013.05.004 Generini S, Tuveri MA, Matucci Cerinic M, Mastinu F,
David S, Aguayo A (1981) Axonal elongation into periph- Manni L, Aloe L (2004) Topical application of nerve
eral nervous system “bridges” after central nervous growth factor in human diabetic foot ulcers. A study of
system injury in adult rats. Science 214:931–933. three cases. Exp Clin Endocrinol Diabetes
https://doi.org/10.1126/science.6171034 112:542–544. https://doi.org/10.1055/s-2004-821313
DeCouto SA, Jones EE, Kudwa AE, Shoemaker SE, Gostynska N, Pannella M, Rocco ML, Giardino L, Aloe L,
Shafer AJ, Brieschke MA, James PF, Vaughn JC, Calzà L (2019) The pleiotropic molecule NGF
Isaacson LG (2003) The effects of deafferentation regulates the in vitro properties of fibroblasts,
and exogenous NGF on neurotrophins and keratinocytes and endothelial cells: implications for
neurotrophin receptor mRNA expression in the adult wound healing. Am J Physiol Physiol. https://doi.org/
superior cervical ganglion. Brain Res Mol Brain Res 10.1152/ajpcell.00180.2019
5 NGF and Endogenous Regeneration: From Embryology Toward Therapies 61

Gravvanis AI, Tsoutsos DA, Tagaris GA, Papalois AE, expression does alter the composition of cellular infil-
Patralexis CG, Iconomou TG, Panayotou PN, trate in experimental autoimmune encephalomyelitis in
Ioannovich JD (2004) Beneficial effect of nerve C57BL/6 mice. J Neuroimmunol 174:92–100. https://
growth factor-7S on peripheral nerve regeneration doi.org/10.1016/j.jneuroim.2006.01.020
through inside-out vein grafts: an experimental study. Laverdet B, Danigo A, Girard D, Magy L, Demiot C,
Microsurgery 24:408–415. https://doi.org/10.1002/ Desmoulière A (2015) Skin innervation: important
micr.20055 roles during normal and pathological cutaneous repair.
Guo J et al (2013) proNGF inhibits proliferation and Histol Histopathol 30:875–892
oligodendrogenesis of postnatal hippocampal neural Lee P, Zhuo H, Helke CJ (2001) Axotomy alters
stem/progenitor cells through p75NTR in vitro. Stem neurotrophin and neurotrophin receptor mRNAs in
Cell Res 11:874–887. https://doi.org/10.1016/j.scr. the vagus nerve and nodose ganglion of the rat. Brain
2013.05.004 Res Mol Brain Res 87:31–41. https://doi.org/10.1016/
Guthrie S (2007) Neurotrophic factors: are they axon s0169-328x(00)00277-1
guidance molecules? Adv Exp Med Biol 621:81–94 Leon A, Buriani A, Dal Toso R, Fabris M, Romanello S,
Horiuchi M, Suzuki-Horiuchi Y, Akiyama T, Itoh A, Aloe L, Levi-Montalcini R (1994) Mast cells synthe-
Pleasure D, Carstens E, Itoh T (2017) Differing intrin- size, store, and release nerve growth factor. Proc Natl
sic biological properties between forebrain and spinal Acad Sci U S A 91:3739–3743. https://doi.org/10.
oligodendroglial lineage cells. J Neurochem 1073/pnas.91.9.3739
142:378–391. https://doi.org/10.1111/jnc.14074 Levi-Montalcini R (1997) The saga of the nerve growth
Inanç B, Elçin AE, Elçin YM (2008) Human embryonic factor. World Scientific, River Edge, NJ
stem cell differentiation on tissue engineering Levi-Montalcini R, Hamburger V (1951) Selective growth
scaffolds: effects of NGF and retinoic acid induction. stimulating effects of mouse sarcoma on the sensory
Tissue Eng Part A. https://doi.org/10.1089/tea.2007. and sympathetic nervous system of the chick embryo. J
0213 Exp Zool 116:321–361. https://doi.org/10.1002/jez.
Indo Y (2012) Nerve growth factor and the physiology of 1401160206
pain: lessons from congenital insensitivity to pain with Li R, Li D, Wu C, Ye L, Wu Y, Yuan Y, Yang S, Xie L,
anhidrosis. Clin Genet 82:341–350 Mao Y, Jiang T, Li Y, Wang J, Zhang H, Li X, Xiao J
Jere SW, Houreld NN, Abrahamse H (2019) Role of the (2020) Nerve growth factor activates autophagy in
PI3K/AKT (mTOR and GSK3β) signalling pathway Schwann cells to enhance myelin debris clearance
and photobiomodulation in diabetic wound healing. and to expedite nerve regeneration. Theranostics
Cytokine Growth Factor Rev. 50:52–59 10:1649–1677. https://doi.org/10.7150/thno.40919
Jessen KR, Mirsky R, Lloyd AC (2015) Schwann cells: Lumelsky N, O’Hayre M, Chander P, Shum L, Somerman
development and role in nerve repair. Cold Spring MJ (2018) Autotherapies: enhancing endogenous
Harb Perspect Biol 7:1–15. https://doi.org/10.1101/ healing and regeneration. Trends Mol Med
cshperspect.a020487 24:919–930
Johnson EM, Gorin PD, Brandeis LD, Pearson J (1980) Maniwa S, Iwata A, Hirata H, Ochi M (2003) Effects of
Dorsal root ganglion neurons are destroyed by expo- neurotrophic factors on chemokinesis of Schwann cells
sure in utero to maternal antibody to nerve growth in culture. Scand J Plast Reconstr Surg Hand Surg
factor. Science 210:916–918. https://doi.org/10.1126/ 37:14–17. https://doi.org/10.1080/alp.37.1.14.17
science.7192014 Marei HES, Althani A, Afifi N, Abd-Elmaksoud A,
Kasemeier-Kulesa JC, Kulesa PM (2018) The convergent Bernardini C, Michetti F, Barba M, Pescatori M,
roles of CD271/p75 in neural crest-derived melanoma Maira G, Paldino E, Manni L, Casalbore P, Cenciarelli
plasticity. Dev Biol 444:S352–S355. https://doi.org/ C (2013) Over-expression of hNGF in adult human
10.1016/j.ydbio.2018.04.008 olfactory bulb neural stem cells promotes cell growth
Kim YS, Jo DH, Lee H, Kim JH, Kim KW, Kim JH (2013) and oligodendrocytic differentiation. PLoS One 8:
Nerve growth factor-mediated vascular endothelial e82206. https://doi.org/10.1371/journal.pone.0082206
growth factor expression of astrocyte in retinal vascu- Martin-Zanca D, Barbacid M, Parada LF (1990) Expres-
lar development. Biochem Biophys Res Commun sion of the trk proto-oncogene is restricted to the sen-
431:740–745. https://doi.org/10.1016/j.bbrc.2013.01. sory cranial and spinal ganglia of neural crest origin in
045 mouse development. Genes Dev 4:683–694. https://
Kristiansen M, Ham J (2014) Programmed cell death dur- doi.org/10.1101/gad.4.5.683
ing neuronal development: the sympathetic neuron Matsuda H, Kannan Y, Ushio H, Kiso Y, Kanemoto T,
model. Cell Death Differ 21:1025–1035 Suzuki H, Kitamura Y (1991) Nerve growth factor
Kuhn S, Gritti L, Crooks D, Dombrowski Y (2019) induces development of connective tissue-type mast
Oligodendrocytes in development, myelin generation cells in vitro from murine bone marrow cells. J Exp
and beyond. Cells 8:1424. https://doi.org/10.3390/ Med 174:7–14. https://doi.org/10.1084/jem.174.1.7
cells8111424 Matsuoka I, Meyer M, Thoenen H (1991) Cell-type-spe-
Küst B, Mantingh-Otter I, Boddeke E, Copray S (2006) cific regulation of nerve growth factor (NGF) synthesis
Deficient p75 low-affinity neurotrophin receptor in non-neuronal cells: comparison of Schwann cells
62 V. A. Baldassarro et al.

with other cell types. J Neurosci 11:3165–3177. https:// Park MJ, Kwak HJ, Lee HC, Yoo DH, Park IC, Kim MS,
doi.org/10.1523/jneurosci.11-10-03165.1991 Lee SH, Chang HR, Hong S Il (2007) Nerve growth
Meberg PJ, Ono S, Minamide LS, Takahashi M, Bamburg factor induces endothelial cell invasion and cord for-
JR (1998) Actin depolymerizing factor and cofilin mation by promoting matrix metalloproteinase-2-
phosphorylation dynamics: response to signals that expression through the phosphatidylinositol 3-kinase/
regulate neurite extension. Cell Motil Cytoskeleton Akt signaling pathway and AP-2 transcription factor. J
39:172–190. https://doi.org/10.1002/(SICI)1097-0169 Biol Chem 282:30485–30496. https://doi.org/10.1074/
(1998)39:2<172::AID-CM8>3.0.CO;2-8 jbc.M701081200
Melville S, Sherburn TE, Coggeshall RE (1989) Preserva- Petratos S, Gonzales MF, Azari MF, Marriott M,
tion of sensory cells by placing stumps of transected Minichiello RA, Shipham KA, Profyris C,
nerve in an impermeable tube. Exp Neurol Nicolaou A, Boyle K, Cheema SS, Kilpatrick TJ
105:311–315. https://doi.org/10.1016/0014-4886(89) (2004) Expression of the low-affinity neurotrophin
90135-0 receptor, p75NTR, is upregulated by oligodendroglial
Mesulam MM, Lalehzari N, Rahmani F, Ohm D, progenitors adjacent to the subventricular zone in
Shahidehpour R, Kim G, Gefen T, Weintraub S, response to demyelination. Glia 48:64–75. https://doi.
Bigio E, Geula C (2019) Cortical cholinergic denerva- org/10.1002/glia.20056
tion in primary progressive aphasia with Alzheimer Piirsoo M, Kaljas A, Tamm K, Timmusk T (2010) Expres-
pathology. Neurology 92:E1580–E1588. https://doi. sion of NGF and GDNF family members and their
org/10.1212/WNL.0000000000007247 receptors during peripheral nerve development and
Micera A, Vigneti E, Aloe L (1998) Changes of NGF differentiation of Schwann cells in vitro. Neurosci
presence in nonneuronal cells in response to experi- Lett 469:135–140. https://doi.org/10.1016/j.neulet.
mental allergic encephalomyelitis in Lewis rats. Exp 2009.11.060
Neurol 154:41–46. https://doi.org/10.1006/exnr.1998. Pyle AD, Lock LF, Donovan PJ (2006) Neurotrophins
6864 mediate human embryonic stem cell survival. Nat
Mietto BS, Mostacada K, Martinez AMB (2015) Biotechnol 24:344–350. https://doi.org/10.1038/
Neurotrauma and inflammation: CNS and PNS nbt1189
responses. Mediators Inflamm. https://doi.org/10. Reichardt LF (2006) Neurotrophin-regulated signalling
1155/2015/251204 pathways. Philos Trans R Soc B Biol Sci
Miron VE, Kuhlmann T, Antel JP (2011) Cells of the 361:1545–1564
oligodendroglial lineage, myelination, and Reinke JM, Sorg H (2012) Wound repair and regeneration.
remyelination. BBA Mol Basis Dis 1812:184–193. Eur Surg Res 49:35–43
https://doi.org/10.1016/j.bbadis.2010.09.010 Richardson PM, Riopelle RJ (1984) Uptake of nerve
Mitsiadis TA, Pagella P (2016) Expression of nerve growth factor along peripheral and spinal axons of
growth factor (NGF), TrkA, and p75NTR in develop- primary sensory neurons. J Neurosci 4:1683–1689.
ing human fetal teeth. Front Physiol 7. https://doi.org/ https://doi.org/10.1523/jneurosci.04-07-01683.1984
10.3389/fphys.2016.00338 Richardson PM, McGuinness UM, Aguayo AJ (1980)
Moscatelli I, Pierantozzi E, Camaioni A, Siracusa G, Axons from CNS neurones regenerate into PNS grafts.
Campagnolo L (2009) p75 neurotrophin receptor is Nature 284:264–265. https://doi.org/10.1038/
involved in proliferation of undifferentiated mouse 284264a0
embryonic stem cells. Exp Cell Res 315:3220–3232. Richner M, Ulrichsen M, Elmegaard SL, Dieu R, Pallesen
https://doi.org/10.1016/j.yexcr.2009.08.014 LT, Vaegter CB (2014) Peripheral nerve injury
Obreja O, Rukwied R, Nagler L, Schmidt M, Schmelz M, modulates neurotrophin signaling in the peripheral and
Namer B (2018) Nerve growth factor locally sensitizes central nervous system. Mol Neurobiol 50:945–970
nociceptors in human skin. Pain 159:416–426. https:// Sacchetti M, Lambiase A, Schmidl D, Schmetterer L,
doi.org/10.1097/j.pain.0000000000001108 Ferrari M, Mantelli F, Allegretti M, Garhoefer G
Oderfeld-Nowak B, Zaremba M, Kwiatkowska-Patzer B, (2020) Effect of recombinant human nerve growth
Lipkowski AW, Kurkowska-Jastrzȩbska I, Triaca V, factor eye drops in patients with dry eye: a phase IIa,
Aloe L (2009) NG2 positive cells of rat spinal cord open label, multiple-dose study. Br J Ophthalmol
activated during experimental autoimmune encephalo- 104:127–135. https://doi.org/10.1136/bjophthalmol-
myelitis are spatially associated with radially oriented 2018-312470
astroglia and express p75 receptor: a role for nerve Sen CK, Gordillo GM, Roy S, Kirsner R, Lambert L, Hunt
growth factor in oligodendrocyte progenitor migra- TK, Gottrup F, Gurtner GC, Longaker MT (2009)
tion? Arch Ital Biol 147:105–115. https://doi.org/10. Human skin wounds: a major and snowballing threat
4449/aib.v147i4.871 to public health and the economy: perspective article.
Pannella M, Giardino L, Calzà L, Fernández M (2018) Wound Repair Regen 17:763–771
Growth and neurotrophic factors in embryonic stem Sima AAF (2003) New insights into the metabolic and
cells. In: Methods in molecular biology. Humana, molecular basis for diabetic neuropathy. Cell Mol Life
Totowa, NJ, pp 275–294 Sci 60:2445–2464
5 NGF and Endogenous Regeneration: From Embryology Toward Therapies 63

Snider WD, McMahon SB (1998) Tackling pain at the nerve growth factor to modulate central axon circuitry.
source: new ideas about nociceptors. Neuron J Cell Biol 210:891–898. https://doi.org/10.1083/jcb.
20:629–632 201504106
Sock E, Wegner M (2019) Transcriptional control of Webber C, Zochodne D (2010) The nerve regenerative
myelination and remyelination. Glia 67 microenvironment: early behavior and partnership of
(11):2153–2165 axons and Schwann cells. Exp Neurol 223:51–59
Starkey GD, Petratos S, Shipham KA, Butzkueven H, Wells JM, Watt FM (2018) Diverse mechanisms for
Bucci T, Lowry K, Cheema SS, Kilpatrick TJ (2001) endogenous regeneration and repair in mammalian
Neurotrophin receptor expression and responsiveness organs. Nature 557:322–328
by postnatal cerebral oligodendroglia. Neuroreport Wiese UH, Ruth JL, Emson PC (1992) Differential expres-
12:4081–4086. https://doi.org/10.1097/00001756- sion of growth-associated protein (GAP-43) mRNA in
200112210-00044 rat primary sensory neurons after peripheral nerve
Sun D (2016) Endogenous neurogenic cell response in the lesion: a non-radioactive in situ hybridisation study.
mature mammalian brain following traumatic injury. Brain Res 592:141–156. https://doi.org/10.1016/0006-
Exp Neurol 275:405–410 8993(92)91669-6
Takano R, Hisahara S, Namikawa K, Kiyama H, Okano H, Wiese S, Metzger F, Holtmann B, Sendtner M (1999) The
Miura M (2000) Nerve growth factor protects role of p75NTR in modulating neurotrophin survival
oligodendrocytes from tumor necrosis factor-alpha- effects in developing motoneurons. Eur J Neurosci
induced injury through Akt-mediated signaling 11:1668–1676. https://doi.org/10.1046/j.1460-9568.
mechanisms. J Biol Chem 275:16360–16365. https:// 1999.00585.x
doi.org/10.1074/jbc.M910419199 Wislet S, Vandervelden G, Rogister B (2018) From neural
Taniuchi M, Clark HB, Schweitzer JB, Johnson EM crest development to cancer and vice versa: how
(1988) Expression of nerve growth factor receptors p75NTR and (Pro)neurotrophins could act on cell
by Schwann cells of axotomized peripheral nerves: migration and invasion? Front Mol Neurosci 11:244
ultrastructural location, suppression by axonal contact, Xiao J, Kilpatrick TJ, Murray SS (2009) The role of
and binding properties. J Neurosci 8:664–681 neurotrophins in the regulation of myelin development.
Terenghi G (1999) Peripheral nerve regeneration and Neurosignals 17:265–276. https://doi.org/10.1159/
neurotrophic factors. J Anat 194:1–14. https://doi.org/ 000231893
10.1046/j.1469-7580.1999.19410001.x Xu D, Wu D, Qin M, Nih LR, Liu C, Cao Z, Ren J,
Tron VA, Coughlin MD, Jang DE, Stanisz J, Sauder DN Chen X, He Z, Yu W, Guan J, Duan S, Liu F, Liu X,
(1990) Expression and modulation of nerve growth Li J, Harley D, Xu B, Hou L, Chen ISY, Wen J,
factor in murine keratinocytes (PAM 212). J Clin Chen W, Pourtaheri S, Lu Y (2019) Efficient delivery
Invest 85:1085–1089. https://doi.org/10.1172/ of nerve growth factors to the central nervous system
JCI114539 for neural regeneration. Adv Mater 31:e1900727.
Tuveri M, Generini S, Matucci-Cerinic M, Aloe L (2000) https://doi.org/10.1002/adma.201900727
NGF, a useful tool in the treatment of chronic Yamashita T, Tucker KL, Barde YA (1999) Neurotrophin
vasculitic ulcers in rheumatoid arthritis. Lancet binding to the p75 receptor modulates Rho activity and
356:1739–1740. https://doi.org/10.1016/S0140-6736 axonal outgrowth. Neuron 24:585–593. https://doi.org/
(00)03212-8 10.1016/S0896-6273(00)81114-9
Verge VMK (1996) Neurotrophins and nerve injury in the Yu X, Qi Y, Zhao T, Fang J, Liu X, Xu T, Yang Q, Dai X
adult. Philos Trans R Soc B Biol Sci 351:423–430. (2019) NGF increases FGF2 expression and promotes
https://doi.org/10.1098/rstb.1996.0038 endothelial cell migration and tube formation through
von Büdingen H-C, Mei F, Greenfield A, Jahn S, Shen PI3K/Akt and ERK/MAPK pathways in human
Y-AA, Reid HH, McKemy DD, Chan JR (2015) The chondrocytes. Osteoarthr Cartil 27:526–534. https://
myelin oligodendrocyte glycoprotein directly binds doi.org/10.1016/j.joca.2018.12.007
The Versatile Roles of Nerve Growth
Factor in Neuronal Attraction, 6
Odontoblast Differentiation,
and Mineral Deposition in Human Teeth

Thimios A. Mitsiadis and Pierfrancesco Pagella

Abstract the NGF secreting cells. These results show


Nerve growth factor (NGF) is an important that NGF is closely linked to the various
molecule for the development and differentia- functions of odontoblasts, including secretory
tion of neuronal and non-neuronal cells. Here and neuronal attraction processes.
we analyze by immunohistochemistry the dis-
tribution of NGF in the dental pulp mesen-
chyme of embryonic and functional human
teeth. In the dental pulp of both embryonic 6.1 Introduction
and healthy functional teeth, NGF is mainly
expressed in the odontoblasts that are respon- Nerve growth factor (NGF) is the first identified
sible for dentine formation, while in functional member of a family of neurotrophic molecules
teeth NGF is also expressed in nerve fibers called neurortophins, which have essential roles
innervating the dental pulp. In injured teeth, in the fate, development, survival, outgrowth, and
NGF is expressed in the newly formed maintenance of selected group of neurons
odontoblastic-like cells, which replace the (Tomellini et al. 2014; Reichardt 2006; Levi-
dying odontoblasts. In these teeth, NGF Montalcini 1987; Lewin and Carter 2014). NGF
expression is also upregulated in the intact binds with low affinity to the transmembrane
odontoblasts, suggesting a role for this mole- glycoprotein receptor p75NTR that is expressed
cule in dental tissue repair. Similarly, in on both neurons and non-neuronal target cells,
cultures of human dental pulp cells, NGF and its activation can induce either cell survival
expression is strongly upregulated during or apoptosis (Lee et al. 2001; Ibanez and Simi
their differentiation into odontoblasts as well 2012; Lewin and Carter 2014). The 140 kDa
as during the mineralization process. In tyrosine kinase TrkA glycoprotein acts as the
microfluidic devices, release of NGF from high-affinity receptor for NGF (Tomellini et al.
cultured human dental pulp cells induced neu- 2014; Reichardt 2006; Levi-Montalcini 1987;
ronal growth from trigeminal ganglia toward Lewin and Carter 2014), and its function is
influenced by the concomitant presence of
p75NTR (Reichardt 2006; Lewin and Carter
T. A. Mitsiadis (*) · P. Pagella
2014; Benedetti et al. 1993; Bibel et al. 1999).
Orofacial Development and Regeneration, Institute of Oral
Biology, Centre for Dental Medicine, University of In teeth, apart from the known functions of
Zurich, Zurich, Switzerland NGF in the nervous system, additional roles
e-mail: thimios.mitsiadis@zzm.uzh.ch; pierfrancesco. have been suggested for this molecule during
pagella@zzm.uzh.ch

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 65


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_6
66 T. A. Mitsiadis and P. Pagella

odontogenesis, dental tissue healing upon injury, (Mitsiadis et al. 1992, 1993) and the mouse anti-
and differentiation of ameloblasts and human p75NTR monoclonal antibody 20.4 (Grob
odontoblasts (Mitsiadis et al. 1992, 1993, 2017; et al. 1985; Ross et al. 1984; Chesa et al. 1988)
Mitsiadis and Luukko 1995; Mitsiadis and have already been described. The p75NTR mono-
Pagella 2016). Indeed, our previous studies and clonal antibody was a kind gift of Dr
work from others have shown that NGF and the E.M. Johnson Jr. and Dr C. Osborn (St. Louis,
p75NTR and TrkA receptors are expressed in the USA). Mouse anti-neurofilament (NFP, Zymed
developing human teeth during embryogenesis, labs, San Francisco, CA, USA), rabbit
thus indicating that NGF regulates dental physio- anti-β-III-tubulin (ab18207, Abcam, Cambridge,
logical processes (Mitsiadis et al. 1992, 1993; UK), mouse anti-NuMA (GT3611; GeneTex,
Byers et al. 1990; Byers 1990; Luukko et al. Irvine, CA, U.S.A.), alexa-488-conjugated anti-
1997; Christensen et al. 1993; Nosrat et al. rabbit IgG (A-32731), and alexa-568-conjugated
2002; Mitsiadis and Pagella 2016). Moreover, anti-mouse IgG (A-11004; Thermo Fisher, Basel,
our recent findings have shown that NGF is Switzerland) antibodies were also used.
involved in dental tissue regeneration and the
behavior of dental pulp cells (Mitsiadis et al. Chemicals Vector Vectastain ABC kit was pur-
2017; Pagella et al. 2020b). Indeed, NGF pro- chased from Biosys (Compiègne, France). For the
duced by dental pulp cells cultured in preparation of the culture media, all materials
microfluidic devices acts as a neuroattractive were purchased from Gibco BRL (Life
agent that allows neurons to establish direct Technologies Inc., NY, USA). Other chemicals
contacts with these cells (Pagella et al. 2020b). were obtained from Sigma (St. Louis, MO, USA).
Therefore, these results, in combination with pre- For cultures, Minimum Essential Medium
viously reported findings in dental pulp (MEM) was supplemented with 10% fetal bovine
fibroblasts (Nosrat et al. 2001), indicate that serum, 2 mM glutamine, 100 UI/mL penicillin,
NGF plays a key role in the attraction and ramifi- 100 μg/mL streptomycin (Biowhittaker, Gagny,
cation of neurons within the dental pulp during France), and 0.25 μg/mL amphotericin B
tooth development and regeneration. Previous (Fungizone®). Trigeminal ganglia were cultured
studies in rodents, dogs, and humans have in Neurobasal Medium (Gibco) supplemented
shown that expression of NGF and p75NTR with B27 (17504-044, Gibco), GlutaMAX, peni-
increased after dental injury (Byers et al. 1990; cillin/streptomycin (10 U/mL), 50 ng/mL Nerve
Byers 1990; Mitsiadis et al. 2017; Asaumi et al. Growth Factor (NGF, R&D Systems), and
2000; Byun et al. 2008). Taken together, these 0.25 pM Arabinose Cytoside.
findings illustrate the importance of NGF in a
variety of biological processes linked to tooth
development, homeostasis, and repair.
6.2.2 Embryonic Human Tissues
In this study, we analyze the in vivo distribution
of NGF in the odontoblasts of developing, func-
Tissues Human fetal tissues were obtained from
tional healthy and injured human teeth, as well as
legal abortions. The material comprises teeth from
in cultured human dental pulp cells either alone or
three fetuses (23 gestational week). The gestation
in the presence of trigeminal neurons in vitro.
age was estimated from the fetal foot length and
from the last menstruation of the mother. Embryos
were non-infected, and all tissues were both mac-
6.2 Materials and Methods
roscopically and microscopically normal. The
fetuses were immediately fixed in 10% buffered
6.2.1 Materials
formalin for 5 days and then decalcified for
3 weeks in formic acid/10% formalin prior to
Antibodies Preparation, purification, and charac-
embedding in Paraplast. 4 to 6 μm thick sections
terization of the polyclonal anti-NGF antibody
6 The Versatile Roles of Nerve Growth Factor in Neuronal Attraction. . . 67

were used for immunohistochemistry. This study healthy patients as previously described
was carried out in compliance with the French (Woloszyk et al. 2016). The dental pulps were
legislation, after the approval of the Regional gently removed with forceps, minced with
Ethics Committee of Development and Reproduc- scalpels, rinsed with PBS, and finally enzymati-
tion of the U.F.R. of Medicine of Reims-France cally digested for one hour at 37  C in a solution
(INSERM 314 Reims). of collagenase (3 mg/mL; Life Technologies BV,
Zug ZG, Switzerland) and Dispase (4 mg/mL;
Sigma-Aldrich Chemie GmbH, Buchs SG,
Switzerland). Dental pulp cells were cultured in
6.2.3 Adult Human Tissues
100-μm diameter culture dishes (Becton
Dickinson Labware, Lincoln Park, NJ, USA) in
All procedures were performed in accordance
MEM supplemented with 2 mM
with the current guidelines. The Cantonal Ethic
β-glycerophosphate (Sigma Chemical Co., St
Commission of Zurich approved experimental
Louis, USA). The cells were cultured at 37  C
procedures involving the use of human specimens
in a humidified atmosphere of 5% CO2, 95% air,
(reference number 2012-0588). All procedures
and the culture medium changed every other day.
were performed after obtaining the informed writ-
Confluent cultures were collected by
ten consent from all patients. Professional dentists
trypsinization (0.2% trypsin and 0.02% EDTA).
performed cavity preparations and tooth
The cells were plated at 3  103/cm2 on tissue
extractions.
culture treated 8-chambers glass slides (Becton
For studies on injured teeth, cavities were
Dickinson Labware, Lincoln Park, NJ, USA).
performed in healthy first premolars scheduled
After 6 weeks of culture, cells were fixed with
for extraction following the orthodontic treatment
70% ethanol for one hour at 4  C and processed
plan. Cavities 2–3 mm wide and 1.3–1.8 mm
for immunohistochemistry.
deep were prepared in the flank of the crown
(Fig. 6.3a). The pulp chambers were not exposed
during the preparation of the cavities. The cavities
6.2.5 Immunohistochemistry
were restored with the calcium hydroxide product
Dycal (Dentsplay, USA). Injured teeth were
Immunoperoxidase staining on sections was
extracted the ninth post-operation week.
performed as previously described (Mitsiadis
Extracted teeth were fixed in 10% neutral-
et al. 1992, 1993, 2003). Briefly, the sections
buffered formalin for 24 h, demineralized in
were deparaffinized, treated with 0.4% pepsin,
sodium formiate for 21 days, and then embedded
exposed to a 0.3% solution of hydrogen peroxide
in paraffin wax. Thereafter teeth were serially
in methanol, and then incubated overnight at 4  C
sectioned (6 μm thick sections) and processed
in a humid atmosphere with the polyclonal anti-
for immunohistochemistry. For the detection of
NGF antibody diluted 1:300 in PBS/0.2% BSA,
NGF in intact functional teeth, we proceeded with
the monoclonal anti-p75NTR antibody 20.4 (dilu-
immediate pulp extraction, followed by 10%
tion 1:1000), and the mouse anti-neurofilament
neutral-buffered formalin fixation for 24 h and
antibody (NFP, dilution 1:50). Peroxidase was
embedding in paraffin without prior
revealed by incubation with either 3.3-
decalcification.
diaminobenzidine tetrahydrochloride (DAB) or
3-amino-9-ethylcarbazole (AEC) reaction
solutions. After staining, the sections were
6.2.4 Cultures of Human Dental mounted with Eukit. In control sections the
Pulp Cells antibodies were omitted.
For immunohistochemistry on cultured dental
Human dental pulp cells were isolated from the pulp cells, the incubation with the NGF antibody
dental pulp of extracted impacted wisdom teeth of was performed overnight at 4  C after
68 T. A. Mitsiadis and P. Pagella

permeabilization of cells for 15 min with 0.5% 6.2.7 Embryonic Mouse Tissues
Triton X-100 in PBS. Upon staining, the slides
were mounted with Aquamount (BDH, Gurr, All mice were maintained and handled according
England). Controls were performed by to the Swiss Animal Welfare Law and in compli-
incubations with unrelated primary antibodies. ance with the regulations of the Cantonal Veteri-
For immunofluorescence identification of nary Office, Zurich (License number 151/2014).
β-III-tubulin and NuMA in microfluidic The animal facility provided standardized hous-
chambers, cells were incubated with the primary ing conditions, with a mean room temperature of
antibodies diluted 1:100 in PBS/0.2% BSA and 21  C, relative humidity of 50%  5%, and
then with a 1:250 dilution of Alexa-488-conju- 15 complete changes of filtered air per hour
gated anti-rabbit IgG and Alexa-568-conjugated (HEPA H 14 filter); air pressure was controlled
anti-mouse IgG. Cells were then counterstained at 50 Pa. The light/dark cycle in the animal rooms
with DAPI and mounted with Fluorescence was set to a 12 h/12 h cycle (lights on at 07:00,
Mounting Medium (S302380-2, DAKO, lights off at 19:00) with artificial light of approxi-
Agilent, Basel, Switzerland) before examination mately 40 Lux in the cage. The animals had
with a Leica DM6000 widefield fluorescent unrestricted access to sterilized drinking water,
microscope. and ad libitum access to a pelleted and extruded
mouse diet in the food hopper (Kliba No. 3436;
Provimi Kliba/Granovit AG, Kaiseraugst,
6.2.6 Preparation of Microfluidic Switzerland). Mice were housed in a barrier-
Devices protected specific pathogen-free unit and were
kept in groups of maximum five adult mice per
Microfluidic devices were prepared as previously cage in standard IVC cages (Allentown Mouse
described (Pagella and Mitsiadis 2020; Pagella 500; 194 mm  181 mm  398 mm, floor area
et al. 2015, Pagella et al. 2020b). Glass coverslips 500 cm2; Allentown, New Jersey, USA) with
were coated overnight at 37  C with 0.1 mg/mL autoclaved dust-free poplar bedding (JRS GmbH
poly-D-lysine and stored in 70% of ethanol at + Co KG, Rosenberg, Germany). A standard
4  C. Polydimethylsiloxane (PDMS) microfluidic cardboard house (Ketchum Manufacturing,
devices (Millipore A150, Switzerland, 2  2 cm) Brockville, Canada) served as a shelter, and tissue
were punched with a 1-mm-diameter biopsy papers were provided as nesting material. Addi-
punch on the neuronal side to enable the insertion tionally, crinklets (SAFE® crinklets natural, JRS
of the trigeminal ganglion and then sterilized GmbH + Co KG, Rosenberg, Germany) were
with 70% of ethanol. Both glass coverslips provided as enrichment and further nesting mate-
and microfluidic devices were then let dry rial. The specific pathogen-free status of the
completely under the laminar flow hood for 2 h. animals was monitored frequently and confirmed
The microfluidic devices were then mounted according to FELASA guidelines by a sentinel
onto the glass coverslips and pressed gently to program. The mice were free of all viral, bacterial,
ensure proper adhesion. After mounting, the and parasitic pathogens listed in FELASA
microfluidic devices were coated with laminin recommendations (FELASA working group on
(5 μg/mL in Neurobasal Medium) overnight at revision of guidelines for health monitoring of
37  C. To prevent the persistence of air bubbles rodents and rabbits et al. 2014).
in the culture chambers, the coated microfluidic C57/BL6J mice were time-mated. Successful
devices were placed under vacuum. After coat- mating was assessed by vaginal plug check, and
ing, the laminin solution was removed and the the day of plug was considered as the day of
culture chambers filled with the appropriate cul- embryonic development 0.5 (E0.5). Trigeminal
ture medium. ganglia were dissected from embryonic day 14.5
6 The Versatile Roles of Nerve Growth Factor in Neuronal Attraction. . . 69

Fig. 6.1 Distribution of NGF protein in dental tissues at d, dentine; df, dental follicle; dp, dental pulp; ide, inner
the bell stage of developing human teeth. (a–c) dental epithelium; o, odontoblasts; ode, outer dental epi-
Immunostaining against NGF (red color). In c, higher thelium; pa, preameloblasts; sr, stellate reticulum. Scale
magnification of the cusp area of the developing tooth bars: (a, b) 100 μm; (c) 25 μm
showing NGF expression in odontoblasts. Abbreviations:

(E14.5) mouse embryos. Dissections were


6.3 Results
performed in cold Dulbecco’s PBS. Dissected
ganglia were preserved in PBS, on ice, until cul-
6.3.1 NGF Distribution
ture. Trigeminal ganglia were then cultured in
in Odontoblasts of Human
Neurobasal Medium (Gibco) supplemented with
Embryonic Teeth
B27 (Gibco 17504-044), GlutaMAX, penicillin/
streptomycin (10 U/mL), 50 ng/mL nerve growth
Odontoblasts start to differentiate and secrete the
factor (NGF, R&D Systems), and 0.25 pM Arab-
dentine matrix during the late bell stage of human
inose Cytoside.
tooth development (23rd gestation week). At the
same time, inner dental epithelial cells stop divid-
ing and differentiate into the enamel-forming
6.2.8 Co-culture of Trigeminal
preameloblasts/ameloblasts. During this stage,
Ganglia and Human Dental
NGF immunoreactrivity was observed in all den-
Pulp Cells
tal epithelial cells, whereas labeling was absent
from the dental pulp (Fig. 6.1a, b). Strong NGF
Trigeminal ganglia were placed through the
staining was observed in differentiated functional
punched hole into the neuronal chamber of the
odontoblasts at the tip of the cusp (Fig. 6.1b, c),
microfluidic device. Ganglia were cultured alone
while immunoreactivity was also detected in the
for 5–7 days, until axons projected into the adja-
dental follicle (Fig. 6.1b).
cent chamber. Cells were then added to the
co-culture system. 20 μl of human dental pulp
cells in suspension were added directly in the
chamber at a density of 1  104 cells/chamber. 6.3.2 NGF and p75NTR Expression
Cells were allowed to be attached for 1 h and in Intact Functional
30 min, then 300 μl of the appropriate medium Human Teeth
were added to all chambers. Co-cultures were
maintained for 4 days. At the end of the culture In healthy functional human teeth (Fig. 6.2a),
period, the medium was removed and the samples NGF staining was observed in odontoblasts
were fixed in paraformaldehyde 4% (PFA 4%; (Fig. 6.2c, d), neurons (Fig. 6.2e), and some of
300 μl/chamber) for 15 min. the pulp fibroblasts (Fig. 6.2c, d) of freshly
70 T. A. Mitsiadis and P. Pagella

Fig. 6.2 Expression of NGF and p75NTR in adult intact subjected to cavity preparation. (g) shows NGF distribu-
and injured human teeth. (a) Histology of an adult intact tion far from the injury site. (h) shows NGF expression in
human tooth. (b) Immunostaining against p75NTR on newly formed odontoblasts under the injured area. (i)
intact tooth. (c–e) Immunostaining against NGF on the shows NGF expression in nerve fibers within the dental
dental pulp of intact teeth. (c, d) show NGF expression pulp. Abbreviations: d, dentine; dp, dental pulp; nf, nerve
in odontoblasts. (e) shows NGF expression in nerve fibers fibers; no, newly formed odontoblasts; o, odontoblasts; td,
localized within the dental pulp. (f) Histology of adult tertiary dentine; v, vessel. Scale bars: (a, f) 5 mm; (b, c)
human tooth after cavity preparation. (g–i) 80 μm; (d, e) 50 μm; (g) 1 mm; (h, i) 20 μm
Immunostaining against NGF on the dental pulp of teeth

extracted dental pulps, while strong p75NTR odontoblast-like cells, which produced a layer
staining was confined to nerve fibers (Fig. 6.2b). of tertiary dentine beneath the injury site
In the dental pulp, NGF immunoreactivity was (Fig. 6.2f, h). Strong NGF immunoreactivity
sporadically observed in few fibroblasts was detected in the odontoblast-like cells
(Fig. 6.2c–e). Immunostaining for NGF and (Fig. 6.2h), but also in odontoblasts located at a
p75NTR was not detected in control sections distance from the cavity preparation site
where the antibodies were omitted (data not (Fig. 6.2g). NGF staining was also observed in
shown). few pulp fibroblasts (Fig. 6.2g) and neurons
(Fig. 6.2i). NGF staining was not detected in
control sections (data not shown).
6.3.3 NGF Distribution in Functional
Human Teeth After Cavity
Preparation 6.3.4 NGF Expression in Human
Dental Pulp Cells In Vitro
Tooth cavity preparation leads to the apoptosis of
the odontoblasts beneath the injury site (Mitsiadis In order to understand better the role of NGF in
et al. 2008). Nine weeks post-operation, the tooth repair, we have analyzed NGF expression in
apoptotic odontoblasts were replaced by dental pulp cells freshly extracted from human
6 The Versatile Roles of Nerve Growth Factor in Neuronal Attraction. . . 71

Fig. 6.3 NGF protein distribution in cultured human growth from the chamber containing the trigeminal gan-
dental pulp cells. (a) Freshly extracted human intact glion toward the chamber containing the dental pulp cells.
teeth, which are used for the collection of dental pulp (g) Higher magnification of f, showing the passage of
cells. (b–d) Expression of NGF in dental pulp cells axons through the microgrooves. (h) Immunofluorescent
cultured under mineralizing conditions for 2 days (not staining against βIII-tubulin (green color) showing trigem-
yet confluent cells, b), 1 week (confluent cells, c), and inal neurons establishing contacts (white arrowheads) with
4 weeks (formation of mineralization nodules, d). (e) dental pulp cells (marked by NuMA nuclear staining, red
Schematic representation of the microfluidic “organ-on- color). DAPI (blue color) marks the nuclei of the cells.
a-chip” device that is used for the co-culture of the trigem- Abbreviations: cc1, culture chamber 1; cc2, culture cham-
inal ganglion with the dental pulp cells. (f) Immunofluo- ber 2; mg, microgrooves; tgg, trigeminal ganglion. Scale
rescent staining against βIII-tubulin showing neuronal bars: (b–d, i) 25 μm; (f) 500 μm; (g) 75 μm

healthy teeth (Fig. 6.3a). Cells were cultured in (Fig. 6.3f, g), human dental pulp cells were added
mineralizing conditions. All dental pulp cells to the other side of the microfluidic device. We
cultured in the presence of ß-glycerophosphate then analyzed the outgrowth of trigeminal
for different periods of time up to 4 weeks neurons co-cultured with dental pulp cells. Tri-
exhibited NGF immunoreactivity (Fig. 6.3b–d). geminal ganglia and dental pulp cells were
The strongest NGF staining was observed in con- co-cultured for 4 days. Staining with βIII-tubulin
fluent cells (Fig. 6.3c) and in cells forming and analysis using a wide-field fluorescent
mineralized nodules (Fig. 6.3d). microscopy showed many trigeminal axonal
projections toward the dental pulp cell compart-
ment (Fig. 6.3b). Rich axonal networks were
6.3.5 Co-culture of Trigeminal observed in the dental cell compartment, where
Ganglia and Human Dental neuronal projections established direct contacts
Pulp Cells in Microfluidic with the dental pulp cells (Fig. 6.3h).
Devices

Trigeminal ganglia from embryonic day 14.5 6.4 Discussion


C57/BL6J mouse embryos were isolated and
cultured alone at the neuronal compartment of A complex network of molecules ensures dental
the microfluidic device (Fig. 6.3e). Upon the ini- tissue integrity and function during tooth devel-
tiation of axonal outgrowths (4 days of culture) opment, homeostasis, and regeneration. Among
72 T. A. Mitsiadis and P. Pagella

these molecules, NGF emerged as an important experiments showed that NGF is upregulated dur-
modulator of innervation, cytodifferentiation, and ing the mineralization of dental pulp fibroblasts
mineralization processes in human and rodent and suggested an active role for this molecule in
teeth (Mitsiadis et al. 1992, 1993, 2017; Mitsiadis hard tissue formation (Mitsiadis et al. 2017), a
and Pagella 2016; Mitsiadis and Luukko 1995), function similar to that recently reported during
thus indicating that the functions of NGF in dental osteogenesis (Tomlinson et al. 2017).
tissues are highly conserved among different spe- The role of innervation in the tooth repair
cies (Mitsiadis et al. 1992, 1993; Mitsiadis and events is not yet well elucidated. It is well
Pagella 2016). In our previous studies, we have established from previous pioneer studies that
demonstrated that NGF and its high and low NGF plays a pivotal role in tissue innervation,
affinity receptors, TrkA and p75NTR respectively, acting as a neurotrophic and neuroattractive factor
are expressed in specific cell types during the (Levi-Montalcini 1987; Levi-Montalcini and
various developmental stages of human teeth Aloe 1980; Ruit et al. 1992; Davies et al. 1981;
(Mitsiadis and Pagella 2016). In the present Cohen et al. 1954). Human tooth is one of the
study, we highlight the roles of NGF in the dif- most innervated organs of the body (Mitsiadis
ferentiation of odontoblasts, formation of dentine, and Luukko 1995; Mitsiadis et al. 2017), and
and establishment of dental pulp innervation. Our therefore NGF could play an important role in
findings confirm the previous studies and show pain perception (Ugolini et al. 2007; Kumar and
that NGF expression in odontoblasts precedes the Mahal 2012). A dense neuronal plexus is com-
dental pulp innervation, thus indicating that this posed around the NGF-secreting odontoblasts
molecule is involved in odontoblasts differentia- (Mitsiadis et al. 2017; Mitsiadis and Pagella
tion, dentine matrix production, and mineraliza- 2016). NGF upregulation upon tooth injury
tion events during odontogenesis. In contrast, the promotes reorganization of the neuronal network
low levels of NGF expression by odontoblasts of within the dental pulp (Mitsiadis et al. 2017). This
functional intact adult teeth are correlated to the reorganization and the neuroattractive properties
slow, throughout life deposition of secondary of dental pulp cells on trigeminal ganglia could be
dentine. However, the synthesis of dentine by studied in vitro using microfluidic “organ-on-a-
odontoblasts is accelerated in carious or injured chip” devices (Pagella et al. 2021). These
teeth (Mitsiadis and Rahiotis 2004; About and miniaturized platforms have already been used
Mitsiadis 2001), a process associated with NGF to emulate the neurotrophic properties of dental
upregulation (Mitsiadis et al. 2017). Deep cavity pulp stem cells, as well as dental epithelial cancer
preparations are more drastic and lead to exten- stem cells (ameloblastoma) (Pagella and
sive apoptosis of the odontoblasts beneath the Mitsiadis 2020; Pagella et al. 2020a, b). These
injured site (Mitsiadis et al. 2008). In this case, two stem cell populations produce and secrete
dental pulp cells are recruited at the injured site; NGF, which attracts the neurons toward them
they differentiate into odontoblast-like cells and (Pagella et al. 2020a, b). Here we show that, in
form a reparative dentin (Mitsiadis and Rahiotis the microfluidic devices, neurons are also
2004). Our present findings show that in these attracted by dental pulp fibroblasts and establish
conditions, NGF expression is upregulated not complex and abundant contacts with them. In
only in odontoblast-like cells but also in addition to this function, NGF can have stimula-
odontoblasts located far from the injury site, in tory roles in regenerative processes by
pulp fibroblasts, and neurons, thus clearly modulating the secretion of neuropeptides and
demonstrating the involvement of this molecule other bioactive molecules by neurons (Pagella
in a complex dental tissue healing process. et al. 2014). Stem cells are involved in dental
Indeed, our present and previous in vitro pulp regeneration, and therefore nerve-derived
6 The Versatile Roles of Nerve Growth Factor in Neuronal Attraction. . . 73

Fig. 6.4 Schematic


representation of the
functions of NGF in
odontoblasts. NGF plays
significant roles in the
differentiation of
odontoblasts, the
mineralization of the dentin
matrix, and the
establishment of a rich
network of neurons in the
dental pulp

molecules affect the behavior of stem cells in the


References
healing processes of many organs, including teeth
(Kumar and Brockes 2012; Orsini et al. 2018; About I, Mitsiadis TA (2001) Molecular aspects of tooth
Mitsiadis et al. 2015). pathogenesis and repair: in vivo and in vitro models.
In conclusion, our present and previous Adv Dent Res 15:59–62
results in human teeth (Mitsiadis et al. 2017; Asaumi K, Nakanishi T, Asahara H, Inoue H, Takigawa M
(2000) Expression of neurotrophins and their receptors
Mitsiadis and Pagella 2016) clearly show that (TRK) during fracture healing. Bone 26:625–633
NGF is associated with the differentiation of Benedetti M, Levi A, Chao MV (1993) Differential
odontoblasts, the formation of dentine, and the expression of nerve growth factor receptors leads to
organization of the neuronal network within the altered binding affinity and neurotrophin
responsiveness. Proc Natl Acad Sci U S A
dental pulp during development, pathology, and 90:7859–7863
regeneration (Fig. 6.4). Bibel M, Hoppe E, Barde YA (1999) Biochemical and
functional interactions between the neurotrophin
Ackowledgments This work was supported by institu- receptors trk and p75NTR. EMBO J 18:616–622
tional grants from the University of Zurich. Byers MR (1990) Segregation of NGF receptor in sensory
receptors, nerves and local cells of teeth and
periodontium demonstrated by EM immunocytochem-
Competing Financial Interests The authors declare istry. J Neurocytol 19:765–775
that there are no conflicts of interest associated with Byers MR, Schatteman GC, Bothwell M (1990) Multiple
this work. functions for NGF receptor in developing, aging and
74 T. A. Mitsiadis and P. Pagella

injured rat teeth are suggested by epithelial, mesenchy- Mitsiadis TA, Luukko K (1995) Neurotrophins in
mal and neural immunoreactivity. Development odontogenesis. Int J Dev Biol 39:195–202
109:461–471 Mitsiadis TA, Pagella P (2016) Expression of nerve
Byun JH, Lee JH, Choi YJ, Kim JR, Park BW (2008) growth factor (NGF), TrkA, and p75(NTR) in devel-
Co-expression of nerve growth factor and p75NGFR oping human fetal teeth. Front Physiol 7:338
in the inferior alveolar nerve after mandibular distrac- Mitsiadis TA, Rahiotis C (2004) Parallels between tooth
tion osteogenesis. Int J Oral Maxillofac Surg development and repair: conserved molecular
37:467–472 mechanisms following carious and dental injury. J
Chesa PG, Rettig WJ, Thomson TM, Old LJ, Melamed Dent Res 83:896–902
MR (1988) Immunohistochemical analysis of nerve Mitsiadis TA, Dicou E, Joffre A, Magloire H (1992)
growth factor receptor expression in normal and malig- Immunohistochemical localization of nerve growth
nant human tissues. J Histochem Cytochem factor (NGF) and NGF receptor (NGF-R) in the devel-
36:383–389 oping first molar tooth of the rat. Differentiation
Christensen LR, Mollgard K, Kjaer I, Janas MS (1993) 49:47–61
Immunocytochemical demonstration of nerve growth Mitsiadis TA, Couble P, Dicou E, Rudkin BB, Magloire H
factor receptor (NGF-R) in developing human fetal (1993) Patterns of nerve growth factor (NGF),
teeth. Anat Embryol (Berl) 188:247–255 proNGF, and p75 NGF receptor expression in the rat
Cohen S, Levi-Montalcini R, Hamburger V (1954) A incisor: comparison with expression in the molar. Dif-
nerve growth-stimulating factor isolated from sarcom ferentiation 54:161–175
as 37 and 180. Proc Natl Acad Sci U S A Mitsiadis TA, Romeas A, Lendahl U, Sharpe PT, Farges
40:1014–1018 JC (2003) Notch2 protein distribution in human teeth
Davies AM, Lumsden AG, Slavkin HC, Burnstock G under normal and pathological conditions. Exp Cell
(1981) Influence of nerve growth factor on the embry- Res 282:101–109
onic mouse trigeminal ganglion in culture. Dev Mitsiadis TA, De Bari C, About I (2008) Apoptosis in
Neurosci 4:150–156 developmental and repair-related human tooth
Felasa Working Group on Revision of Guidelines for remodeling: a view from the inside. Exp Cell Res
Health Monitoring of Rodents and Rabbits, Mahler 314:869–877
Convenor M, Berard M, Feinstein R, Gallagher A, Mitsiadis TA, Orsini G, Jimenez-Rojo L (2015) Stem cell-
Illgen-Wilcke B, Pritchett-Corning K, Raspa M based approaches in dentistry. Eur Cell Mater
(2014) FELASA recommendations for the health mon- 30:248–257
itoring of mouse, rat, hamster, guinea pig and rabbit Mitsiadis TA, Magloire H, Pagella P (2017) Nerve growth
colonies in breeding and experimental units. Lab Anim factor signalling in pathology and regeneration of
48:178–192 human teeth. Sci Rep 7:1327
Grob PM, Ross AH, Koprowski H, Bothwell M (1985) Nosrat IV, Widenfalk J, Olson L, Nosrat CA (2001) Dental
Characterization of the human melanoma nerve growth pulp cells produce neurotrophic factors, interact with
factor receptor. J Biol Chem 260:8044–8049 trigeminal neurons in vitro, and rescue motoneurons
Ibanez CF, Simi A (2012) p75 neurotrophin receptor sig- after spinal cord injury. Dev Biol 238:120–132
naling in nervous system injury and degeneration: par- Nosrat I, Seiger A, Olson L, Nosrat CA (2002) Expression
adox and opportunity. Trends Neurosci 35:431–440 patterns of neurotrophic factor mRNAs in developing
Kumar A, Brockes JP (2012) Nerve dependence in tissue, human teeth. Cell Tissue Res 310:177–187
organ, and appendage regeneration. Trends Neurosci Orsini G, Pagella P, Mitsiadis TA (2018) Modern trends in
35:691–699 dental medicine: an update for internists. Am J Med
Kumar V, Mahal BA (2012) NGF—the TrkA to successful 131:1425–1430
pain treatment. J Pain Res 5:279–287 Pagella P, Mitsiadis TA (2020) Analysis of tooth innerva-
Lee R, Kermani P, Teng KK, Hempstead BL (2001) Reg- tion in microfluidic coculture devices. Methods Mol
ulation of cell survival by secreted proneurotrophins. Biol 2155:99–106
Science 294:1945–1948 Pagella P, Jimenez-Rojo L, Mitsiadis TA (2014) Roles of
Levi-Montalcini R (1987) The nerve growth factor innervation in developing and regenerating orofacial
35 years later. Science 237:1154–1162 tissues. Cell Mol Life Sci 71:2241–2251
Levi-Montalcini R, Aloe L (1980) Tropic, trophic, and Pagella P, Miran S, Mitsiadis T (2015) Analysis of devel-
transforming effects of nerve growth factor. Adv oping tooth germ innervation using microfluidic
Biochem Psychopharmacol 25:3–15 co-culture devices. J Vis Exp e53114
Lewin G, Carter BD (2014) Neurotrophic factors. Pagella P, Caton J, Meisel CT, Mitsiadis TA (2020a)
Springer, Berlin Ameloblastomas exhibit stem cell potential, possess
Luukko K, Arumae U, Karavanov A, Moshnyakov M, neurotrophic properties, and establish connections
Sainio K, Sariola H, Saarma M, Thesleff I (1997) with trigeminal neurons. Cells 9
Neurotrophin mRNA expression in the developing Pagella P, Miran S, Neto E, Martin I, Lamghari M,
tooth suggests multiple roles in innervation and organ- Mitsiadis TA (2020b) Human dental pulp stem cells
ogenesis. Dev Dyn 210:117–129 exhibit enhanced properties in comparison to human
6 The Versatile Roles of Nerve Growth Factor in Neuronal Attraction. . . 75

bone marrow stem cells on neurites outgrowth. FASEB Tomellini E, Lagadec C, Polakowska R, Le Bourhis X
J 34:5499–5511 (2014) Role of p75 neurotrophin receptor in stem cell
Pagella P, Cordiale A, Marconi GD, Trubiani O, Rasponi biology: more than just a marker. Cell Mol Life Sci
M, Mitsiadis TA (2021) Bioengineered tooth emula- 71:2467–2481
tion systems for regenerative and pharmacological Tomlinson RE, Li Z, Li Z, Minichiello L, Riddle RC,
purposes. Eur Cell Mater 41:502–516 Venkatesan A, Clemens TL (2017) NGF-TrkA signal-
Reichardt LF (2006) Neurotrophin-regulated signalling ing in sensory nerves is required for skeletal adaptation
pathways. Philos Trans R Soc Lond B Biol Sci to mechanical loads in mice. Proc Natl Acad Sci U S A
361:1545–1564 114:E3632–E3E41
Ross AH, Grob P, Bothwell M, Elder DE, Ernst CS, Ugolini G, Marinelli S, Covaceuszach S, Cattaneo A,
Marano N, Ghrist BF, Slemp CC, Herlyn M, Atkinson Pavone F (2007) The function neutralizing anti-TrkA
B et al (1984) Characterization of nerve growth factor antibody MNAC13 reduces inflammatory and neuro-
receptor in neural crest tumors using monoclonal pathic pain. Proc Natl Acad Sci U S A 104:2985–2990
antibodies. Proc Natl Acad Sci U S A 81:6681–6685 Woloszyk A, Buschmann J, Waschkies C, Stadlinger B,
Ruit KG, Elliott JL, Osborne PA, Yan Q, Snider WD Mitsiadis TA (2016) Human dental pulp stem cells and
(1992) Selective dependence of mammalian dorsal gingival fibroblasts seeded into silk fibroin scaffolds
root ganglion neurons on nerve growth factor during have the same ability in attracting vessels. Front
embryonic development. Neuron 8:573–587 Physiol 7:140
An Overview of Adult Neurogenesis
7
Filipa F. Ribeiro and Sara Xapelli

Abstract review the main properties of the adult neuro-


Neurogenesis is maintained in the mammalian genic niches.
brain throughout adulthood in two main
regions: the subventricular zone (SVZ) of the
lateral ventricles and the subgranular zone
(SGZ) of the hippocampal dentate gyrus. 7.1 Adult Neurogenesis: An
Adult neurogenesis is a process composed of Historical Perspective
multiple steps by which neurons are generated
from dividing adult neural stem cells and For over 100 years, a central dogma in the field of
migrate to be integrated into existing neuronal neuroscience was that new neurons are not added
circuits. Alterations in any of these steps to the adult mammalian brain. Indeed, until
impair neurogenesis and may compromise recently, it was assumed that neurogenesis, the
brain function, leading to cognitive formation of new neurons, in the mammalian
impairment and neurodegenerative diseases. brain was thought to occur only during develop-
Therefore, understanding the cellular and ment, stopping soon after birth. Then, the brain
molecular mechanisms that modulate adult would remain a fixed structure, incapable of pro-
neurogenesis is the centre of attention of ducing new neurons during adulthood. This cen-
regenerative research. In this chapter, we tral dogma in neuroscience was defended by
influential figures, such as the Spanish Nobel
Prize Winner, the neuroanatomist Santiago
Ramón y Cajal (Ramón y Cajal and May 1928),
who contributed for the scepticism of the scien-
tific community.
In 1962, with the introduction of [3H]-thymi-
dine autoradiography, a technique where [3H]-
thymidine is incorporated into the DNA of divid-
F. F. Ribeiro · S. Xapelli (*)
ing cells, Joseph Altman reported the first evi-
Instituto de Farmacologia e Neurociências, Faculdade de
Medicina, Universidade de Lisboa, Lisbon, Portugal dence of [3H]-thymidine presence in neurons
(Altman 1962). He also demonstrated that new
Instituto de Medicina Molecular João Lobo Antunes,
Faculdade de Medicina, Universidade de Lisboa, Lisbon, neurons are present in different areas of the post-
Portugal natal brain, such as in the neocortex, the dentate
e-mail: anaribeiro@medicina.ulisboa.pt; gyrus (DG) of the hippocampus, the olfactory
sxapelli@medicina.ulisboa.pt

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 77


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_7
78 F. F. Ribeiro and S. Xapelli

bulb (OB) and the cerebellar cortex of different towards a more plastic brain, where new neurons
mammals (Altman 1963; Altman and Das 1965, are continuously being formed throughout adult-
1966, 1967). hood in specific areas.
Despite Altman’s discoveries, the evidence of
postnatal neurogenesis was ignored for two more
decades. It was then that, with electron micros- 7.2 The Doubts Behind Adult
copy, Kaplan and his colleagues confirmed Human Neurogenesis
Altman’s results by showing the existence of
[3H]-thymidine-labelled neurons in the DG, OB The existence of adult human neurogenesis was
and visual cortex, as well as mitotic cells in the controversial, with great scepticism among the
SVZ of adult macaque monkeys (Kaplan and scientific community. In 2013, Jonas Friesen’s
Hinds 1977; Kaplan 1981, 1983; Kaplan and lab used the retrospective birth dating of cells
Bell 1984). Simultaneously, Nottebohm and with 14C, which constituted an innovative strat-
colleagues demonstrated that neurogenesis occurs egy to study neurogenesis in the human adult
during adulthood in the brain of songbirds, brain. This carbon isotype, which resulted from
namely in the SVZ and in the forebrain, including nuclear bomb tests of the Cold War, was spread in
in the hippocampus. These SVZ new neurons the atmosphere, and incorporated in the food
migrated to the vocal control nucleus to replace through plant photosynthesis and then in animals
pre-existent neurons in a process dependent on eating plants, and finally into human body. In the
synaptic inputs (Goldman and Nottebohm 1983; human body, mitotic cells integrated 14C into
Paton and Nottebohm 1984; Nottebohm 1985; DNA, thus allowing to determinate cell birth dat-
Burd and Nottebohm 1985). Later, Reynolds ing by measuring the concentration of 14C in the
and Weiss isolated proliferating cells from the DNA of post-mortem brain. Carbon dating has
adult mouse striatum, demonstrating that these demonstrated that neuronal turnover mainly
cells have the potential to originate neurons and occurs in hippocampus and striatum, with
astrocytes (Reynolds and Weiss 1992). 700 neurons being added each day into the
By the nineties, the phenotypic identification human DG. Moreover, this method failed to
of new generated cells was facilitated by the reveal neuronal turnover in the OB or in the
emergence of new techniques, namely the syn- cerebral neocortex of the adult human brain
thetic thymidine analogue BrdU (5-bromo-2- (Bhardwaj et al. 2006; Bergmann et al. 2012;
0
-deoxyuridine) which is incorporated during the Spalding et al. 2013; Ernst et al. 2014). However,
S-phase of the cell cycle and cell-type specific the use of carbon dating was also pointed to be
markers associated with immunohistochemistry prone to contamination, providing inaccurate
and confocal microscopy. These techniques results. Consequently, the occurrence of adult
contributed to demonstrate in vivo adult neurogenesis in the human brain was still not
neurogenesis in mammals, such as in rodents consensual amongst the scientific community.
(Kuhn et al. 1996) and primates (Gould et al. In the past 5 years, several studies have
1998, 1999a; Kornack and Rakic 1999). This focused in resolving the unsolved mystery of
kind of prospective labelling of newborn cells whether new neurons are formed or not in the
with BrdU was a huge improvement for adult human adult brain. In 2016, Dennis and
mammalian neurogenesis studies, showing its colleagues using immunohistochemical methods
conservation in almost all mammals, including failed to demonstrate any continued neurogenesis
in humans, by demonstrating the presence of in the adult human. They examined cell prolifera-
newborn neurons in the adult human DG tion and neurogenesis in 23 individuals aged
(Eriksson et al. 1998). 0.2–59 years and observed a marked decline in
Together, this evidence allowed concluding neurogenesis in both SVZ and SGZ regions in
that the brain does not have an immutable neuro- early childhood. Moreover, microglia were the
nal composition, with the concept moving only proliferating cells found in both niches and
7 An Overview of Adult Neurogenesis 79

adjacent parenchyma after 3 years of age (Dennis up to the ninth decade of life. In contrast,
et al. 2016). In 2018, Sorrells and colleagues neurogenesis declined in the DG of patients with
collected post-mortem tissue from healthy adults Alzheimer’s disease with disease progression
and postoperative hippocampal tissue from (Moreno-Jiménez et al. 2019). Tobin and
humans with epilepsy. The subjects ranged from colleagues not only reported that hippocampal
foetuses with 14 gestational weeks to adults with neurogenesis is persistent in aged human brains,
77 years old. Samples were stained with the fluo- through the tenth decade of life, but also that
rescent marker antibodies Ki-67 (nuclear protein hippocampal neurogenesis is still present,
associated with cellular proliferation), SOX1 (Sex although not as significantly, in patients with
determining region Y-box 1) and SOX2 (Sex mild cognitive impairments and Alzheimer’s dis-
determining region Y-box 2) to identify neural ease (Tobin et al. 2019).
progenitor cells, and DCX (Doublecortin) and Bioinformatic methods have also been used to
PSA-NCAM (Polysialylated-neural cell adhesion analyse the differential expression of
molecule) to identify immature neurons. Results neurogenesis signature markers in the human hip-
showed that the density of proliferating cells and pocampal transcriptome, from prenatal, through
young neurons decreased with age, with the latter infancy and adolescence, to adulthood. Results
reducing from around 1618 cells/mm2 at birth to found persistent but minimal hippocampal
12 cells/mm2 by age 7, to 2 cells/mm2 by age neurogenesis in adult humans, suggesting that a
13, and to almost none in the samples of significant number of newborn hippocampal cells
individuals with more than 18 years old. With could be glial cells and not neurons (Kumar et al.
this study, the authors concluded that the recruit- 2019).
ment of new neurons to the primate hippocampus Apart from the DG, Ernst and colleagues
decreases rapidly during the first years of life, observed that new neurons integrate in the adult
with neurogenesis in the DG being extremely human striatum. They showed that DCX-positive
rare, or extinct, in adult humans (Sorrells et al. and PSA-NCAM-positive neuroblasts are present
2018). both in the SVZ and in the adjacent striatum of
On the other hand, at the same time, other control human brains, with these cells being
studies emerged supporting the existence of responsible for the turnover of striatal
adult human hippocampal neurogenesis. Using interneurons (Ernst et al. 2014). While in rodents,
similar immunohistochemical methods, Boldrini new neurons generated in the SVZ migrate to the
and colleagues studied the DG from post-mortem OB to become interneurons, only entering the
healthy humans aged 14–79 years and found that striatum under pathological conditions, such as
the total numbers of intermediate neural in ischemia, in humans SVZ-derived cells mainly
progenitors and immature neurons, which were migrate to the adjacent striatum to become
thousands, were stable across ages. Moreover, interneurons, and rarely to the OB (Kempermann
they observed angiogenesis and changes in the 2014). Apart from the physiological role, which is
volume of DG with aging. They employed unbi- yet unknown, it is thought that striatal adult
ased stereology, which is the gold standard for neurogenesis may be important in pathological
counting the number of neurons, and they conditions, such as in cerebral ischemia, where
concluded that while DG neurogenesis and vol- upregulation of neurogenesis could reduce dam-
ume is unchanged with aging, there is a decline in age, and psychiatric disorders, where
quiescent stem cell pools, angiogenesis, and neurogenesis could enhance susceptibility to ill-
neuroplasticity (Boldrini et al. 2018). More ness or support its treatment (Inta et al. 2015).
recently, Moreno-Jiménez and colleagues Whether and/or how much adult neurogenesis
identified thousands of immature neurons really occurs in humans is a question of great
exhibiting variable degrees of maturation along importance to understand its functional role in
the differentiation stages of adult hippocampal our species (Ghosh 2019). Importantly, adult
neurogenesis in neurologically healthy humans neurogenesis has been widely studied in other
80 F. F. Ribeiro and S. Xapelli

organisms. These findings paved the way for a Weiss 1992; Kilpatrick and Bartlett 1993; Palmer
new research field that, in the last two decades, et al. 1997; Gage et al. 1998).
has been extensively explored and has provided In contrast to what is observed in vitro, in vivo
several insights on the identity of neural stem clonal studies have shown limited NSC self-
cells (NSCs), on the properties and cellular com- renewal capacity with a fast progeny expansion
position of neurogenic niches, the cellular dynam- (Encinas et al. 2011; Calzolari et al. 2015), with a
ics and the migratory behaviour of newly NSC being mostly unilineage, generating
generated cells, as well as the survival, integration neurons, and, sometimes, having the capacity to
and functional relevance of these adult born cells. generate either a neuron or a glial cell (Encinas
et al. 2011; Bonaguidi et al. 2011; Calzolari et al.
2015). Although an adult NSC might be intrinsi-
cally multipotent, displaying the capacity to gen-
7.3 Neural Stem Cells
erate cells from the neuronal and glial lineages,
the niche environment seems to limit, in some
One of the things that was against the acceptance
way, its potential and long-term self-renewal
of adult neurogenesis was the unconceivable idea
capacity. The extracellular environment, such as
that mature neurons, considered to be terminally
the different types of neighbouring cells that
differentiated and unable to re-enter the cell-
secrete or present distinct signals on their plasma
division cycle, could be able to divide to originate
membrane, as well as molecules of the extracellu-
new neurons. The concept of NSC (Kempermann
lar matrix, is essential to establish a difference
and Gage 1999), being both proliferative cells,
between lineage and potential (Götz et al. 2015).
with the capacity to self-renew, and multipotent
Therefore, this will have an impact in cell genesis,
cells, meaning that after exiting the cell cycle
turning this process region-specific. For instance,
initiate differentiation towards a specialized neu-
in the SVZ, the higher number of neuronal line-
ral cell type, was introduced more recently (Hall
age progenitors is located in the lateral wall, with
and Watt 1989; Gage 2000; Temple 2001)
NSCs being more prone for neurogenesis, while
(Fig. 7.1).
oligodendrocyte progenitors are enriched in the
NSCs were first identified (Reynolds and
septal wall, with gliogenesis being more substan-
Weiss 1992) from isolated cells from the striatum
tial in this area (Mizrak et al. 2019). While the
of the adult mouse brain. When cultured in the
lineage of a cell is what a single NSC originates
presence of epidermal growth factor (EGF), these
in vivo, in a conditioned environment, the poten-
isolated cells were able to extensively proliferate
tial corresponds to what a single NSC can do
as non-adherent spherical structures, the
when it is exposed to different environments
neurospheres, which were composed of clonally
(Götz et al. 2015).
derived precursors. These cells were able to give
rise to more neurospheres with the same pheno-
type, thus showing self-renewal capacity. In con-
7.4 Adult Neurogenic Niches
trast, when removing the growth factors from the
medium and providing an adhesion substrate of
In the adult mammalian brain, NSCs are mainly
poly-L-ornithine, neurosphere cells formed a
restricted to specific brain regions, the SVZ in the
pseudomonolayer culture which differentiated
lateral ventricles and the subgranular zone (SGZ)
into astrocytes and neurons, thus exhibiting
in the DG of the hippocampus where constitutive
multipotency (Reynolds and Weiss 1992).
neurogenesis takes place throughout adulthood
The two main properties that are attributed to
(Fig. 7.2). These regions have a specialized and
NSCs are the capacity to self-renew through cell
diverse microenvironment that embeds NSCs, the
division and to generate a differentiated progeny
reason why it is called a niche. In these niches,
of the three neural lineages, namely neurons,
NSCs generate neural progenitor cells (NPCs), or
astrocytes and oligodendrocytes (Reynolds and
transit amplifying progenitors, which rapidly
7 An Overview of Adult Neurogenesis 81

Fig. 7.1 Neural stem cells


are self-renewing and
multipotent cells with the
capacity to generate
neurons, astrocytes and
oligodendrocytes (from
Ribeiro 2017)

divide and form migrating neuroblasts. The generation of new neurons from the SVZ
Neuroblasts differentiate into neurons that inte- and SGZ is possible because adult NSCs are
grate into the pre-existing circuits. While SGZ embedded in a specialized and diverse microen-
newborn neurons migrate short distances to inte- vironment that is the niche and that is permissive
grate into the circuits of the DG granule cell layer, for neurogenesis to occur. Apart from NSCs and
SVZ newborn neurons migrate long distances NPCs, the neurogenic niche is also composed of
towards the OB (Alvarez-Buylla and Lim 2004). glial, endothelial and immune (as microglia and
macrophages) cells. Giving all this cell diversity,
the neurogenic niche provides an unique milieu
consisting of extracellular matrix, short and long-
ranged signalling factors, cell-to-cell contacts,
which is responsible for tightly regulating NSC
self-renewal and differentiation (Palmer et al.
2000; Alvarez-Buylla and Lim 2004; Ma et al.
2005; Merkle and Alvarez-Buylla 2006).
Vasculature and systemic factors also have a
great impact on adult neurogenesis. The impor-
tance of this relationship has been demonstrated
Fig. 7.2 The main neurogenic niches in the adult mam- by heterochronic parabiosis experiments. In this
malian brain are the subventricular zone (SVZ), in the parabiosis experiments, the blood circulations of
lateral ventricles (LV), and the subgranular zone (SGZ),
in the dentate gyrus (DG) of the hippocampus. These a young and old mice were surgically connected
neurogenic niches contain neural stem cells (NSCs) with through the attachment of their skins on the lateral
the capacity to divide and generate new neurons. Newborn torso. Heterochronic parabiosis experiments
neurons from SGZ migrate short distances to be integrated showed that the presence of young mouse blood
into the circuits of the DG, whereas SVZ newborn neurons
migrate long distances, through the rostral migratory in the circulatory system of an older animal
stream (RMS), towards the olfactory bulb (OB) (from resulted in increased proliferation of NPCs and
Ribeiro 2017) increased adult neurogenesis in the neurogenic
82 F. F. Ribeiro and S. Xapelli

niches. Moreover, old heterochronic parabionts Although more limited, this process has been
displayed increased vessel volume and cerebral observed in neocortex (Gould et al. 1999b;
blood flow comparable to young mice. In con- Dayer et al. 2005; Ohira et al. 2010), amygdala
trast, adult neurogenesis in the younger animal (Bernier et al. 2002), substantia nigra (Lie et al.
was reduced (Villeda et al. 2011; Katsimpardi 2002; Zhao et al. 2003; Zhao and Janson Lang
et al. 2014). 2009), hypothalamus (Kokoeva et al. 2005, 2007)
SVZ NSCs are in contact with the cerebrospi- and striatum (Bédard et al. 2006). This evidence
nal fluid (CSF) which is produced inside the contributes to the idea that the potential of the
ventricles by a highly vascular structure, the cho- brain to generate new neurons is greater than
roid plexus (CP). The CP acts as an interface initially thought. Nevertheless, the neurogenic
between the brain and the periphery. Moreover, process in these non-classic neurogenic areas is
many altered factors in the blood have been still in the beginning of its characterisation, with
shown to influence SVZ neurogenesis by affect- some evidence still controversial (Breunig et al.
ing the activated NSCs. Besides not being in 2007). Progenitor cells residing in non-classic
contact with the CSF, DG NSCs have an ample neurogenic locations, such as the substantia
interaction with the DG vasculature, which starts nigra, can give rise to neurons when transplanted
from the arterioles of the hippocampal fissure, into neurogenic niches, but the contrary is not
passing as capillaries through the granule cell possible. In in vitro conditions, these restricted
(GC) layer along the rostro-caudal axis (Ghosh progenitors can give rise to both neurons and glia.
2019). These experiments suggest that the niches have
Due to differences in the microenvironment of the capacity to provide both ‘restrictive’ and
SGZ and SVZ, self-renewal and differentiation of ‘instructive’ cues to a neural progenitor for a
NSCs are differently regulated. On the one hand, certain fate, depending on its location and
self-renewal through symmetric division requirements (Ghosh 2019).
represents the fate of a minority of DG NSCs,
with most of them undergoing asymmetric cell
division resulting in one NSCs and one 7.5 The Subventricular Zone (SVZ)
differentiated cell, either an astrocyte or neuron, Neurogenic Niche
but never an oligodendrocyte under normal
conditions (Encinas et al. 2011; Bonaguidi et al. The SVZ is the major neurogenic niche of the
2011). On the other hand, self-renewal and differ- adult mammalian brain, and it is localised along
entiation of SVZ NSCs occur through symmetric the ependymal cell layer that lines the walls of the
cell division. About 20–30% of SVZ NSCs divide lateral ventricles (Fig. 7.3) (Doetsch et al. 1997).
symmetrically to maintain their pool, whereas SVZ contains a subpopulation of quiescent cells
most NSCs undergo differentiative symmetric with astrocyte-like characteristics, the type B
divisions, undergoing neuronal lineage progres- cells, which function as NSCs (Morshead et al.
sion (Obernier et al. 2018). In addition to this, 1994; Nam and Benezra 2009).
adult DG NSCs possess limited self-renewal, pro- In the SVZ niche, the type B cells are slow
liferation and migration capacities (Seaberg and dividing cells. These cells are in direct contact
van der Kooy 2002; Bull and Bartlett 2005), with the CSF, which fills the lumen of the lateral
when compared to SVZ NSCs. In pathological ventricles, through a specialized apical process
conditions, such as in multiple sclerosis, SVZ surrounded by ependymal cells (Shen et al.
NSCs can also give raise to oligodendrocyte pre- 2008; Falcão et al. 2012; Fuentealba et al.
cursor cells (OPCs), thus generating oligoden- 2012). The adult CP, present in the ventricle
drocytes (Menn et al. 2006). lumen, expresses and secretes to the CSF not
Apart from these two niches in the adult mam- only numerous trophic factors but also cytokines,
malian brain, non-typical NSC niches have also which can influence the behaviour of SVZ NSCs
been postulated where neurogenesis can occur. and NPCs, by modulating their self-renewal
7 An Overview of Adult Neurogenesis 83

Fig. 7.3 Schematic


representation of the adult
subventricular zone (SVZ)
neurogenic niche. SVZ
lines the lateral walls of the
lateral ventricles and is
comprised of three main
cell types: the multipotent
type B NSCs that give rise
to type C cells (fast dividing
transient amplifying cells)
that, in turn, generate type
A neuroblasts. Type B cells
interact basally with blood
vessels and apically with
the cerebrospinal fluid
(CSF). The composition of
the CSF is modified by the
choroid plexus, a thin
vascularized membrane
mainly composed by
epithelial cells, which
secretes several cytokines
and trophic factors to the
CSF (from Armada-
Moreira et al. 2015)

capacity, proliferation and differentiation (Falcão neuroblasts commonly migrate in a saltatory


et al. 2012; Fuentealba et al. 2012). On the basal manner which involves three steps: leading-
side, they are also in contact with blood vessels process extension; swelling formation and
through the extension of a basal process centrosomal migration; and somal translocation
(Mirzadeh et al. 2008). Type B NSCs give rise (Kaneko et al. 2017). Neuroblasts express DCX,
to intermediate progenitors (IPCs), or fast divid- as well as PSA-NCAM and N-cadherin, which
ing transient amplifying progenitors, known as enable them to migrate along one another through
type C cells (Fig. 7.4) (Doetsch et al. 1999). homophilic interaction, thus forming elongated
In rodents, type C cells mostly generate clusters called chains (Rousselot et al. 1995;
neuroblasts, or type A cells, which migrate Lois et al. 1996; Yagita et al. 2009). After a
along the rostral migratory stream (RMS) towards brain injury, apart from individual migration, sim-
the OB where they differentiate into olfactory ilar chain formations are observed for neuroblasts
interneurons and are integrated into the neuronal migrating towards injured areas (Zhang et al.
circuitry involved in olfaction (Altman 1969; 2009).
Luskin 1993; Lois and Alvarez-Buylla 1994; Externally to the chains of neuroblasts, there
Carleton et al. 2003; Belluzzi et al. 2003). are astrocytic tunnels called glial tubes that sur-
Neuroblasts migrate tangentially along the round and separate the chains of neuroblasts from
RMS forming chains surrounded by a meshwork the nearby brain parenchyma, which helps to
of specialized GFAP-positive astrocytes that form maintain the long-distance migration route
a tubular structure longitudinally linking the SVZ (Kaneko et al. 2010). This astroglial matrix in
to the OB (Lois et al. 1996; Peretto et al. 1997). the RMS is composed by specialized astrocytes
Migrating neuroblasts have a bipolar morphol- that overexpress non-soluble factors such as
ogy, displaying a long leading process and a extracellular matrix and cell adhesion molecules,
short trailing process. Individual bipolar-shaped and promote neuronal migration in a contact-
84 F. F. Ribeiro and S. Xapelli

Fig. 7.4 Adult neurogenesis from the subventricular zone (4) radial migration of immature neurons in the olfactory
of the lateral ventricle to the olfactory bulb in rodents. bulb (OB); (5) synaptic integration and maturation of
Schematic representation of adult subventricular zone granule cells and periglomerular neurons (PG) in the olfac-
(SVZ) neurogenesis in five developmental stages: (1) acti- tory bulb. Also shown is the expression of stage-specific
vation of radial glia-like cells in the SVZ in the lateral markers during SVZ-OB adult neurogenesis. GFAP, glial
ventricle (LV) and proliferation of transient amplifying fibrillary acidic protein; Dlx2, distal-less homeobox 2;
progenitors; (2) fate specification with the generation of DCX, doublecortin; PSA-NCAM, polysialylated neural
neuroblasts/type A cells; (3) chain tangential migration of cell adhesion molecule; NeuN, neuronal nuclei (from
neuroblasts within the rostral migratory stream (RMS); Ribeiro 2017)

dependent manner (García-Marqués et al. 2010). vessels have a very important function in rostral
These astrocytes also modulate neuroblast migra- migration by retaining neuroblasts in the migra-
tion by secreting or trapping soluble factors tory stream, by serving as a physical substrate
(Mason et al. 2001; Bolteus and Bordey 2004; towards the OB, or even to an injured area, as
Snapyan et al. 2009). Apart from this, blood well as by providing and regulating molecular
7 An Overview of Adult Neurogenesis 85

cues (Snapyan et al. 2009; Whitman et al. 2009; the granule cell layer to become granule cells
Grade et al. 2013). (Winner et al. 2002; Carleton et al. 2003) and
The OB is a complex and multi-layered struc- the others to the glomerular layer to become
ture. The main OB is composed of the glomerular periglomerular interneurons (Belluzzi et al.
layer, external plexiform layer, mitral cell layer, 2003). These newborn cells differentiate more
internal plexiform layer and granule cell layer slowly into periglomerular interneurons, with a
(Lazarini and Lledo 2011). Although yet not delay of more than 1 month when compared to
clear, it is thought that OB neurogenesis may be the granule cells (Winner et al. 2002).
used for plasticity, olfactory discrimination, and Neuroblasts differentiate into GABAergic
olfactory learning and memory (Lledo and Valley interneurons, such as calretinin+ or calbindin+,
2016). In the OB, there is a continuous cell turn- into dopaminergic tyrosine hydroxylase+ neurons
over, with newly generated granule cells thought (Batista-Brito et al. 2008), or into glutamatergic
to replace older ones. Importantly, cell death neurons (Brill et al. 2009). These neurons estab-
occurs through the whole SVZ-OB pathway, lish dendro-dendritic synapses with the tufted
being more marked in the OB (Biebl et al. 2000; cells (Abrous et al. 2005) and begin a maturation
Petreanu and Alvarez-Buylla 2002). process by receiving GABAergic and
Directional migration of neuroblasts towards glutamatergic synaptic inputs (Belluzzi et al.
the OB is regulated by several signalling 2003). A large amount of SVZ-derived cells that
molecules that mediate not only the interaction arrive to the OB firstly differentiate into GABA-
between type A cells, astrocytes and blood containing granule cells, followed by the forma-
vessels, but also the interaction of these cells tion of glutamatergic synaptic contacts (Carleton
with the surrounding tissues, which make possi- et al. 2003; Belluzzi et al. 2003). At earlier stages
ble this long-way migration process. Among of differentiation in OB, newborn neurons remain
them are attractive cues, such as netrin-1 and unable to fire action potentials (Carleton et al.
prokineticin 2, that are produced by the OB (Liu 2003). This delay in excitability may be important
and Rao 2003; Prosser et al. 2007); repulsive to protect circuits from uncontrolled neurotrans-
cues, like Slit proteins, that are expressed in the mitter release and neural network disruption
septum (Hu and Rutishauser 1996; Hu 1999) and (Lledo et al. 2006; Pignatelli and Belluzzi
CP (Wu et al. 1999; Nguyen-Ba-Charvet et al. 2010). The majority of cells that disperse
2004). Other signalling molecules known to con- throughout the granule cell layer develop into
trol neuroblast migration are ephrins (Conover GABAergic granule cells, while a small percent-
et al. 2000), integrins (Murase and Horwitz age that move into the periglomerular region
2002), growth factors such as glial cell line- develop mainly into interneurons of either
derived neurotrophic factor (GDNF) (Paratcha GABAergic or dopaminergic phenotypes (Kuhn
et al. 2006), brain-derived neurotrophic factor et al. 2005b).
(BDNF) (Chiaramello et al. 2007; Snapyan et al. Within the SVZ, thousands of cells are born
2009) and vascular endothelial growth factor each day (Winner et al. 2002). The majority of
(VEGF) (Wittko et al. 2009), and gamma- new generated cells, whether periglomerular or
aminobutyric acid (GABA) (Bolteus and Bordey granule cells, that reach the OB die, with around
2004; Snapyan et al. 2009). 40% of newly born cells surviving up to 19 months
Once arrived to the OB, neuroblasts detach (Petreanu and Alvarez-Buylla 2002; Winner et al.
from rostral migratory chains and radially migrate 2002). These data suggest that OB neurogenesis
into the OB, with the majority of them using promotes an overproduction and turnover of
blood vessels in a process called vasophilic neu- young neurons, rather than replacing the old
ronal migration (Bovetti et al. 2007). SVZ adult- ones. Per month, only around 6–10% of the
born cells differentiate into olfactory existing cellular population within the OB granule
interneurons, with the majority migrating towards cell layer is replaced (Winner et al. 2002). The
86 F. F. Ribeiro and S. Xapelli

survival of neurons is largely influenced by the 7.6 The Subgranular Zone (SGZ)
odour environment (Rochefort et al. 2002; Neurogenic Niche
Petreanu and Alvarez-Buylla 2002; Yamaguchi
and Mori 2005). Enriched odour exposure The other neurogenic niche in the adult mamma-
increases the number of newborn neurons in the lian brain where NSCs reside is the SGZ of the
adult OB and improves odour memory (Rochefort hippocampal DG. The DG is a V-shaped structure
et al. 2002). Therefore, the system may function that is composed of three layers (Fig. 7.5): the
by overproducing potential neurons which will be molecular layer, which is mainly occupied by the
selected through an experience-dependent dendrites of the dentate granule cells and the
process. fibres of the perforant path coming from the ento-
The route newborn SVZ neurons take in direc- rhinal cortex; the granule cell layer, which is
tion to OB is not fixed. In fact, although mainly made by densely packed granule cells
neuroblasts migrate to the OB in healthy with a 6–8 neuron-thickness and encloses the
conditions, they can also be called upon demand. third layer; and the hilus or the polymorphic cell
For instance, when a lesion occurs in the cerebral layer, which is a region composed of a variety of
cortex, corpus callosum or striatum, newborn cell types, including the mossy cells (Amaral
neuroblasts can migrate from the SVZ to the et al. 2007). The SGZ is a thin band of cells that
injured areas and form functional neurons is located at the border between the inner granule
(Bernier et al. 2002; Arvidsson et al. 2002; Parent cell layer and the hilus of the DG, which contains
et al. 2002; Bédard et al. 2006; Yamashita et al. NSCs and NPCs, as well as the bodies of
2006). Interestingly, neuroblasts migrating in an parvalbumin-expressing interneurons, i.e. the
ischemic striatum display higher exploratory GABAergic basket cells (Toni and Schinder
behaviour and longer stationary periods than 2016).
cells migrating in the RMS (Grade et al. 2013). NSCs that reside in the SGZ are called type
Some reports show that, in addition to the germi- 1 cells and are radial glial-like precursor cells
nal zone located in the walls of the lateral (Fig. 7.6). They display astrocytic electrophysio-
ventricles facing the striatum (lateral walls), logical properties and express the astrocytic
other areas also have cells that in culture possess marker GFAP, as well as other markers such as
stem cell-like properties, as it is the case of parts the intermediate filament nestin and Sox2 (Seri
of the lateral ventricular wall facing the septum, et al. 2001; Filippov et al. 2003; Fukuda et al.
as well as the whole rostral migratory stream 2003; Steiner et al. 2006). With the cell body in
(RMS) (Doetsch et al. 1999; Gritti et al. 2002). the SGZ, these early precursor cells have pro-
These stem cell-like properties were also cesses that allow them to be in permanent contact
observed in vivo (Merkle et al. 2007), showing not only with the molecular layer, through an
to possess neurogenic potential (Gritti et al. 2002; extending apical process, but also with blood
Aguirre and Gallo 2004). Moreover, several lines vessels (Filippov et al. 2003; Fukuda et al.
of evidence also suggest that the adult OB is a 2003). Type 1 cells are early progenitor cells
source of local NSCs (Pagano et al. 2000; Liu and that rarely divide. Responsive to tonic GABA
Martin 2003; Giachino and Taylor 2009; from local interneurons, these cells are
Moreno-Estellés et al. 2012). maintained in quiescence by the GABA released
Finally, a minority of type C cells enter in the from GABAergic basket cells (Song et al. 2012).
oligodendroglial lineage. They produce oligoden- When type 1 cells exit the quiescent state, they
drocyte progenitor cells (OPCs), which migrate generate intermediate progenitor cells, the type
radially out of the SVZ into the surrounding cor- 2 cells (Fig. 7.6). Unlike the former, type 2 cells
tex and white matter where they differentiate into display a high proliferative activity that allows the
oligodendrocytes (Suzuki and Goldman 2003; SGZ pool of precursor cells to expand. Moreover,
Menn et al. 2006). they have a simpler morphology, without radial
7 An Overview of Adult Neurogenesis 87

Fig. 7.5 Structure and circuits of the hippocampus. Sche- border between the inner granule cell layer and the hilus of
matic representation of a transversal slice of the hippocam- the DG is the subgranular zone (SGZ), containing NSCs
pus depicting the dentate gyrus (DG), cornus ammonis and NPCs. Granule cells are synaptically connected via the
(CA)3 and CA1. The DG is composed of three layers: so-called trisynaptic circuit. The main circuit input is the
the molecular layer, which is mainly occupied by the performant path, composed of axons coming from the
dendrites of the dentate granule cells and the fibres of the lateral and the medial entorhinal cortex. Then, granule
perforant path; the granule cell layer, made by densely neurons project mossy fibres to CA3 pyramidal neurons,
packed granule cells and the hilus, mostly composed of which then project Schaffer collaterals to CA1 pyramidal
basket and mossy cells, apart from other cell types. In the neurons (from Ribeiro 2017)
88 F. F. Ribeiro and S. Xapelli

Fig. 7.6 Cell types and the main markers expressed dur- type 3 cells to the granule cell layer; (4) axons and dendrite
ing the process of adult neurogenesis in the dentate gyrus development of immature neurons and (5) adult-born den-
of the hippocampus. Schematic representation of adult tate granule cell synaptic integration. The expressed cell
hippocampal neurogenesis in five developmental stages: stage-specific markers are also represented during adult
(1) activation of quiescent radial glia-like cell in the hippocampal neurogenesis. GFAP, glial fibrillary acidic
subgranular zone (SGZ) and proliferation of non-radial protein; DCX, doublecortin; PSA-NCAM, polysialylated
precursor and intermediate progenitors; (2) fate specifica- neural cell adhesion molecule; NeuN, neuronal nuclei
tion with the generation of neuronal precursors/type 2b (from Ribeiro 2017)
cells; (3) short-distance radial migration of neuroblasts/

but horizontally oriented short processes (Fukuda proliferative activity. They lack nestin and glial
et al. 2003). Two distinct subsets of type 2 cells markers and express neuronal lineage markers,
can be identified, the type 2a and the type 2b. including DCX and PSA-NCAM. With a great
Similarly to type 1 cells, type 2a cells still express diversity of morphologies, type 3 cells can have
glial markers and give rise to a subpopulation of either the radial or tangential process. Further-
type 2b lacking GFAP and expressing the neuro- more, these cells migrate radially short distances
nal markers DCX and PSA-NCAM (Fukuda et al. to the granule cell layer where they exit cell cycle
2003; Steiner et al. 2006). Type 3 cells are then to become granule neurons that express the
generated from type 2b cells and have little postmitotic neuronal markers neuronal nuclei
7 An Overview of Adult Neurogenesis 89

(NeuN) and the transient calcium-binding protein survives and becomes permanently integrated in
calretinin, which is later replaced by the mature the granule cell layer.
neuronal marker calbindin (Brandt et al. 2003). Adult hippocampal neurogenesis is important
Once they arrive to their destination in the granule for several hippocampal-dependent functions.
cell layer, newborn neurons continue to maturate Numerous studies over the past years have
their processes. Initially they project the axon associated hippocampal neurogenesis with con-
through the mossy fibre tract in the hilar region text learning and spatial memory, as well as emo-
to contact hippocampal CA3 and then extend tional behaviours related to stress, anxiety and
their dendrites towards the molecular layer, depression, and also with memory formation,
establishing synaptic contacts with axons from consolidation and decay, pattern separation and
the entorhinal cortex (Fig. 7.5) (Hastings and discrimination behaviours, addiction, and atten-
Gould 1999; Zhao et al. 2006; Sun et al. 2013). tion (Snyder and Drew 2020).
Hippocampal activity regulates neurogenesis
in the adult DG. Type 2 cells, as type 1 cells,
also respond to GABA. However, while GABA 7.7 Conclusion
maintains type 1 cells quiescent, type 2 cells start
to be responsive to active GABAergic excitatory Over the last years, there has been a growing
inputs from the hippocampal circuitry, which are increase in the number of reports highlighting
responsible for promoting neuronal differentia- the existence of adult neurogenesis both in animal
tion (Tozuka et al. 2005) and to promote dendritic models and in humans. In fact, adult-generated
development and synaptic integration of young neurons have important functional contributions
newborn neurons (Ge et al. 2006). In rodents, by to neural plasticity and cognition across human
the time glutamatergic synapses are formed, new lifespan. Importantly, the prospect of using NSC
neurons present a very high synaptic plasticity as regenerative therapies is very promising.
during the “critical period” of 4–6 weeks of the Therefore, the potential actions of pharmacologi-
cell age, with immature neurons displaying a cal agents such as neurotrophins used to modulate
lower threshold than mature neurons to induce NSC activity are highly relevant and will be
long-term potentiation (LTP) (Wang et al. 2000; discussed in the second part of this chapter.
Schmidt-Hieber et al. 2004; Ge et al. 2007).
These adult-born neurons might be preferentially Acknowledgements This work was supported by
recruited into hippocampal neural circuits to IF/01227/2015 project funded by Fundação para a Ciência
mediate novelty recognition, contextual fear con- e a Tecnologia (FCT). F.F.R. (IMM/CT/35-2018) received
a fellowship from FCT.
ditioning, spatial information processing and
memory formation (Gu et al. 2013). The critical
period finishes when new neurons start to be
inhibited by the surrounding interneurons, like References
mature neurons, when GABAergic contacts
Abrous DN, Koehl M, Le Moal M (2005) Adult
switch to inhibitory (Espósito at al. 2005; Ge
neurogenesis: from precursors to network and physiol-
et al. 2006). ogy. Physiol Rev 85:523–569. https://doi.org/10.1152/
During the postmitotic phase, there is a dra- physrev.00055.2003
matic decrease in the number of new neurons, Aguirre A, Gallo V (2004) Postnatal neurogenesis and
gliogenesis in the olfactory bulb from
through apoptosis (Biebl et al. 2000; Kuhn et al.
NG2-expressing progenitors of the subventricular
2005a). In fact, most of the cells are eliminated zone. J Neurosci 24:10530–10541. https://doi.org/10.
before establishing synaptic contacts with the tar- 1523/JNEUROSCI.3572-04.2004
get area in CA3 or receiving correct dendritic Altman J (1962) Are new neurons formed in the brains of
adult mammals? Science 135:1127–1128
input from the entorhinal cortex in the molecular
Altman J (1963) Autoradiographic investigation of cell
layer of the DG. From the 10,000 cells that are proliferation in the brains of rats and cats. Anat Rec
produced every day, just a small percentage 145:573–591
90 F. F. Ribeiro and S. Xapelli

Altman J (1969) Autoradiographic and histological studies Biebl M, Cooper CM, Winkler J, Kuhn HG (2000) Analy-
of postnatal neurogenesis. IV. Cell proliferation and sis of neurogenesis and programmed cell death reveals
migration in the anterior forebrain, with special refer- a self-renewing capacity in the adult rat brain. Neurosci
ence to persisting neurogenesis in the olfactory bulb. J Lett 291:17–20
Comp Neurol 137:433–457. https://doi.org/10.1002/ Boldrini M, Fulmore CA, Tartt AN, et al (2018) Human
cne.901370404 hippocampal neurogenesis persists throughout aging.
Altman J, Das GD (1965) Autoradiographic and histologi- Cell Stem Cell 22:589–599.e5. https://doi.org/10.
cal evidence of postnatal hippocampal neurogenesis in 1016/j.stem.2018.03.015
rats. J Comp Neurol 124:319–335 Bolteus AJ, Bordey A (2004) GABA release and uptake
Altman J, Das GD (1966) Autoradiographic and histologi- regulate neuronal precursor migration in the postnatal
cal studies of postnatal neurogenesis. I. A longitudinal subventricular zone. J Neurosci 24:7623–7631. https://
investigation of the kinetics, migration and transforma- doi.org/10.1523/JNEUROSCI.1999-04.2004
tion of cells incorporating tritiated thymidine in neo- Bonaguidi MA, Wheeler MA, Shapiro JS et al (2011) In
nate rats, with special reference to postnatal vivo clonal analysis reveals self-renewing and
neurogenesis in some brain regions. J Comp Neurol multipotent adult neural stem cell characteristics. Cell
126:337–389. https://doi.org/10.1002/cne.901260302 145:1142–1155. https://doi.org/10.1016/j.cell.2011.
Altman J, Das GD (1967) Postnatal neurogenesis in the 05.024
guinea-pig. Nature 214:1098–1101 Bovetti S, Hsieh Y-C, Bovolin P et al (2007) Blood vessels
Alvarez-Buylla A, Lim DA (2004) For the long run: form a scaffold for neuroblast migration in the adult
maintaining germinal niches in the adult brain. Neuron olfactory bulb. J Neurosci 27:5976–5980. https://doi.
41:683–686 org/10.1523/JNEUROSCI.0678-07.2007
Amaral DG, Scharfman HE, Lavenex P (2007) The den- Brandt MD, Jessberger S, Steiner B et al (2003) Transient
tate gyrus: fundamental neuroanatomical organization calretinin expression defines early postmitotic step of
(dentate gyrus for dummies). Prog Brain Res neuronal differentiation in adult hippocampal
163:3–22. https://doi.org/10.1016/S0079-6123(07) neurogenesis of mice. Mol Cell Neurosci 24:603–613
63001-5 Breunig JJ, Arellano JI, Macklis JD, Rakic P (2007)
Armada-Moreira A, Ribeiro F, Sebastião A, Xapelli S Everything that glitters isn’t gold: a critical review of
(2015) Neuroinflammatory modulators of oligoden- postnatal neural precursor analyses. Cell Stem Cell
drogenesis. Neuroimmunol Neuroinflamm 2:263. 1:612–627. https://doi.org/10.1016/j.stem.2007.11.
https://doi.org/10.4103/2347-8659.167311 008
Arvidsson A, Collin T, Kirik D et al (2002) Neuronal Brill MS, Ninkovic J, Winpenny E et al (2009) Adult
replacement from endogenous precursors in the adult generation of glutamatergic olfactory bulb
brain after stroke. Nat Med 8:963–970. https://doi.org/ interneurons. Nat Neurosci 12:1524–1533. https://doi.
10.1038/nm747 org/10.1038/nn.2416
Batista-Brito R, Close J, Machold R, Fishell G (2008) The Bull ND, Bartlett PF (2005) The adult mouse hippocampal
distinct temporal origins of olfactory bulb interneuron progenitor is neurogenic but not a stem cell. J Neurosci
subtypes. J Neurosci 28:3966–3975. https://doi.org/10. 25:10815–10821. https://doi.org/10.1523/
1523/JNEUROSCI.5625-07.2008 JNEUROSCI.3249-05.2005
Bédard A, Gravel C, Parent A (2006) Chemical character- Burd GD, Nottebohm F (1985) Ultrastructural characteri-
ization of newly generated neurons in the striatum of zation of synaptic terminals formed on newly
adult primates. Exp Brain Res 170:501–512. https:// generated neurons in a song control nucleus of the
doi.org/10.1007/s00221-005-0233-5 adult canary forebrain. J Comp Neurol 240:143–152.
Belluzzi O, Benedusi M, Ackman J, LoTurco JJ (2003) https://doi.org/10.1002/cne.902400204
Electrophysiological differentiation of new neurons in Calzolari F, Michel J, Baumgart EV et al (2015) Fast
the olfactory bulb. J Neurosci 23:10411–10418 clonal expansion and limited neural stem cell self-
Bergmann O, Liebl J, Bernard S et al (2012) The age of renewal in the adult subependymal zone. Nat Neurosci
olfactory bulb neurons in humans. Neuron 18:490–492. https://doi.org/10.1038/nn.3963
74:634–639. https://doi.org/10.1016/j.neuron.2012. Carleton A, Petreanu LT, Lansford R et al (2003) Becom-
03.030 ing a new neuron in the adult olfactory bulb. Nat
Bernier PJ, Bedard A, Vinet J et al (2002) Newly Neurosci 6:507–518. https://doi.org/10.1038/nn1048
generated neurons in the amygdala and adjoining cor- Chiaramello S, Dalmasso G, Bezin L et al (2007) BDNF/
tex of adult primates. Proc Natl Acad Sci USA TrkB interaction regulates migration of SVZ precursor
99:11464–11469. https://doi.org/10.1073/pnas. cells via PI3-K and MAP-K signalling pathways. Eur J
172403999 Neurosci 26:1780–1790. https://doi.org/10.1111/j.
Bhardwaj RD, Curtis MA, Spalding KL et al (2006) Neo- 1460-9568.2007.05818.x
cortical neurogenesis in humans is restricted to devel- Conover JC, Doetsch F, Garcia-Verdugo JM et al (2000)
opment. Proc Natl Acad Sci USA 103:12564–12568. Disruption of Eph/ephrin signaling affects migration
https://doi.org/10.1073/pnas.0605177103 and proliferation in the adult subventricular zone. Nat
Neurosci 3:1091–1097. https://doi.org/10.1038/80606
7 An Overview of Adult Neurogenesis 91

Dayer AG, Cleaver KM, Abouantoun T, Cameron HA Ge S, Yang C-H, Hsu K-S et al (2007) A critical period for
(2005) New GABAergic interneurons in the adult neo- enhanced synaptic plasticity in newly generated
cortex and striatum are generated from different neurons of the adult brain. Neuron 54:559–566.
precursors. J Cell Biol 168:415–427. https://doi.org/ https://doi.org/10.1016/j.neuron.2007.05.002
10.1083/jcb.200407053 Ghosh HS (2019) Adult neurogenesis and the promise of
Dennis CV, Suh LS, Rodriguez ML et al (2016) Human adult neural stem cells. J Exp Neurosci 13. https://doi.
adult neurogenesis across the ages: an immunohisto- org/10.1177/1179069519856876
chemical study. Neuropathol Appl Neurobiol Giachino C, Taylor V (2009) Lineage analysis of quiescent
42:621–638. https://doi.org/10.1111/nan.12337 regenerative stem cells in the adult brain by genetic
Doetsch F, García-Verdugo JM, Alvarez-Buylla A (1997) labelling reveals spatially restricted neurogenic niches
Cellular composition and three-dimensional organiza- in the olfactory bulb. Eur J Neurosci 30:9–24. https://
tion of the subventricular germinal zone in the adult doi.org/10.1111/j.1460-9568.2009.06798.x
mammalian brain. J Neurosci 17:5046–5061 Goldman SA, Nottebohm F (1983) Neuronal production,
Doetsch F, Caillé I, Lim DA et al (1999) Subventricular migration, and differentiation in a vocal control
zone astrocytes are neural stem cells in the adult mam- nucleus of the adult female canary brain. Proc Natl
malian brain. Cell 97:703–716 Acad Sci USA 80:2390–2394
Encinas JM, Michurina TV, Peunova N et al (2011) Götz M, Sirko S, Beckers J, Irmler M (2015) Reactive
Division-coupled astrocytic differentiation and astrocytes as neural stem or progenitor cells: in vivo
age-related depletion of neural stem cells in the adult lineage, In vitro potential, and genome-wide expres-
hippocampus. Cell Stem Cell 8:566–579. https://doi. sion analysis. Glia 63:1452–1468. https://doi.org/10.
org/10.1016/j.stem.2011.03.010 1002/glia.22850
Eriksson PS, Perfilieva E, Björk-Eriksson T et al (1998) Gould E, Tanapat P, McEwen BS et al (1998) Proliferation
Neurogenesis in the adult human hippocampus. Nat of granule cell precursors in the dentate gyrus of adult
Med 4:1313–1317. https://doi.org/10.1038/3305 monkeys is diminished by stress. Proc Natl Acad Sci
Ernst A, Alkass K, Bernard S et al (2014) Neurogenesis in USA 95:3168–3171
the striatum of the adult human brain. Cell Gould E, Reeves AJ, Fallah M et al (1999a) Hippocampal
156:1072–1083. https://doi.org/10.1016/j.cell.2014. neurogenesis in adult Old World primates. Proc Natl
01.044 Acad Sci USA 96:5263–5267
Falcão AM, Marques F, Novais A et al (2012) The path Gould E, Reeves AJ, Graziano MS, Gross CG (1999b)
from the choroid plexus to the subventricular zone: go Neurogenesis in the neocortex of adult primates. Sci-
with the flow! Front Cell Neurosci 6:34. https://doi. ence 286:548–552
org/10.3389/fncel.2012.00034 Grade S, Weng YC, Snapyan M et al (2013) Brain-derived
Filippov V, Kronenberg G, Pivneva T et al (2003) Sub- neurotrophic factor promotes vasculature-associated
population of nestin-expressing progenitor cells in the migration of neuronal precursors toward the ischemic
adult murine hippocampus shows electrophysiological striatum. PLoS One 8:e55039. https://doi.org/10.1371/
and morphological characteristics of astrocytes. Mol journal.pone.0055039
Cell Neurosci 23:373–382 Gritti A, Bonfanti L, Doetsch F et al (2002) Multipotent
Fuentealba LC, Obernier K, Alvarez-Buylla A (2012) neural stem cells reside into the rostral extension and
Adult neural stem cells bridge their niche. Cell Stem olfactory bulb of adult rodents. J Neurosci 22:437–445
Cell 10:698–708. https://doi.org/10.1016/j.stem.2012. Gu Y, Janoschka S, Ge S (2013) Neurogenesis and hippo-
05.012 campal plasticity in adult brain. Curr Top Behav
Fukuda S, Kato F, Tozuka Y et al (2003) Two distinct Neurosci 15:31–48. https://doi.org/10.1007/7854_
subpopulations of nestin-positive cells in adult mouse 2012_217
dentate gyrus. J Neurosci 23:9357–9366 Hall PA, Watt FM (1989) Stem cells: the generation and
Gage FH (2000) Mammalian neural stem cells. Science maintenance of cellular diversity. Development
287:1433–1438 106:619–633
Gage FH, Kempermann G, Palmer TD et al (1998) Hastings NB, Gould E (1999) Rapid extension of axons
Multipotent progenitor cells in the adult dentate into the CA3 region by adult-generated granule cells. J
gyrus. J Neurobiol 36:249–266 Comp Neurol 413:146–154
García-Marqués J, Carlos JAD, Greer CA, López- Hu H (1999) Chemorepulsion of neuronal migration by
Mascaraque L (2010) Different astroglia permissivity Slit2 in the developing mammalian forebrain. Neuron
controls the migration of olfactory bulb interneuron 23:703–711
precursors. Glia 58:218–230. https://doi.org/10.1002/ Hu H, Rutishauser U (1996) A septum-derived
glia.20918 chemorepulsive factor for migrating olfactory interneu-
Ge S, Goh ELK, Sailor KA et al (2006) GABA regulates ron precursors. Neuron 16:933–940
synaptic integration of newly generated neurons in the Inta D, Cameron HA, Gass P (2015) New neurons in the
adult brain. Nature 439:589–593. https://doi.org/10. adult striatum: from rodents to humans. Trends
1038/nature04404 Neurosci 38:517–523. https://doi.org/10.1016/j.tins.
2015.07.005
92 F. F. Ribeiro and S. Xapelli

Kaneko N, Marín O, Koike M et al (2010) New neurons continued but minimal neurogenesis in the adult
clear the path of astrocytic processes for their rapid human hippocampus. bioRxiv. https://doi.org/10.
migration in the adult brain. Neuron 67:213–223. 1101/664995
https://doi.org/10.1016/j.neuron.2010.06.018 Lazarini F, Lledo P-M (2011) Is adult neurogenesis essen-
Kaneko N, Sawada M, Sawamoto K (2017) Mechanisms tial for olfaction? Trends Neurosci 34:20–30. https://
of neuronal migration in the adult brain. J Neurochem doi.org/10.1016/j.tins.2010.09.006
141:835–847. https://doi.org/10.1111/jnc.14002 Lie DC, Dziewczapolski G, Willhoite AR et al (2002) The
Kaplan MS (1981) Neurogenesis in the 3-month-old rat adult substantia nigra contains progenitor cells with
visual cortex. J Comp Neurol 195:323–338. https://doi. neurogenic potential. J Neurosci 22:6639–6649.
org/10.1002/cne.901950211 https://doi.org/10.1523/JNEUROSCI.22-15-06639.
Kaplan MS (1983) Proliferation of subependymal cells in 2002
the adult primate CNS: differential uptake of DNA Liu Z, Martin LJ (2003) Olfactory bulb core is a rich
labelled precursors. J Hirnforsch 24:23–33 source of neural progenitor and stem cells in adult
Kaplan MS, Bell DH (1984) Mitotic neuroblasts in the rodent and human. J Comp Neurol 459:368–391.
9-day-old and 11-month-old rodent hippocampus. J https://doi.org/10.1002/cne.10664
Neurosci 4:1429–1441 Liu G, Rao Y (2003) Neuronal migration from the fore-
Kaplan MS, Hinds JW (1977) Neurogenesis in the adult brain to the olfactory bulb requires a new attractant
rat: electron microscopic analysis of light persistent in the olfactory bulb. J Neurosci
radioautographs. Science 197:1092–1094 23:6651–6659
Katsimpardi L, Litterman NK, Schein PA et al (2014) Lledo P-M, Valley M (2016) Adult olfactory bulb
Vascular and neurogenic rejuvenation of the aging neurogenesis. Cold Spring Harb Perspect Biol 8:
mouse brain by young systemic factors. Science a018945. https://doi.org/10.1101/cshperspect.a018945
344:630–634. https://doi.org/10.1126/science. Lledo P-M, Alonso M, Grubb MS (2006) Adult
1251141 neurogenesis and functional plasticity in neuronal
Kempermann G (2014) Off the beaten track: new neurons circuits. Nat Rev Neurosci 7:179–193. https://doi.org/
in the adult human striatum. Cell 156:870–871. https:// 10.1038/nrn1867
doi.org/10.1016/j.cell.2014.02.027 Lois C, Alvarez-Buylla A (1994) Long-distance neuronal
Kempermann G, Gage FH (1999) New nerve cells for the migration in the adult mammalian brain. Science
adult brain. Sci Am 280:48–53 264:1145–1148
Kilpatrick TJ, Bartlett PF (1993) Cloning and growth of Lois C, García-Verdugo JM, Alvarez-Buylla A (1996)
multipotential neural precursors: requirements for pro- Chain migration of neuronal precursors. Science
liferation and differentiation. Neuron 10:255–265 271:978–981
Kokoeva MV, Yin H, Flier JS (2005) Neurogenesis in the Luskin MB (1993) Restricted proliferation and migration
hypothalamus of adult mice: potential role in energy of postnatally generated neurons derived from the fore-
balance. Science 310:679–683. https://doi.org/10. brain subventricular zone. Neuron 11:173–189
1126/science.1115360 Ma DK, Ming G-L, Song H (2005) Glial influences on
Kokoeva MV, Yin H, Flier JS (2007) Evidence for consti- neural stem cell development: cellular niches for adult
tutive neural cell proliferation in the adult murine neurogenesis. Curr Opin Neurobiol 15:514–520.
hypothalamus. J Comp Neurol 505:209–220. https:// https://doi.org/10.1016/j.conb.2005.08.003
doi.org/10.1002/cne.21492 Mason HA, Ito S, Corfas G (2001) Extracellular signals
Kornack DR, Rakic P (1999) Continuation of that regulate the tangential migration of olfactory bulb
neurogenesis in the hippocampus of the adult macaque neuronal precursors: inducers, inhibitors, and
monkey. Proc Natl Acad Sci USA 96:5768–5773 repellents. J Neurosci 21:7654–7663
Kuhn HG, Dickinson-Anson H, Gage FH (1996) Menn B, Garcia-Verdugo JM, Yaschine C et al (2006)
Neurogenesis in the dentate gyrus of the adult rat: Origin of oligodendrocytes in the subventricular zone
age-related decrease of neuronal progenitor prolifera- of the adult brain. J Neurosci 26:7907–7918. https://
tion. J Neurosci 16:2027–2033 doi.org/10.1523/JNEUROSCI.1299-06.2006
Kuhn HG, Biebl M, Wilhelm D et al (2005a) Increased Merkle FT, Alvarez-Buylla A (2006) Neural stem cells in
generation of granule cells in adult Bcl-2- mammalian development. Curr Opin Cell Biol
overexpressing mice: a role for cell death during 18:704–709. https://doi.org/10.1016/j.ceb.2006.09.
continued hippocampal neurogenesis. Eur J Neurosci 008
22:1907–1915. https://doi.org/10.1111/j.1460-9568. Merkle FT, Mirzadeh Z, Alvarez-Buylla A (2007) Mosaic
2005.04377.x organization of neural stem cells in the adult brain.
Kuhn HG, Cooper-Kuhn C, Eriksson P, Nilsson M Science 317:381–384. https://doi.org/10.1126/sci
(2005b) Signals regulating neurogenesis in the adult ence.1144914
olfactory bulb. Chem Senses 30:i109–i110. https://doi. Mirzadeh Z, Merkle FT, Soriano-Navarro M et al (2008)
org/10.1093/chemse/bjh138 Neural stem cells confer unique pinwheel architecture
Kumar A, Pareek V, Faiq MA et al (2019) Transcriptomic to the ventricular surface in neurogenic regions of the
analysis of the neurogenesis signature suggests
7 An Overview of Adult Neurogenesis 93

adult brain. Cell Stem Cell 3:265–278. https://doi.org/ the rostral migratory stream. Mol Cell Neurosci
10.1016/j.stem.2008.07.004 31:505–514. https://doi.org/10.1016/j.mcn.2005.11.
Mizrak D, Levitin HM, Delgado AC et al (2019) Single- 007
cell analysis of regional differences in adult V-SVZ Parent JM, Vexler ZS, Gong C et al (2002) Rat forebrain
neural stem cell lineages. Cell Rep 26:394–406.e5. neurogenesis and striatal neuron replacement after
https://doi.org/10.1016/j.celrep.2018.12.044 focal stroke. Ann Neurol 52:802–813. https://doi.org/
Moreno-Estellés M, González-Gómez P, Hortigüela R 10.1002/ana.10393
et al (2012) Symmetric expansion of neural stem cells Paton JA, Nottebohm FN (1984) Neurons generated in the
from the adult olfactory bulb is driven by astrocytes via adult brain are recruited into functional circuits. Sci-
WNT7A. Stem Cells 30:2796–2809. https://doi.org/ ence 225:1046–1048
10.1002/stem.1243 Peretto P, Merighi A, Fasolo A, Bonfanti L (1997) Glial
Moreno-Jiménez EP, Flor-García M, Terreros-Roncal J tubes in the rostral migratory stream of the adult rat.
et al (2019) Adult hippocampal neurogenesis is abun- Brain Res Bull 42:9–21
dant in neurologically healthy subjects and drops Petreanu L, Alvarez-Buylla A (2002) Maturation and
sharply in patients with Alzheimer’s disease. Nat death of adult-born olfactory bulb granule neurons:
Med 25:554–560. https://doi.org/10.1038/s41591- role of olfaction. J Neurosci 22:6106–6113
019-0375-9 Pignatelli A, Belluzzi O (2010) Neurogenesis in the adult
Morshead CM, Reynolds BA, Craig CG et al (1994) olfactory bulb. In: Menini A (ed) The neurobiology of
Neural stem cells in the adult mammalian forebrain: a olfaction. CRC/Taylor & Francis, Boca Raton, FL
relatively quiescent subpopulation of subependymal Prosser HM, Bradley A, Caldwell MA (2007) Olfactory
cells. Neuron 13:1071–1082 bulb hypoplasia in Prokr2 null mice stems from defec-
Murase S, Horwitz AF (2002) Deleted in colorectal carci- tive neuronal progenitor migration and differentiation.
noma and differentially expressed integrins mediate the Eur J Neurosci 26:3339–3344. https://doi.org/10.1111/
directional migration of neural precursors in the rostral j.1460-9568.2007.05958.x
migratory stream. J Neurosci 22:3568–3579. https:// Ramón y Cajal S, May RM (1928) Degeneration & regen-
doi.org/10.1523/JNEUROSCI.22-09-03568.2002 eration of the nervous system. Oxford University
Nam H, Benezra R (2009) High levels of Id1 expression Press, Humphrey Milford, London
define B1 type adult neural stem cells. Cell Stem Cell Reynolds BA, Weiss S (1992) Generation of neurons and
5:515–526. https://doi.org/10.1016/j.stem.2009.08. astrocytes from isolated cells of the adult mammalian
017 central nervous system. Science 255:1707–1710
Nguyen-Ba-Charvet KT, Picard-Riera N, Tessier-Lavigne Ribeiro FF (2017) Unveiling the trophic actions of adeno-
M et al (2004) Multiple roles for slits in the control of sine A2A receptors in neurite outgrowth and postnatal
cell migration in the rostral migratory stream. J neurogenesis: interaction with brain-derived
Neurosci 24:1497–1506. https://doi.org/10.1523/ neurotrophic factor. PhD thesis. Universidade de
JNEUROSCI.4729-03.2004 Lisboa, Lisbon, Portugal
Nottebohm F (1985) Neuronal replacement in adulthood. Rochefort C, Gheusi G, Vincent J-D, Lledo P-M (2002)
Ann N Y Acad Sci 457:143–161 Enriched odor exposure increases the number of new-
Obernier K, Cebrian-Silla A, Thomson M, et al (2018) born neurons in the adult olfactory bulb and improves
Adult neurogenesis is sustained by symmetric self- odor memory. J Neurosci 22:2679–2689. https://doi.
renewal and differentiation. Cell Stem Cell org/10.1523/JNEUROSCI.22-07-02679.2002
22:221–234.e8. https://doi.org/10.1016/j.stem.2018. Rousselot P, Lois C, Alvarez-Buylla A (1995) Embryonic
01.003 (PSA) N-CAM reveals chains of migrating neuroblasts
Ohira K, Furuta T, Hioki H et al (2010) Ischemia-induced between the lateral ventricle and the olfactory bulb of
neurogenesis of neocortical layer 1 progenitor cells. adult mice. J Comp Neurol 351:51–61. https://doi.org/
Nat Neurosci 13:173–179. https://doi.org/10.1038/nn. 10.1002/cne.903510106
2473 Schmidt-Hieber C, Jonas P, Bischofberger J (2004)
Pagano SF, Impagnatiello F, Girelli M et al (2000) Isola- Enhanced synaptic plasticity in newly generated gran-
tion and characterization of neural stem cells from the ule cells of the adult hippocampus. Nature
adult human olfactory bulb. Stem Cells 18:295–300. 429:184–187. https://doi.org/10.1038/nature02553
https://doi.org/10.1634/stemcells.18-4-295 Seaberg RM, van der Kooy D (2002) Adult rodent neuro-
Palmer TD, Takahashi J, Gage FH (1997) The adult rat genic regions: the ventricular subependyma contains
hippocampus contains primordial neural stem cells. neural stem cells, but the dentate gyrus contains
Mol Cell Neurosci 8:389–404. https://doi.org/10. restricted progenitors. J Neurosci 22:1784–1793
1006/mcne.1996.0595 Seri B, García-Verdugo JM, McEwen BS, Alvarez-Buylla
Palmer TD, Willhoite AR, Gage FH (2000) Vascular niche A (2001) Astrocytes give rise to new neurons in the
for adult hippocampal neurogenesis. J Comp Neurol adult mammalian hippocampus. J Neurosci
425:479–494 21:7153–7160
Paratcha G, Ibáñez CF, Ledda F (2006) GDNF is a Shen Q, Wang Y, Kokovay E et al (2008) Adult SVZ stem
chemoattractant factor for neuronal precursor cells in cells lie in a vascular niche: a quantitative analysis of
94 F. F. Ribeiro and S. Xapelli

niche cell-cell interactions. Cell Stem Cell 3:289–300. Wang S, Scott BW, Wojtowicz JM (2000) Heterogenous
https://doi.org/10.1016/j.stem.2008.07.026 properties of dentate granule neurons in the adult rat. J
Snapyan M, Lemasson M, Brill MS et al (2009) Vascula- Neurobiol 42:248–257
ture guides migrating neuronal precursors in the adult Whitman MC, Fan W, Rela L et al (2009) Blood vessels
mammalian forebrain via brain-derived neurotrophic form a migratory scaffold in the rostral migratory
factor signaling. J Neurosci 29:4172–4188. https:// stream. J Comp Neurol 516:94–104. https://doi.org/
doi.org/10.1523/JNEUROSCI.4956-08.2009 10.1002/cne.22093
Snyder JS, Drew MR (2020) Functional neurogenesis over Winner B, Cooper-Kuhn CM, Aigner R et al (2002) Long-
the years. Behav Brain Res 382:112470. https://doi. term survival and cell death of newly generated
org/10.1016/j.bbr.2020.112470 neurons in the adult rat olfactory bulb. Eur J Neurosci
Song J, Zhong C, Bonaguidi MA et al (2012) Neuronal 16:1681–1689. https://doi.org/10.1046/j.1460-9568.
circuitry mechanism regulating adult quiescent neural 2002.02238.x
stem-cell fate decision. Nature 489:150–154. https:// Wittko IM, Schänzer A, Kuzmichev A et al (2009)
doi.org/10.1038/nature11306 VEGFR-1 regulates adult olfactory bulb neurogenesis
Sorrells SF, Paredes MF, Cebrian-Silla A et al (2018) and migration of neural progenitors in the rostral
Human hippocampal neurogenesis drops sharply in migratory stream in vivo. J Neurosci 29:8704–8714.
children to undetectable levels in adults. Nature https://doi.org/10.1523/JNEUROSCI.5527-08.2009
555:377–381. https://doi.org/10.1038/nature25975 Wu W, Wong K, Chen J et al (1999) Directional guidance
Spalding KL, Bergmann O, Alkass K et al (2013) Dynam- of neuronal migration in the olfactory system by the
ics of hippocampal neurogenesis in adult humans. Cell protein Slit. Nature 400:331–336. https://doi.org/10.
153:1219–1227. https://doi.org/10.1016/j.cell.2013. 1038/22477
05.002 Yagita Y, Sakurai T, Tanaka H et al (2009) N-cadherin
Steiner B, Klempin F, Wang L et al (2006) Type-2 cells as mediates interaction between precursor cells in the
link between glial and neuronal lineage in adult hippo- subventricular zone and regulates further differentia-
campal neurogenesis. Glia 54:805–814. https://doi.org/ tion. J Neurosci Res 87:3331–3342. https://doi.org/10.
10.1002/glia.20407 1002/jnr.22044
Sun GJ, Sailor KA, Mahmood QA et al (2013) Seamless Yamaguchi M, Mori K (2005) Critical period for sensory
reconstruction of intact adult-born neurons by serial experience-dependent survival of newly generated
end-block imaging reveals complex axonal guidance granule cells in the adult mouse olfactory bulb. Proc
and development in the adult hippocampus. J Neurosci Natl Acad Sci USA 102:9697–9702. https://doi.org/10.
33:11400–11411. https://doi.org/10.1523/ 1073/pnas.0406082102
JNEUROSCI.1374-13.2013 Yamashita T, Ninomiya M, Acosta PH et al (2006)
Suzuki SO, Goldman JE (2003) Multiple cell populations Subventricular zone-derived neuroblasts migrate and
in the early postnatal subventricular zone take distinct differentiate into mature neurons in the post-stroke
migratory pathways: a dynamic study of glial and adult striatum. J Neurosci 26:6627–6636. https://doi.
neuronal progenitor migration. J Neurosci org/10.1523/JNEUROSCI.0149-06.2006
23:4240–4250 Zhang RL, Chopp M, Gregg SR et al (2009) Patterns and
Temple S (2001) The development of neural stem cells. dynamics of subventricular zone neuroblast migration
Nature 414:112–117. https://doi.org/10.1038/ in the ischemic striatum of the adult mouse. J Cereb
35102174 Blood Flow Metab 29:1240–1250. https://doi.org/10.
Tobin MK, Musaraca K, Disouky A et al (2019) Human 1038/jcbfm.2009.55
hippocampal neurogenesis persists in aged adults and Zhao M, Janson Lang AM (2009) Bromodeoxyuridine
Alzheimer’s disease patients. Cell Stem Cell infused into the cerebral ventricle of adult mice labels
24:974–982.e3. https://doi.org/10.1016/j.stem.2019. nigral neurons under physiological conditions--a
05.003 method to detect newborn nerve cells in regions with
Toni N, Schinder AF (2016) Maturation and functional a low rate of neurogenesis. J Neurosci Methods
integration of new granule cells into the adult hippo- 184:327–331. https://doi.org/10.1016/j.jneumeth.
campus. Cold Spring Harb Perspect Biol 8:a018903. 2009.08.007
https://doi.org/10.1101/cshperspect.a018903 Zhao M, Momma S, Delfani K et al (2003) Evidence for
Tozuka Y, Fukuda S, Namba T et al (2005) GABAergic neurogenesis in the adult mammalian substantia nigra.
excitation promotes neuronal differentiation in adult Proc Natl Acad Sci USA 100:7925–7930. https://doi.
hippocampal progenitor cells. Neuron 47:803–815. org/10.1073/pnas.1131955100
https://doi.org/10.1016/j.neuron.2005.08.023 Zhao C, Teng EM, Summers RG et al (2006) Distinct
Villeda SA, Luo J, Mosher KI et al (2011) The ageing morphological stages of dentate granule neuron matu-
systemic milieu negatively regulates neurogenesis and ration in the adult mouse hippocampus. J Neurosci
cognitive function. Nature 477:90–94. https://doi.org/ 26:3–11. https://doi.org/10.1523/JNEUROSCI.3648-
10.1038/nature10357 05.2006
Intervention of Brain-Derived
Neurotrophic Factor and Other 8
Neurotrophins in Adult Neurogenesis

Filipa F. Ribeiro and Sara Xapelli

Abstract the most studied neurotrophin. In this chapter,


Cell survival during adult neurogenesis and we review the current knowledge about the
the modulation of each step, namely, prolifer- role of neurotrophins in the regulation of
ation, lineage differentiation, migration, matu- adult neurogenesis in both the subventricular
ration, and functional integration of the zone (SVZ) and the hippocampal subgranular
newborn cells into the existing circuitry, is zone (SGZ).
regulated by intrinsic and extrinsic factors.
Transduction of extracellular niche signals
triggers the activation of intracellular
mechanisms that regulate adult neurogenesis 8.1 Neurotrophins
by affecting gene expression. While the intrin-
sic factors include transcription factors and Neurotrophins are pleiotropic molecules that not
epigenetic regulators, the extrinsic factors are only play important roles in regulating cell sur-
molecular signals that are present in the neuro- vival but also exert a wide variety of other
genic niche microenvironment. These include biological functions, including the regulation of
morphogens, growth factors, neuronal differentiation, axonal and dendritic
neurotransmitters, and signaling molecules growth, and synaptic transmission and plasticity.
secreted as soluble factors or associated to Hence, neurotrophins are essential for the devel-
the extracellular matrix. Among these molecu- opment of the architecture of the embryonic
lar mechanisms are neurotrophins and brain, and also for its maintenance during adult-
neurotrophin receptors which have been hood, having important roles in learning and
implicated in the regulation of adult memory processes (Bothwell 2014). Among the
neurogenesis at different levels, with brain- family of neurotrophins, BDNF has been the most
derived neurotrophic factor (BDNF) being studied neurotrophin in the regulation of postnatal
neurogenesis, with NGF, neurotrophin-3 (NT-3),
and neurotrophin-4 (NT-4) being less studied
F. F. Ribeiro · S. Xapelli (*)
(Huang and Reichardt 2001; Chao 2003; Vilar
Instituto de Farmacologia e Neurociências, Faculdade de
Medicina, Universidade de Lisboa, Lisbon, Portugal and Mira 2016).
All neurotrophins are initially synthesized as
Instituto de Medicina Molecular João Lobo Antunes,
Faculdade de Medicina, Universidade de Lisboa, Lisbon, precursors or proneurotrophins containing signal
Portugal peptides followed by the pro-region at the
e-mail: anaribeiro@medicina.ulisboa.pt; N-terminal. Proneurotrophins may be secreted
sxapelli@medicina.ulisboa.pt

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 95


L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_8
96 F. F. Ribeiro and S. Xapelli

from cells or undergo intracellular proteolytic other neurotrophins, BDNF is not translated from
cleavage to yield mature neurotrophins, which the mRNA as a mature protein, but it is initially
dimerize after translation (Kolbeck et al. 1994; synthesized as a precursor protein, the pre-pro-
Foltran and Diaz 2016). Proneurotrophins and BDNF. The presence of a signal peptide directs
neurotrophins have distinct biological functions. this pre-pro-neurotrophin to the endoplasmic
reticulum where the signal peptide is cleaved to
form a 32 kDa pro-BDNF (Halban and Irminger
8.2 Brain-Derived Neurotrophic 1994). Pro-BDNF is then transported to the Golgi
Factor (BDNF) for sorting into two different secretory pathways
(Fig. 8.1): the constitutive pathway, which
BDNF, a small basic protein with 252 amino acid comprises small vesicles that are transported to
residues, is abundantly expressed in the nervous the cell periphery to fuse with the plasma mem-
system (Katoh-Semba et al. 1997). BDNF synthe- brane, in an intracellular Ca2+-independent man-
sis is highly regulated in space and time at several ner; and the regulated pathway, which comprises
levels. The BDNF gene comprises one common large vesicles that fuse with the plasma membrane
encoding region and multiple promoters that reg- in a Ca2+-dependent manner (Lessmann et al.
ulate the transcription of the BDNF gene. In 2003). While the constitutive secretory pathway
rodents, the main encoding region is spliced to for BDNF secretion occurs in both non-neuronal
one out of eleven upstream different 50 UTR alter- and neuronal cells, the regulated secretory path-
native exons. In addition, BDNF transcripts can way exclusively occurs in neurons (Seidah et al.
be subjected to polyadenylation in two different 1996; Mowla et al. 2001). Through these pro-
sites, generating two different transcripts, one cesses, pro-BDNF is secreted to the extracellular
with a long and other with a short 30 UTR space. Either pro-BDNF can be simply secreted
(untranslated region), raising the resultant without further digestion, thus serving as an
transcripts to a total of 22 variants (Aid et al. endogenous ligand by itself, or pro-BDNF can
2007; Pruunsild et al. 2007). This structural dif- be secreted and extracellularly processed to
ference determines the subcellular localization of mature BDNF by the endopeptidases tissue plas-
the BDNF mRNAs: BDNF mRNAs with a short minogen activator (tPA)/plasmin system (Pang
30 UTR are restricted to the soma, where they are et al. 2004) and matrix-metalloproteinases (Lee
constitutively translated to maintain the basal et al. 2001; Hwang et al. 2005; Mizoguchi et al.
levels of BDNF protein, whilst long 30 UTR 2011).
mRNAs are targeted to dendritic spines, where Alternatively, pro-BDNF can suffer intracellu-
they are locally translated in response to neuronal larly proteolytic cleavage in the trans-Golgi
activation (An et al. 2008; Lau et al. 2010). There through the action of furin, to be constitutively
is also evidence that BDNF 50 UTR mRNA splice released, or within secretory granules by
variants have differences in the expression and convertases in the regulated pathway (Seidah
localization of individual BDNF transcripts et al. 1996; Mowla et al. 1999). In these
between cell types, creating a spatial code that situations, pro-BDNF is intracellularly converted
facilitates local structural and functional plasticity and secreted as a 14 kDa mature BDNF. As a
in specific synaptic connections between hippo- result, there are two secreted BDNF forms, the
campal neurons (Maynard et al. 2017). In pro-BDNF and the mature BDNF, which increase
neurons, BDNF mRNA variants have a distinct the complexity of biological effects.
subcellular distribution, constituting a “spatial Pro-BDNF seems not only to represent a pre-
code,” with exon 1, 3, 5, 7, and 8 located in cursor for the biologically active BDNF but also
neuronal soma, exon 4 extending into proximal to play biologically active functions, such as
dendrites, and exon 2 and 6 reaching distal modulation of synaptic plasticity, neuronal mor-
dendrites (Aliaga et al. 2009; Baj et al. 2011; phology, or cell survival (Lee et al. 2001;
Colliva and Tongiorgi 2020). In a similar way to Nagappan et al. 2009; Yang et al. 2009; Sun
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 97

Fig. 8.1 Production and processing of BDNF. BDNF by convertase enzymes. If pro-BDNF reaches the extracel-
mRNAs with a short 30 UTR are restricted to the soma, lular milieu, it can be processed by the endopeptidases
while long 30 UTR mRNAs are targeted to dendritic spines. tPA/plasmin system and matrix-metalloproteinases, and
Pro-BDNF may be processed to mature BDNF by several the resulting mature BDNF then activates cell surface
cellular mechanisms. Pro-BDNF can be cleaved in the TrkB receptors. Alternatively, extracellular pro-BDNF
trans-Golgi by furin and in regulated secretory vesicles can bind to p75NTR (from Ribeiro 2017)

et al. 2012; Dieni et al. 2012). Indeed, pro-BDNF hippocampal neurons or electroconvulsive stimu-
was shown to be predominantly secreted in lation of rat brains, extracellular mature BDNF
response to low frequency stimulation, being levels were highly increased mainly due to
responsible for facilitating long-term depression co-secretion of pro-BDNF and tPA that extracel-
(LTD), while mature BDNF was mostly secreted lularly produce mature BDNF, and probably also
following high frequency stimulation, promot- to increased levels of intracellular prohormone
ing long-term potentiation (LTP) (LTP and LTD convertase 1, which intracellularly converts
are cellular mechanisms of long-term synaptic pro-BDNF into mature BDNF (Nagappan et al.
plasticity underlying learning and memory forma- 2009; Segawa et al. 2013). Apparently,
tion, maintenance, and decay). In fact, as a conse- pro-BDNF is the main form secreted in vivo and
quence of high frequency stimulation of cultured the ratio pro-BDNF/mature BDNF is crucial for
98 F. F. Ribeiro and S. Xapelli

the regulation of long-term synaptic plasticity, as above mature neurotrophins, bind with high affin-
well as pathological conditions (Foltran and Diaz ity to this second class of receptors (Lee et al.
2016). 2001), also signaling through a specific
Moreover, BDNF can be anterogradely co-receptor, sortilin (Teng et al. 2005).
transported from the cell soma to axon terminals The mature BDNF elicits its trophic actions
(Smith et al. 1997; Adachi et al. 2005; Lu et al. through high-affinity binding to TrkB, which
2014). For instance, in hippocampal mossy fibers, triggers receptor dimerization and subsequent
BDNF is transcribed and translated at the soma of autophosphorylation of multiple tyrosine residues
granule neurons and then transported in the cytoplasmic receptor domain. These
anterogradely to provide trophic support of CA3 phosphorylated tyrosine residues promote the
pyramidal neurons (Smith et al. 1997). For that, activation of three main intracellular signaling
BDNF and pro-BDNF have been suggested to be pathways (Fig. 8.2), by the recruitment of various
stored in presynaptic large dense core vesicles adaptor proteins, such as Src homologous and
(Dieni et al. 2012). collagen-like (Shc) adaptor protein, or phospholi-
As mentioned above, pro-BDNF is enzymati- pase C (PLC)γ. Shc adaptor protein links the
cally cleaved to generate mature BDNF. Apart activated TrkB receptor to two separated intracel-
from mature BDNF, as a result of this proteolytic lular signaling cascades, namely, phosphatidy-
processing, it is generated a N-terminal fragment linositol 3-kinase (PI3K)/Akt pathway and Ras
from the pro-BDNF, which called the pathway which leads to the activation of
pro-peptide. Interestingly, this pro-peptide also mitogen-activated protein kinases/extracellular
has biological activity. Both BDNF and its signal-regulated kinases (MAPK/ERK) cascade.
pro-peptide have been found to be stored in pre- In addition, PLCγ binds to activated Trk receptors
synaptic dense core vesicles in hippocampal and initiates an intracellular signaling cascade
neurons, and the BDNF pro-peptide also seems resulting in the release of inositol phosphates,
to be involved in neuronal plasticity, such as in with consequent release of intracellular calcium
facilitating LTD (Mizui et al. 2017; Dieni et al. (Ca2+), and activation of protein kinase C (PKC).
2012). There are several isoforms of TrkB receptors.
The afore described isoform is called the TrkB
full-length (TrkB-FL). Other isoforms contain an
8.3 Neurotrophin Receptors extracellular ligand binding domain and a single
transmembrane domain, lacking, however, the
Neurotrophins interact with two types of trans- intracellular tyrosine kinase domain. These are
membrane cell surface receptors: the high-affinity the truncated TrkB isoforms that are generated
tropomyosin-related kinase receptor (Trk), which from alternative splicing (Klein et al. 1990;
belongs to the tyrosine kinase family of receptors; Middlemas et al. 1991; Stoilov et al. 2002;
and the low-affinity pan-neurotrophin receptor Luberg et al. 2010). There are two most known
p75NTR, which belongs to the tumor necrosis C-terminal truncated TrkB isoforms, TrkB-T1
factor (TNF) receptor superfamily. The mature and TrkB-T2 (Klein et al. 1990; Middlemas
neurotrophins bind to the class of the Trk family et al. 1991). In humans, however, three isoforms
of tyrosine protein kinase receptors. These are the have been described, the C-terminal truncated
major membrane signal-transducing receptors receptor (TrkB-T1), the C-terminal truncated
that include the TrkA, TrkB, and TrkC receptors, receptor containing a Shc-binding site (TrkB-T-
for which the main ligands are, respectively, Shc), and a C-terminal truncated receptor
NGF, BDNF/NT-4, and NT-3 (Chao 2003). In retaining tyrosine kinase activity (TrkB-T-TK)
addition, all neurotrophins can also bind to (Stoilov et al. 2002; Luberg et al. 2010). TrkB-
p75NTR with similar low affinity (approximately T1 and TrkB-T2 are similar to TrkB-FL except at
100-fold less than to Trk receptors). On the other the intracellular domain, where they only have,
hand, pro-neurotrophins, the precursors of the respectively, 23 and 21 amino acid residues and
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 99

Fig. 8.2 Signaling


cascades activated by the
neurotrophin receptor
TrkB. BDNF binding to
TrkB triggers the activation
of signaling cascades, such
as Ras,
phosphatidylinositol
3-kinase (PI3K), and
phospholipase C (PLC)γ
pathways, resulting in gene
expression, neuronal
survival, and neurite
outgrowth (from Ribeiro
2017)

lack the kinase domain (Klein et al. 1990; internalized (Biffo et al. 1995). The truncated
Middlemas et al. 1991). The first 12 amino acid receptors may also function as dominant negative
residues of the intracellular domain of both receptors through the formation of non-functional
truncated TrkB isoforms are common with those heterodimers with TrkB-FL, thus inhibiting BDNF
of TrkB-FL, and the remaining residues of the signaling (Eide et al. 1996; Haapasalo et al. 2002).
intracellular domain are isoform-specific of each However, other studies suggest that TrkB-T1 and
truncated receptor (Baxter et al. 1997). TrkB-FL TrkB-T2 are able to induce signaling transduction
is abundantly expressed in neuronal cells, while in response to BDNF binding, through their short
truncated receptors are mainly found in glial cells, cytoplasmic domain (Baxter et al. 1997). Also,
being widely expressed during development and TrkB-T1 has been shown to evoke rapid calcium
in the adult brain (Reichardt 2003). TrkB-T-Shc transients in astrocytes (Rose et al. 2003) and to
has been identified only in human neurons alter astrocytic morphology via Rho GTPase
(Stoilov et al. 2002). In addition to these truncated (Ohira et al. 2005) in response to BDNF binding,
receptors originated from alternative splicing, as well as to allow glycine (Aroeira et al. 2015) and
TrkB-FL protein can be cleaved at the cytosolic GABA (Vaz et al. 2011) transport. In fact,
domain by calpains to produce the TrkB truncated astrocytes were shown to predominantly express
form TrkB-T0 and the TrkB-ICD fragment of TrkB-T1, which is a TrkB isoform that was
32 kDa containing the protein kinase domain demonstrated to increase astrocyte morphological
(Jerónimo-Santos et al. 2015; Tanqueiro et al. complexity in response to BDNF, during the astro-
2018; Fonseca-Gomes et al. 2019). cytic maturation process (Holt et al. 2019), as well
There is no clear answer about the function or as to be important in cell migration and prolifera-
signaling pathways activated by truncated tion (Matyas et al. 2017).
receptors. Some findings suggest that truncated Regarding p75NTR, it is composed of an
TrkB isoforms function as selective scavengers extracellular cysteine-rich domain, a single trans-
that prevent extracellular BDNF diffusion since, membrane domain and a cytoplasmic domain that
when BDNF binds to these receptors, it is contains a “death” domain. Activation of
100 F. F. Ribeiro and S. Xapelli

p75NTR triggers nuclear factor-kB (NF-kB) and contradictory findings, BDNF constitutes a key
Jun kinase-signaling pathways, thus, respec- regulatory player in the neurogenic process. Both
tively, regulating cell survival and death BDNF and its high-affinity TrkB receptor are
(Reichardt 2006). Besides lacking an intracellular expressed in the adult mouse hippocampus in
kinase domain, p75NTR can cooperate with other neurons and astrocytes (Ivanova and Beyer
proteins to enhance biological responses, forming 2001; Li et al. 2008). Within the hippocampus,
functional receptor complexes with Trk receptors TrkB is strongly expressed in the granule layer of
to increase binding affinity to their preferred the DG, as well as in the stratum pyramidale of
neurotrophins (Barker 2004), and with sortilin, areas CA1–CA3 and within the subiculum, and in
forming a p75NTR-sortilin complex to which moderate levels in the stratum radiatum, stratum
pro-neurotrophins bind, like pro-BDNF (Teng lacunosum moleculare, and the molecular layer of
et al. 2005). Evidence suggests that sortilin also the DG (Yan et al. 1997). TrkB expression levels
interacts with TrkA, TrkB, and TrkC receptors, have been shown to increase with the progression
enabling their anterograde axonal transport, toward neuronal maturity. However, stem-like
thereby enhancing neurotrophin signaling cells, which rarely divide, express the TrkB pro-
(Vaegter et al. 2011). Moreover, there are two tein, in contrast to proliferating progenitors
known p75NTR isoforms: a short (s-p75NTR) (Donovan et al. 2008). Using primary murine
and a full-length isoform. The full-length isoform hippocampal cultures, it has been shown that
is able to bind to neurotrophins, while the short BDNF levels increased stepwise from embryonic
isoform, whose functions are largely unknown, day 15 until the second postnatal week, whereas
lacks the neurotrophin binding site (Fujii and TrkB expression was stable throughout develop-
Kunugi 2009). ment. BDNF was shown to be strongly expressed
These receptors have shown to mediate utterly in the granule layer of the DG (Li et al. 2008), and
opposite effects. For instance, studies have shown particularly in mossy fiber axons of the dentate
that pro-BDNF binds to p75NTR to facilitate granule neurons (Conner et al. 1997). However,
LTD in hippocampal neurons, initiates in humans, the levels of BDNF mRNA did not
programmed neuronal death, collapses neurite change significantly in the hippocampus with
outgrowth, or negatively regulates dendritic com- aging, whereas the levels of TrkB mRNA,
plexity and spine density (Lee et al. 2001; Woo whether of the TrkB-T isoform or the TrkB-FL
et al. 2005; Nagappan et al. 2009; Sun et al. 2012; isoform, decreased (Webster et al. 2006). More-
Yang et al. 2014). In opposition, when activated over, post-mortem findings from patients with
by BDNF, TrkB enhances LTP, cell survival, and bipolar disorder, major depressive disorder,
dendritic complexity (Pang et al. 2004; Nagappan schizophrenia, and Alzheimer’s disease showed
et al. 2009). Hence, a bidirectional regulation that BDNF levels are decreased in hippocampus
seems to exist between both receptors and pro- (Hock et al. 2000; Reinhart et al. 2015).
and mature BDNF. On the other hand, the lower-affinity receptor
for BDNF, p75NTR, can also be found within the
hippocampal formation. In young mice, both
granule cells of the DG and pyramidal neurons
8.4 BDNF Role in Adult
of CA1-CA3 express high levels of p75NTR
Hippocampal Neurogenesis
mRNA (Zagrebelsky et al. 2005). Within the
DG, p75NTR is mainly localized presynaptically,
The role of BDNF in adult neurogenesis has been
but can also be found in astrocytes and in the
the subject of several studies. This neurotrophin
dendrites and cell bodies of some granule cells
has been implicated in the regulation of adult
(Dougherty and Milner 1999). p75NTR has been
neurogenesis at different levels, in both dentate
shown to localize in dendritic spines, in addition
gyrus (DG) and SVZ neurogenic niches. Despite
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 101

to afferent terminals, of CA1 neurons (Woo et al. through TrkB cascade, favored cell survival and
2005), and in cell bodies within the DG (Barrett migration of murine adult hippocampal precursor
et al. 2005). Neural progenitor cell (NPC)-specific cells during differentiation to glial and neuronal
knockout of p75NTR did not affect proliferation lineages, thus increasing these cell types. (Ortiz-
of NPCs nor altered neurogenesis in the adult López et al. 2017). Moreover, BDNF, through
hippocampus (Groves et al. 2019). Regarding TrkB signaling, has been consistently shown to
the co-receptor sortilin, it is highly expressed in have an essential regulatory role in dendritic com-
the hippocampus, and particularly in the soma plexity, as well as in synaptic formation, matura-
and dendrites of neurons located in CA1-CA3 tion, and plasticity of newborn SGZ neurons
and within the DG (Sarret et al. 2003). (Chan et al. 2008; Gao et al. 2009; Wang et al.
BDNF has been described to be an important 2015).
player in DG neurogenesis (Scharfman et al. BDNF is an important factor that is involved in
2005; Taliaz et al. 2010; Ferreira et al. 2018). the regulation of hippocampal spine densities and
Using antibodies to scavenge BDNF in vitro led spine shape, and these effects seemed to be
to decreased neuronal differentiation of progeni- mediated by TrkB receptors, since receptor dele-
tor cells (Babu et al. 2009). However, there are tion in adult neural progenitors reduced the den-
contradictory reports on the role of BDNF upon dritic complexity and spine density in adult-born
proliferation, with some showing an increase hippocampal neurons (Bergami et al. 2008), thus
(Li et al. 2008; Ferreira et al. 2018), while others suggesting the relevance of BDNF/TrkB signal-
no changes in progenitor cell proliferation in the ing for dendrite morphogenesis of adult-born
presence of BDNF (Lee et al. 2002; Choi et al. neurons. In in vivo, during adult neurogenesis in
2009), and others demonstrating that BDNF the DG, BDNF has been described to be locally
depletion induces SGZ progenitor cell prolifera- synthesized and released from the dendrites of
tion, concluding that BDNF is required for the mature granule cells which through stimulation
terminal differentiation of new neurons in the of TrkB receptors on parvalbumin (PV)-
adult hippocampus (Chan et al. 2008). Similarly, expressing GABAergic interneurons leads to mat-
there are also conflicting data regarding the uration of adult-born neurons through GABA
BDNF role upon survival of new neurons in the inputs (Waterhouse et al. 2012). In fact, ErbB4
adult DG (Sairanen et al. 2005; Chan et al. 2008; ablation in PV neurons inhibited adult hippocam-
Choi et al. 2009), with reports showing that pal neurogenesis in mice through down-
BDNF signaling is required for the survival of regulation of the BDNF–TrkB pathway (Zhang
newborn neurons in mouse hippocampus et al. 2018). Rather than a paracrine action, den-
(Sairanen et al. 2005; Choi et al. 2009), and others drite morphogenesis of adult-born granule cells is
that it is not necessary for cell survival (Chan mainly regulated by an autocrine action of BDNF
et al. 2008). in these neurons, possibly as a result of
The high-affinity BDNF receptor TrkB seems GABAergic or glutamatergic inputs (Wang et al.
to play a role in these BDNF-mediated actions on 2015), whereas BDNF released from adult-born
adult hippocampal neurogenesis. Deletion of the granule cells also proved to have an impact, even
BDNF or NTRK2 (TrkB gene) in progenitor cells though minor, in adult-born neuronal dendrite
blocked BDNF-induced neurosphere growth growth and formation of the dendritic branches
in vitro, and impaired proliferation and (Wang et al. 2015). Mice, in which long 30 UTR
neurogenesis in vivo, together with a reduction BDNF mRNA is not generated, displayed
of DG thickness (Lee et al. 2002; Li et al. 2008). decreased differentiation, maturation, and inte-
The generation of new neurons during adulthood gration of neuronal precursors in the dentate
involves local precursor cell migration and termi- gyrus (Waterhouse et al. 2012). Using specific
nal differentiation in the DG, which is influenced knockout mice lacking BDNF production from
by the hippocampal microenvironment. BDNF, exons 1, 2, 4, or 6 splice variants, it was found
102 F. F. Ribeiro and S. Xapelli

that loss of BDNF from different BDNF mRNA 8.5 Role of Environmental Factors
variants differentially affected dendritic complex- on BDNF and Adult
ity and spine morphology in the hippocampus Hippocampal Neurogenesis
(Maynard et al. 2017). These data demonstrate
that the diverse actions of BDNF rely on different Environmental factors can also stimulate the pro-
splice variants. duction of BDNF in the hippocampus and thus
Not only TrkB activation promotes adult regulate adult hippocampal neurogenesis, such as
neurogenesis enhancement, but also p75NTR physical exercise (Cotman and Berchtold 2002;
(Bernabeu and Longo 2010). p75NTRs are Berchtold et al. 2005; Marlatt et al. 2012), and
expressed by adult dentate progenitor persisting enriched environment (Kuzumaki et al. 2011).
until the early stages of neuronal and glial cell Adult hippocampal neurogenesis can be
maturation. These receptors are important for pro- enhanced by voluntary exercise. Indeed, running
moting proliferation and/or early maturation of increases cell proliferation and neurogenesis in
not only neural, but also glial cells, being impor- the adult mouse DG (van Praag et al. 1999).
tant for regulating neuronal and non-neuronal cell Rats subjected to forced run on a treadmill for
genesis. In fact, mice lacking the p75NTR recep- 30 min 5 times a week for 4 weeks showed
tor gene (p75(NTR-/-)) have significantly reduced improvements in short-term memory which was
hippocampal neurogenesis (Bernabeu and Longo impaired by maternal lipopolysaccharide (LPS)
2010). P75NTR is also important for axon speci- exposure. Moreover, increased cell proliferation
fication. In cultured hippocampal neurons, local and neurogenesis in the DG and increased BDNF
exposure to neurotrophins leads to p75NTR and TrkB expression were observed in the hippo-
asymmetrical localization in the differentiating campus of rats with treadmill exercise born from
neuron, accumulating into that undifferentiated LPS-exposed maternal rats (Kim et al. 2015). On
neurite. This process leads to specification of the the other hand, in mice allowed to voluntarily
future axon. Moreover, knockout or knockdown exercise, with free access to a running wheel,
of p75NTR results in axon initiation failure dur- only a few days of exercise was enough to induce
ing in vivo adult hippocampal neurogenesis an increase in BDNF mRNA levels in the DG
(Zuccaro et al. 2014). As a p75NTR ligand, (Neeper et al. 1995). By either inhibiting BDNF
BDNF induces the specification of axons in action or blocking TrkB, the benefit induced by
cultured developing hippocampal neurons (Shelly exercise on cognitive function, such as learning
et al. 2007; Cheng et al. 2011). These beneficial and recall abilities, was reduced to levels similar
effects of p75NTR may be possible through to sedentary control (Vaynman et al. 2004, 2006).
partnering with TrkB and binding to mature Other studies have shown beneficial effects of
BDNF. Contrary to the beneficial effects of exercise on hippocampal neurogenesis through-
mature BDNF on adult neurogenesis, pro-BDNF out life span. For example, aerobic exercise train-
seems to attenuate neurogenesis in the hippocam- ing has shown to increase hippocampal volume in
pus (Chen et al. 2016), possibly through binding adult humans by 2%, thus reversing age-related
to the complex p75NTR/sortilin. Moreover, loss- loss in volume by 1–2 years. Also, increased
of-function experiments show that mutant lines of hippocampal volume was associated with greater
p75NTR have a higher spine density and greater serum levels of BDNF and spatial memory
dendritic complexity than wild-type (WT) mice. improvement (Erickson et al. 2011). Another
On the contrary, p75NTR overexpression signifi- work studied the effect of a 30-min running
cantly reduced dendritic complexity and spine once a day for 6 weeks in hippocampal
density in all dendritic compartments. These neurogenesis of young and adult rats. Results
results show that p75NTR negatively modulates showed an increase in hippocampal neurogenesis
dendritic morphology in hippocampal neurons, in in both ages, being more active in young than in
opposition to TrkB (Zagrebelsky et al. 2005). adult rats. BDNF and TrkB expression in the
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 103

hippocampus were also enhanced in both ages, wheels. These data demonstrate that voluntary
but they were greater in the adult rats. Results exercise is sufficient to enhance neurogenesis in
showed that adults have much better spatial the adult mouse DG (van Praag et al. 1999), also
learning abilities than young rats in the absence suggesting that the benefits from environmental
of exercise. But the improvement of exercise- enrichment are highly dependent on the access to
induced spatial learning ability through running wheels (Russo-Neustadt et al. 2000;
neurogenesis was better in younger age (Jin Kobilo et al. 2011), as voluntary exercise in run-
et al. 2017). Exercise was also reported to provide ning wheels was, by itself, able to increase BDNF
cognitive benefit to a mouse model of levels and promote adult hippocampal
Alzheimer’s disease, 5FAD, by inducing adult neurogenesis (Neeper et al. 1995). Mice lacking
hippocampal neurogenesis and elevating the expression of BDNF through promoter IV
levels of BDNF. In fact, combining adult (BDNF-KIV mice) exhibit a depression-like phe-
neurogenesis with BDNF mimicked exercise notype (Sakata et al. 2009). Three weeks in an
effects on cognition in an Alzheimer’s mouse enriched environment, including running wheels,
model (Choi et al. 2018). Long-term exercise rescued depression-like behavior of BDNF-KIV
not only elevates the rate of adult neurogenesis mice and increased other BDNF transcripts,
and BDNF levels (Marlatt et al. 2012), but also restoring BDNF protein levels in the hippocam-
pro-BDNF levels in the hippocampus. In addi- pus. An enriched environment also restored the
tion, voluntary exercise increases the activity of reduction in cell survival and dendritic length and
the serine protease tPA that facilitates pro-BDNF increased cell proliferation in BDNF-KIV mice.
cleavage into mature BDNF. This suggests that These results suggest that other BDNF promoters
the beneficial effects on adult neurogenesis compensate the loss of a specific BDNF
depend on cleavage of pro-BDNF to mature promotor, rescuing BDNF levels and BDNF-
BDNF, which, in turn, can signal through TrkB mediated function associated to environmental
(Ding et al. 2011). enrichment (Jha et al. 2011).
Exposure to an enriched environment Nutrition is also an environmental factor that
increases neurogenesis in the DG of adult rodents. can influence BDNF levels and adult hippocam-
BDNF is required for the enhancement of hippo- pal neurogenesis. For instance, rodents fasted
campal neurogenesis following environmental intermittently exhibit enhanced hippocampal
enrichment (Rossi et al. 2006). Indeed, environ- neurogenesis and LTP at hippocampal synapses
mental enrichment increases BDNF mRNA compared with sedentary animals fed an ad
expression via sustained epigenetic modification libitum diet. Curiously, an increase in BDNF
in the mouse hippocampus (Kuzumaki et al. was also observed in animals subjected to inter-
2011). However, environmental enrichment mittent fasting (Baik et al. 2020). Conversely,
consists of many components, such as expanded obesity and psychosocial stress combined in the
learning opportunities, increased social interac- same individual, as it happens frequently in the
tion, more physical activity, and larger housing. Western society, augmented anxiety-like behav-
A study assigned adult mice to a separate envi- ior, and impaired spatial memory of adult mice.
ronmental enrichment component: water-maze These changes were associated with reduced hip-
learning (learner), swim-time-yoked control pocampal volume, neurogenesis, local BDNF and
(swimmer), voluntary wheel running (runner), TrkB levels, and amplified astrogliosis (Agrimi
and enriched (enriched) and standard housing et al. 2019).
(control) groups and evaluated the number of Neurogenesis in the SGZ of the DG is also
BrdU+ cells. They observed that neither maze negatively regulated by stressful experiences,
training nor yoked swimming had any effect on leading to a depression condition, and positively
BrdU+ cell number, but running doubled the by treatment with antidepressant drugs (Hanson
number of surviving newborn cells, in amounts et al. 2011). In fact, chronic antidepressant
like enriched conditions, which included running administration also promotes an increase in
104 F. F. Ribeiro and S. Xapelli

BDNF expression (Nibuya et al. 1995; Russo- have demonstrated that in vitro BDNF enhances
Neustadt et al. 2000; Sairanen et al. 2005), while cell proliferation and increases the number of
BDNF infusion mimics the antidepressant behav- neurospheres, favoring symmetric divisions
ioral effects (Shirayama et al. 2002). More toward self-renewal (Ferreira et al. 2018). How-
recently, TrkB-dependent adult hippocampal ever, Tervonen and colleagues demonstrated that
neurogenesis was shown to mediate sustained the number of neurosphere-forming progenitors,
ketamine antidepressant response (Ma et al. obtained from SVZ cells from mice
2017). overexpressing TrkB-T1, was reduced, and apo-
ptosis increased, when compared with WT
neurospheres. Interestingly, these overexpressing
8.6 BDNF Role in SVZ-Derived TrkB-T1 cells showed a higher cell proliferation
Neurogenesis rate, measured by the increase in the diameter of
postnatal mice SVZ-derived neurospheres
The SVZ expresses low levels of BDNF (Galvão (Tervonen et al. 2006). These results reveal a
et al. 2008), with levels not changing throughout role for truncated TrkB receptors in modulating
the adult lifespan (Werry et al. 2010), and with postnatal neurogenesis, namely, the capacity of
SVZ astrocytes and ependymal cells being the neural progenitors to proliferate, and suggest that
possible source of BDNF (Tonchev 2011). these receptors may be modifying the action
Proliferating cells in the SVZ have been reported of BDNF.
to express both the full-length and the truncated Work aimed at testing the role of BDNF in the
form of the TrkB receptors, but not p75NTR migration of SVZ-derived cells has been most
(Tervonen et al. 2006; Galvão et al. 2008). consensual. Along the rostral migratory stream
p75NTRs are expressed in SVZ intermediate (RMS), BDNF is synthesized and secreted by
progenitors (type C cells) and migrating the endothelial cells of the blood vessels that
neuroblasts (type A cells) (Hosomi et al. 2003; outline the migratory stream (Snapyan et al.
Galvão et al. 2008). 2009), being a key player in the regulation of
BDNF has been shown to increase the number neuroblast migration (Snapyan et al. 2009;
of new neurons in the olfactory bulb (OB) (Zigova Bagley and Belluscio 2010). Indeed, BDNF scav-
et al. 1998; Benraiss et al. 2001; Henry et al. enging using TrkB-Fc (a fusion protein compris-
2007). However, studies on the role of BDNF in ing the Fc domain of human IgG and the
regulating SVZ-neuronal survival have provided ectodomain of TrkB) or genetical ablation of
conflicting results. First studies, in rat cell BDNF from vascular endothelial cells decreased
cultures, showed BDNF as a mediator of neuronal neuroblast migration in vitro and in vivo
survival (Kirschenbaum and Goldman 1995). In (Snapyan et al. 2009). However, the type of
agreement, later in vivo studies showed increased receptors through which BDNF signals, either
cell death in animals where BDNF is genetically TrkB-FL, TrkB-T or p75NTR, to regulate
knocked out or mutated (Linnarsson et al. 2000; neuroblast migration is not yet established.
Bath et al. 2008), or enhanced survival of new Some studies have shown that TrkB-FL is not
neurons in BDNF overexpressing animals essential for RMS migration (Galvão et al. 2008;
(Benraiss et al. 2001). In contradiction, others Bergami et al. 2013) since it is not expressed in
have shown that BDNF does not affect the sur- neuroblasts until they arrive to the OB, but only in
vival of SVZ-derived neurons (Galvão et al. astrocytes and ependymal cells (Galvão et al.
2008). These different responses to BDNF may 2008; Bergami et al. 2013). In fact, TrkB-T was
rely on BDNF receptor expression in the different shown to be abundantly expressed by nestin+
cells throughout neurogenic process. cells within the neurosphere, in differentiated
Galvão and colleagues found that TrkB knock- glia cells, and in immature neurons within adher-
out mice decreased cell proliferation and survival ent differentiated neurospheres, whereas TrkB-
in the SVZ (Galvão et al. 2008). Accordingly, we FL was expressed in fully differentiated post-
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 105

mitotic neurons and in immature astrocytes produced neuroblasts that migrated to the OB in
(Frisén et al. 1993; Rudge et al. 1994; Climent the absence of TrkB. The survival and differenti-
et al. 2000; Galvão et al. 2008; Islam et al. 2009). ation of granule cells and calbindin+
Instead, migrating neuroblasts express TrkB-T1 periglomerular interneurons seemed unaffected
and p75NTR (Galvão et al. 2008; Snapyan et al. by the loss of TrkB, but dopaminergic
2009), with p75NTR being pointed as a good periglomerular neurons were reduced (Galvão
candidate (Snapyan et al. 2009). It is suggested et al. 2008). On the other hand, overexpressing
that BDNF promotes neuroblast migration via BDNF in the granule cell layer did not affect
p75NTR. Then, GABA released from neuroblasts survival, as well as the numbers of new granule
induces Ca2+-dependent insertion of high-affinity cells when mice were sensory deprived by unilat-
TrkB receptors on the plasma membrane of eral naris occlusion (McDole et al. 2020).
astrocytes that surround neuronal precursors. As Although not affecting the turnover of granule
a result, astrocytes trap extracellular BDNF, cell layer in vivo, sustained BDNF over-
reducing the amount of BDNF that binds to the expression produces a marked increase in granule
low-affinity receptor p75NTR on neuroblast, cell spine density, leading to spine maturation on
hence regulating cell migration (Snapyan et al. their apical dendrites (McDole et al. 2015). In
2009; Grade et al. 2013). In accordance, in fact, BDNF has been previously demonstrated to
p75NTR null animals, a significant reduction in promote dendritic growth of SVZ-derived neuro-
OB volume was observed, concomitant with a nal precursors during the initial 3 days in culture
decrease in the number of proliferating and (Gascon et al. 2005), and to selectively regulate
migrating neuroblasts (Young et al. 2007). How- dendritogenesis of PV-expressing interneurons in
ever, complete ablation of p75NTR gene in mice the main OB in vivo (Berghuis et al. 2006). Mor-
did not affect newly born cell density in the OB phometric analysis demonstrated that changes in
(Bath et al. 2008). spine density in apical dendrites of granule cell
In a similar way to the RMS, BDNF is highly neurons were most observed in the distal and
expressed in the OB (Bath et al. 2008), being proximal domains, suggesting that multiple excit-
suggested to be released by local glutamatergic atory inputs are regulated by BDNF (McDole
neurons and long projecting neurons from the et al. 2015).
anterior olfactory nucleus and the piriform cortex In addition to neurogenic regions, intraventric-
(Bergami et al. 2013). Only when they arrive to ular administration of BDNF for 12 days has been
the OB, immature neurons start to express TrkB- shown to increase the number of BrdU+ cells in
FL. Through TrkB-FL, BDNF has a physiologi- specific parenchymal structures lining the lateral
cal role during the integration of newborn and third ventricles, including the striatum, sep-
neurons, regulating the degree of connectivity in tum, thalamus, and hypothalamus, thus showing
integrating neurons, particularly in an association between BDNF and a positive
periglomerular cells, by shaping their dendritic regulation of adult neurogenesis in these noncon-
complexity and spine density, through, respec- ventional neurogenic areas (Pencea et al. 2001).
tively, Shc/PI3K and PLCγ activation (Bergami
et al. 2013). Indeed, TrkB-deficient neurons
displayed a marked impairment in dendritic 8.7 Regulation of Adult
arborization and spine growth. Moreover, BDNF Neurogenesis by NGF
has also a critical role in balancing the turnover of
adult-born periglomerular cells, as TrkB deletion The role of pro-NGF in the biology of adult
compromised the survival of the new dopaminer- neural stem cells (NSCs) is still not clear. How-
gic neurons, but not of the granule cells (Bergami ever, there are several reports that study the inter-
et al. 2013). In accordance, when SVZ cells from vention of this proneurotrophin in the adult
TrkB knockout mice (TrkB-KO) were grafted hippocampal neurogenesis. Interestingly,
into the SVZ of WT, grafted progenitors pro-NGF has been reported to be highly
106 F. F. Ribeiro and S. Xapelli

expressed in aging brains and in the brains of with a reduction in hippocampal neurogenesis
patients with mild cognitive impairment and was reported. In accordance, AD11 hippocampal
with Alzheimer’s disease (Peng et al. 2004; Guo NSCs proliferated more in vitro but were unable
et al. 2013). Indeed pro-NGF accumulation in the to differentiate into morphologically mature
adult hippocampus has been correlated with neurons. Moreover, radial glial-like cell
memory deficits and loss of cognitive function expressed high levels of p75NTR and responded
(Peng et al. 2004; Guo et al. 2013). Pro-NGF to chronic pro-NGF by reactivating their prolifer-
infusion into adult mouse hippocampus signifi- ation, leading to the formation of neurospheres
cantly reduced the density of BrdU+ cells and the (Corvaglia et al. 2019). Altogether, these studies
density of BrdU/DCX-double positive cells in the are consensual in showing that pro-NGF impairs
SGZ, indicating an inhibitory effect of pro-NGF hippocampal neurogenesis.
on hippocampal neurogenesis. Moreover, Regarding the mature form, NGF is highly
pro-NGF infusion induced prominent cell apopto- expressed in the adult hippocampus
sis and activated resident astrocyte and microglia, (Maisonpierre et al. 1990). Chronic intracerebro-
which might further contribute to an impairment ventricular infusion of NGF for 6 weeks
in the hippocampal neurogenesis (Guo et al. enhanced recognition memory and increased the
2013). With the high-affinity receptor of expression of TrkA, synaptogenesis, and cell pro-
pro-NGF, p75NTR, being expressed on both liferation in the DG (Birch and Kelly 2013).
proliferating cells and postmitotic mature cells However, other data suggest that treating adult
in the adult mouse hippocampus (Guo et al. and aged male rats intracerebroventricularly
2013), the pro-NGF role in inhibiting with NGF for 6 or 20 days increased hippocampal
neurogenesis in this region could be possible cholinergic activity, without affecting the prolif-
through the creation of a toxic signaling complex eration of progenitor cells in the granule cell layer
by simultaneously binding to p75NTR and of the DG. Nevertheless, continuous NGF infu-
sortilin, given that sortilin is expressed in the sion improved survival of new neurons in the
adult hippocampus and is essential for pro-NGF- granule cell layer of adult, but not of aged rats
induced neuronal cell death (Nykjaer et al. 2004; (Frielingsdorf et al. 2007).
Xu et al. 2019). In aging and related In the absence of exercise, short-term environ-
neurodegeneration, studies have shown that the mental enrichment improves memory, and
prefrontal cortex and hippocampus have around increases NGF concentration, early neuronal sur-
two-fold increase in the levels of the vival, and synaptogenesis in the DG in a time-
proneurotrophin, pro-NGF, p75NTR receptors, dependent manner (Birch et al. 2013). A six-week
and sortilin, and decreased levels of mature period of housing in an enriched environment,
NGF and phospho-TrkA receptors (Jansen et al. without access to exercise equipment, improved
2007; Al-Shawi et al. 2008; Terry et al. 2011; recognition memory. The improvement in cogni-
Meeker and Williams 2014). These data support tive function was coincident with increased
the argument that the sortilin/TrkA ratio is expression of NGF, but not BDNF in the rat
responsible for NGF-altered signaling in the hippocampus, and increased expression of synap-
aging brain, and that such changes may contribute tic vesicle proteins and cell proliferation in the
to an age-related decline in cognitive function. On DG (Birch et al. 2013).
the other hand, pro-NGF has been shown to act as Very little is known regarding the role of NGF
mitogen on hippocampal neurogenesis. Using upon SVZ-derived neurogenesis in physiological
AD11 transgenic mice, in which the constitutive conditions. It was demonstrated that swimming
expression of anti-NGF antibody leads to an exercise in adult rats for 5 days/week over a
imbalance of pro-NGF over mature NGF, an period of 8 weeks increases neurogenesis, neuro-
increase in the proliferation of progenitor cells nal survival, and neuronal maintenance in the
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 107

SVZ. At the same time, swimming exercise was 1990). In fact, the differentiation of NPCs in DG
associated to an increase in the levels of NGF in lacking NT-3 is compromised, rather than cell
the OB (Chae et al. 2014). Therefore, it is possible proliferation. Particularly, NT-3 affects the num-
that NGF may have a role in regulating OB ber of newly differentiated neurons, but not glia.
neurogenesis. Importantly, experiments Moreover, NT-3 conditional knockout mice
performed with adult male Sprague-Dawley rats displayed impaired LTP in the lateral, but not
subjected to middle cerebral artery occlusion for medial perforant path-granule neuron synapses,
2 h showed that treatment with intranasal NGF and exhibited deficits in spatial memory tasks,
failed to increase cell proliferation but improved indicating that NT-3 facilitates hippocampal plas-
survival of newly generated cells in ipsilateral ticity and learning and memory by regulating
striatum and SVZ (Zhu et al. 2011). In fact, B hippocampal neurogenesis (Shimazu et al. 2006).
cells in the SVZ do not express TrkA (Delgado Conditional deletion of NT-3 in endothelial
et al. 2014), which is in accordance with the cells leads to the loss of NT-3 protein in both
absence of an effect in cell proliferation. More- SVZ and CSF, showing that these cells are a
over, cells double labeled with BrdU and the major source of NT-3 in SVZ stem cell niche.
mature neuronal marker NeuN were increased in Type B cells and ependymal cells do not synthe-
striatum and SVZ in rats treated with intranasal size NT-3, but type B cells express TrkC
NGF. Therefore, these results demonstrate that receptors. In young adult (60-day-old) heterozy-
intranasal NGF promotes neurogenesis and sur- gous NT-3 mice, the proportion of dividing
vival of newly generated cells, an action that was BrdU-labeled retaining cells over GFAP+ cells
accompanied by functional recovery after cere- was increased, which was followed by higher
bral ischemia. (Zhu et al. 2011). numbers of OB neurons and significant changes
in olfactory behavior. Moreover, NT-3 promotes
an increase in the number of neurospheres
8.8 Regulation of Adult through a mechanism dependent on TrkC. Inter-
Neurogenesis by NT-3 estingly, while heterozygous NT-3 young adult
mice exhibited an increase in cell proliferation, in
Contrary to NGF and BDNF, NT-3 is by far the middle-aged mice there was a decrease in
most highly expressed neurotrophin in the imma- neurosphere formation, with B1 stem cells
ture regions of the CNS in which proliferation, becoming depleted (Delgado et al. 2014).
migration, and differentiation of neuronal
precursors is ongoing. NT-3 expression dramati-
cally decreases with the maturation of these 8.9 Regulation of Adult
regions. High levels of NT-3 are synthesized by Neurogenesis by NT-4
discrete populations of neurons and astrocytes in
the nervous system; however, it can be also pro- NT-4 is also known as neurotrophin-5 (NT-5) and
duced, at lower levels, by endothelial cells and by is the least well-understood member of the
the microvasculature in the choroid plexus. neurotrophin family. Together with BDNF,
Therefore, the NT-3 protein is also found in NT-4 is a ligand for TrkB receptor. Therefore, it
the CSF. would be expected for them to have similar
In the adult hippocampus, in a similar way to effects. In fact, both ligands have been reported
BDNF and NGF, NT-3 is highly expressed to promote survival and differentiation of hippo-
(Maisonpierre et al. 1990) as well as TrkC campal neurons (Ip et al. 1993), as well as neurite
receptors (Ip et al. 1993). Indeed, in the adult extension and survival of differentiated cerebellar
brain, the expression of NT-3 is largely confined granule cells, through TrkB (Gao et al. 1995),
to the hippocampal DG (Maisonpierre et al. among other findings. However, evidence
108 F. F. Ribeiro and S. Xapelli

suggests that TrkB receptor can also be differen- 8.10 Conclusion


tially stimulated by BDNF and NT-4, and conse-
quently, depending on the nature of the BDNF is the neurotrophin that has been the center
stimulation, each neurotrophin can trigger differ- of most attention over the last two decades, in
ent synaptic signals, and thus mediate different terms of regulating adult neurogenesis. Alto-
effects (Hernandez-Echeagaray 2020). For gether, this chapter strongly suggests that BDNF
instance, both BDNF and NT-4 are known to is an important regulator at different levels of the
modulate corticostriatal synaptic transmission. process, whether through binding as a
When corticostriatal synapses were exposed to pro-neurotrophin or as a mature neurotrophin to
BDNF and NT-4 separately, they increased the TrkB and/or p75NTR and sortilin. All these dif-
phosphorylated levels of TrkB receptors, thus ferent mechanisms may result in distinct actions
demonstrating no differences in the regulation of of BDNF, demonstrating the richness and fine-
the receptor. However, sequential exposure to tune regulation of adult neurogenesis exerted by
both neurotrophins results in different modulatory this molecule. Hence, this field needs and
effects on corticostriatal transmission, depending deserves to be further explored to contribute to a
on the exposure order. When NT-4 is added after better and deeper knowledge about the actions of
BDNF, it elicits an antagonistic effect, with BDNF and its receptors in physiological and
phosphorylated levels of TrkB being reduced. pathological conditions.
On the other hand, if BDNF follows NT-4, it Although being less explored, the few studies
elicits a synergistic effect, with activated levels that focus on the actions of NGF, NT-3, and NT-4
of TrkB being enhanced. Each effect was also strongly suggest an important role for these
demonstrated to be respectively dependent on neurotrophins in modulating adult neurogenesis.
TrkB-T and TrkB-FL (Torres-Cruz et al. 2019). Alterations in the expression levels of some of
Moreover, exposure to an enriched environment these mature and pro-neurotrophins are observed
is known to enhance the number of newly with aging and in neurodegenerative diseases.
generated neurons in the DG. WT mice and Therefore, future research should pay attention
NT-4–/– mice placed in an enriched environment to these ligands to further understand the intricate
for 8 weeks showed a two-fold increase in the modulatory mechanisms of adult neurogenesis
mitotic marker BrdU in the DG, as well as in the that involve all the neurotrophins, and therefore
number of cells double-stained for BrdU and the contribute to develop new strategies for brain
neuronal marker NeuN. This enhancement in hip- regeneration.
pocampal neurogenesis was, however, not
observed in BDNF+/– mice under enriched Acknowledgments This work was supported by
conditions. As a consequence, these data demon- IF/01227/2015 project funded by Fundação para a Ciência
strate that NT-4 is not required for the environ- e a Tecnologia (FCT). F.F.R. (IMM/CT/35-2018) received
a fellowship from FCT.
mental induction of neurogenesis, although
BDNF is (Rossi et al. 2006).
Consequently, besides NT-4 acting on the
same receptor as BDNF, the role of the two References
neurotrophins upon adult neurogenesis may be
different. These different actions could be depen- Adachi N, Kohara K, Tsumoto T (2005) Difference in
trafficking of brain-derived neurotrophic factor
dent on the different availability of each TrkB
between axons and dendrites of cortical neurons,
isoform on the membrane surface, as well as in revealed by live-cell imaging. BMC Neurosci 6:42.
the extracellular concentration of each ligand and https://doi.org/10.1186/1471-2202-6-42
their different affinity for each receptor. This is an Agrimi J, Spalletti C, Baroni C et al (2019) Obese mice
exposed to psychosocial stress display cardiac and
area that is very poorly explored and whose
hippocampal dysfunction associated with local brain-
knowledge will contribute to further understand derived neurotrophic factor depletion. EBioMedicine
the role of TrkB signaling.
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 109

47:384–401. https://doi.org/10.1016/j.ebiom.2019.08. discrimination. J Neurosci 28:2383–2393. https://doi.


042 org/10.1523/JNEUROSCI.4387-07.2008
Aid T, Kazantseva A, Piirsoo M et al (2007) Mouse and rat Baxter GT, Radeke MJ, Kuo RC et al (1997) Signal
BDNF gene structure and expression revisited. J transduction mediated by the truncated trkB receptor
Neurosci Res 85:525–535. https://doi.org/10.1002/jnr. isoforms, trkB.T1 and trkB.T2. J Neurosci
21139 17:2683–2690
Aliaga EE, Mendoza I, Tapia-Arancibia L (2009) Distinct Benraiss A, Chmielnicki E, Lerner K et al (2001) Adeno-
subcellular localization of BDNF transcripts in viral brain-derived neurotrophic factor induces both
cultured hypothalamic neurons and modification by neostriatal and olfactory neuronal recruitment from
neuronal activation. J Neural Transm (Vienna) endogenous progenitor cells in the adult forebrain. J
116:23–32. https://doi.org/10.1007/s00702-008-0159- Neurosci 21:6718–6731
8 Berchtold NC, Chinn G, Chou M et al (2005) Exercise
Al-Shawi R, Hafner A, Olsen J et al (2008) Neurotoxic and primes a molecular memory for brain-derived
neurotrophic roles of proNGF and the receptor sortilin neurotrophic factor protein induction in the rat hippo-
in the adult and ageing nervous system. Eur J Neurosci campus. Neuroscience 133:853–861. https://doi.org/
27:2103–2114. https://doi.org/10.1111/j.1460-9568. 10.1016/j.neuroscience.2005.03.026
2008.06152.x Bergami M, Rimondini R, Santi S et al (2008) Deletion of
An JJ, Gharami K, Liao G-Y et al (2008) Distinct role of TrkB in adult progenitors alters newborn neuron inte-
long 30 UTR BDNF mRNA in spine morphology and gration into hippocampal circuits and increases
synaptic plasticity in hippocampal neurons. Cell anxiety-like behavior. Proc Natl Acad Sci U S A
134:175–187. https://doi.org/10.1016/j.cell.2008.05. 105:15570–15575. https://doi.org/10.1073/pnas.
045 0803702105
Aroeira RI, Sebastião AM, Valente CA (2015) BDNF, via Bergami M, Vignoli B, Motori E et al (2013) TrkB signal-
truncated TrkB receptor, modulates GlyT1 and GlyT2 ing directs the incorporation of newly generated
in astrocytes. Glia 63:2181–2197. https://doi.org/10. periglomerular cells in the adult olfactory bulb. J
1002/glia.22884 Neurosci 33:11464–11478. https://doi.org/10.1523/
Babu H, Ramirez-Rodriguez G, Fabel K et al (2009) JNEUROSCI.4812-12.2013
Synaptic network activity induces neuronal differenti- Berghuis P, Agerman K, Dobszay MB et al (2006) Brain-
ation of adult hippocampal precursor cells through derived neurotrophic factor selectively regulates
BDNF signaling. Front Neurosci 3:49. https://doi.org/ dendritogenesis of parvalbumin-containing
10.3389/neuro.22.001.2009 interneurons in the main olfactory bulb through the
Bagley JA, Belluscio L (2010) Dynamic imaging reveals PLCgamma pathway. J Neurobiol 66:1437–1451.
that brain-derived neurotrophic factor can indepen- https://doi.org/10.1002/neu.20319
dently regulate motility and direction of neuroblasts Bernabeu RO, Longo FM (2010) The p75 neurotrophin
within the rostral migratory stream. Neuroscience receptor is expressed by adult mouse dentate progeni-
169:1449–1461. https://doi.org/10.1016/j.neurosci tor cells and regulates neuronal and non-neuronal cell
ence.2010.05.075 genesis. BMC Neurosci 11:136. https://doi.org/10.
Baik S-H, Rajeev V, Fann DY-W et al (2020) Intermittent 1186/1471-2202-11-136
fasting increases adult hippocampal neurogenesis. Biffo S, Offenhäuser N, Carter BD, Barde YA (1995)
Brain Behav 10:e01444. https://doi.org/10.1002/brb3. Selective binding and internalisation by truncated
1444 receptors restrict the availability of BDNF during
Baj G, Leone E, Chao MV, Tongiorgi E (2011) Spatial development. Development 121:2461–2470
segregation of BDNF transcripts enables BDNF to Birch AM, Kelly ÁM (2013) Chronic intracerebroven-
differentially shape distinct dendritic compartments. tricular infusion of nerve growth factor improves rec-
PNAS 108:16813–16818. https://doi.org/10.1073/ ognition memory in the rat. Neuropharmacology
pnas.1014168108 75:255–261. https://doi.org/10.1016/j.neuropharm.
Barker PA (2004) p75NTR is positively promiscuous: 2013.07.023
novel partners and new insights. Neuron 42:529–533. Birch AM, McGarry NB, Kelly ÁM (2013) Short-term
https://doi.org/10.1016/j.neuron.2004.04.001 environmental enrichment, in the absence of exercise,
Barrett GL, Greferath U, Barker PA et al (2005) improves memory, and increases NGF concentration,
Co-expression of the P75 neurotrophin receptor and early neuronal survival, and synaptogenesis in the den-
neurotrophin receptor-interacting melanoma antigen tate gyrus in a time-dependent manner. Hippocampus
homolog in the mature rat brain. Neuroscience 23:437–450. https://doi.org/10.1002/hipo.22103
133:381–392. https://doi.org/10.1016/j.neuroscience. Bothwell M (2014) NGF, BDNF, NT3, and NT4. Handb
2005.01.067 Exp Pharmacol 220:3–15. https://doi.org/10.1007/978-
Bath KG, Mandairon N, Jing D et al (2008) Variant brain- 3-642-45106-5_1
derived neurotrophic factor (Val66Met) alters adult Chae C-H, Jung S-L, An S-H et al (2014) Swimming
olfactory bulb neurogenesis and spontaneous olfactory exercise stimulates neuro-genesis in the subventricular
zone via increase in synapsin I and nerve growth factor
110 F. F. Ribeiro and S. Xapelli

levels. Biol Sport 31:309–314. https://doi.org/10.5604/ vesicles in brain neurons. J Cell Biol 196:775–788.
20831862.1132130 https://doi.org/10.1083/jcb.201201038
Chan JP, Cordeira J, Calderon GA et al (2008) Depletion Ding Q, Ying Z, Gómez-Pinilla F (2011) Exercise
of central BDNF in mice impedes terminal differentia- influences hippocampal plasticity by modulating
tion of new granule neurons in the adult hippocampus. BDNF processing. Neuroscience 192:773–780.
Mol Cell Neurosci 39:372–383. https://doi.org/10. https://doi.org/10.1016/j.neuroscience.2011.06.032
1016/j.mcn.2008.07.017 Donovan MH, Yamaguchi M, Eisch AJ (2008) Dynamic
Chao MV (2003) Neurotrophins and their receptors: a expression of TrkB receptor protein on proliferating
convergence point for many signalling pathways. Nat and maturing cells in the adult mouse dentate gyrus.
Rev Neurosci 4:299–309. https://doi.org/10.1038/ Hippocampus 18:435–439. https://doi.org/10.1002/
nrn1078 hipo.20410
Chen J, Li C-R, Yang H et al (2016) proBDNF Attenuates Dougherty KD, Milner TA (1999) p75NTR immunoreac-
Hippocampal Neurogenesis and Induces Learning and tivity in the rat dentate gyrus is mostly within presyn-
Memory Deficits in Aged Mice. Neurotox Res aptic profiles but is also found in some astrocytic and
29:47–53. https://doi.org/10.1007/s12640-015-9568-2 postsynaptic profiles. J Comp Neurol 407:77–91.
Cheng P-L, Song A-H, Wong Y-H et al (2011) Self- https://doi.org/10.1002/(sici)1096-9861(19990428)
amplifying autocrine actions of BDNF in axon devel- 407:1<77::aid-cne6>3.0.co;2-s
opment. Proc Natl Acad Sci U S A 108:18430–18435. Eide FF, Vining ER, Eide BL et al (1996) Naturally
https://doi.org/10.1073/pnas.1115907108 occurring truncated trkB receptors have dominant
Choi SH, Li Y, Parada LF, Sisodia SS (2009) Regulation inhibitory effects on brain-derived neurotrophic factor
of hippocampal progenitor cell survival, proliferation signaling. J Neurosci 16:3123–3129
and dendritic development by BDNF. Mol Erickson KI, Voss MW, Prakash RS et al (2011) Exercise
Neurodegener 4:52. https://doi.org/10.1186/1750- training increases size of hippocampus and improves
1326-4-52 memory. Proc Natl Acad Sci USA 108:3017–3022.
Choi SH, Bylykbashi E, Chatila ZK et al (2018) Combined https://doi.org/10.1073/pnas.1015950108
adult neurogenesis and BDNF mimic exercise effects Ferreira FF, Ribeiro FF, Rodrigues RS et al (2018) Brain-
on cognition in an Alzheimer’s mouse model. Science derived neurotrophic factor (BDNF) role in
361(6406):eaan8821. https://doi.org/10.1126/science. cannabinoid-mediated neurogenesis. Front Cell
aan8821 Neurosci 12:441. https://doi.org/10.3389/fncel.2018.
Climent E, Sancho-Tello M, Miñana R et al (2000) 00441
Astrocytes in culture express the full-length Trk-B Foltran RB, Diaz SL (2016) BDNF isoforms: a round trip
receptor and respond to brain derived neurotrophic ticket between neurogenesis and serotonin? J
factor by changing intracellular calcium levels: effect Neurochem 138:204–221. https://doi.org/10.1111/jnc.
of ethanol exposure in rats. Neurosci Lett 288:53–56 13658
Colliva A, Tongiorgi E (2020) Distinct role of 50 UTR Fonseca-Gomes J, Jerónimo-Santos A, Lesnikova A et al
sequences in dendritic trafficking of BDNF mRNA: (2019) TrkB-ICD fragment, originating from BDNF
additional mechanisms for the BDNF splice variants receptor cleavage, is translocated to cell nucleus and
spatial code. Mol Brain. https://doi.org/10.1186/ phosphorylates nuclear and axonal proteins. Front Mol
s13041-020-00680-8 Neurosci 12:4. https://doi.org/10.3389/fnmol.2019.
Conner JM, Lauterborn JC, Yan Q et al (1997) Distribu- 00004
tion of brain-derived neurotrophic factor (BDNF) pro- Frielingsdorf H, Simpson DR, Thal LJ, Pizzo DP (2007)
tein and mRNA in the normal adult rat CNS: evidence Nerve growth factor promotes survival of new neurons
for anterograde axonal transport. J Neurosci in the adult hippocampus. Neurobiol Dis 26:47–55.
17:2295–2313 https://doi.org/10.1016/j.nbd.2006.11.015
Corvaglia V, Cilli D, Scopa C et al (2019) ProNGF is a Frisén J, Verge VM, Fried K et al (1993) Characterization
cell-type-specific mitogen for adult hippocampal and of glial trkB receptors: differential response to injury in
for induced neural stem cells. Stem Cells the central and peripheral nervous systems. Proc Natl
37:1223–1237. https://doi.org/10.1002/stem.3037 Acad Sci USA 90:4971–4975
Cotman CW, Berchtold NC (2002) Exercise: a behavioral Fujii T, Kunugi H (2009) p75NTR as a therapeutic target
intervention to enhance brain health and plasticity. for neuropsychiatric diseases. Curr Mol Pharmacol
Trends Neurosci 25:295–301 2:70–76. https://doi.org/10.2174/
Delgado AC, Ferrón SR, Vicente D et al (2014) Endothe- 1874467210902010070
lial NT-3 delivered by vasculature and CSF promotes Galvão RP, Garcia-Verdugo JM, Alvarez-Buylla A (2008)
quiescence of subependymal neural stem cells through Brain-derived neurotrophic factor signaling does not
nitric oxide induction. Neuron 83:572–585. https://doi. stimulate subventricular zone neurogenesis in adult
org/10.1016/j.neuron.2014.06.015 mice and rats. J Neurosci 28:13368–13383. https://
Dieni S, Matsumoto T, Dekkers M et al (2012) BDNF and doi.org/10.1523/JNEUROSCI.2918-08.2008
its pro-peptide are stored in presynaptic dense core Gao WQ, Zheng JL, Karihaloo M (1995) Neurotrophin-4/
5 (NT-4/5) and brain-derived neurotrophic factor
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 111

(BDNF) act at later stages of cerebellar granule cell via astrocytic TrkB.T1. eLife 8:e44667. https://doi.org/
differentiation. J Neurosci 15(4):2656–2667. https:// 10.7554/eLife.44667
pubmed.ncbi.nlm.nih.gov/7722620/. Accessed 10 Sept Hosomi S, Yamashita T, Aoki M, Tohyama M (2003) The
2020 p75 receptor is required for BDNF-induced differenti-
Gao X, Smith GM, Chen J (2009) Impaired dendritic ation of neural precursor cells. Biochem Biophys Res
development and synaptic formation of postnatal- Commun 301:1011–1015
born dentate gyrus granular neurons in the absence of Huang EJ, Reichardt LF (2001) Neurotrophins: roles in
brain-derived neurotrophic factor signaling. Exp neuronal development and function. Annu Rev
Neurol 215:178–190. https://doi.org/10.1016/j. Neurosci 24:677–736. https://doi.org/10.1146/
expneurol.2008.10.009 annurev.neuro.24.1.677
Gascon E, Vutskits L, Zhang H et al (2005) Sequential Hwang JJ, Park M-H, Choi S-Y, Koh J-Y (2005) Activa-
activation of p75 and TrkB is involved in dendritic tion of the Trk signaling pathway by extracellular zinc.
development of subventricular zone-derived neuronal Role of metalloproteinases. J Biol Chem
progenitors in vitro. Eur J Neurosci 21:69–80. https:// 280:11995–12001. https://doi.org/10.1074/jbc.
doi.org/10.1111/j.1460-9568.2004.03849.x M403172200
Grade S, Weng YC, Snapyan M et al (2013) Brain-derived Ip N, Li Y, Yancopoulos G, Lindsay R (1993) Cultured
neurotrophic factor promotes vasculature-associated hippocampal neurons show responses to BDNF, NT-3,
migration of neuronal precursors toward the ischemic and NT-4, but not NGF. J Neurosci 13:3394–3405.
striatum. PLoS One 8:e55039. https://doi.org/10.1371/ https://doi.org/10.1523/JNEUROSCI.13-08-03394.
journal.pone.0055039 1993
Groves N, O’Keeffe I, Lee W et al (2019) Blockade of Islam O, Loo TX, Heese K (2009) Brain-derived
TrkB but not p75NTR activates a subpopulation of neurotrophic factor (BDNF) has proliferative effects
quiescent neural precursor cells and enhances on neural stem cells through the truncated TRK-B
neurogenesis in the adult mouse hippocampus. Dev receptor, MAP kinase, AKT, and STAT-3 signaling
Neurobiol 79:868–879. https://doi.org/10.1002/dneu. pathways. Curr Neurovasc Res 6:42–53
22729 Ivanova T, Beyer C (2001) Pre- and postnatal expression
Guo J, Wang J, Zhang Z et al (2013) proNGF inhibits of brain-derived neurotrophic factor mRNA/protein
neurogenesis and induces glial activation in adult and tyrosine protein kinase receptor B mRNA in the
mouse dentate gyrus. Neurochem Res 38:1695–1703. mouse hippocampus. Neurosci Lett 307:21–24. https://
https://doi.org/10.1007/s11064-013-1071-7 doi.org/10.1016/s0304-3940(01)01905-x
Haapasalo A, Sipola I, Larsson K et al (2002) Regulation Jansen P, Giehl K, Nyengaard JR et al (2007) Roles for the
of TRKB surface expression by brain-derived pro-neurotrophin receptor sortilin in neuronal develop-
neurotrophic factor and truncated TRKB isoforms. J ment, aging and brain injury. Nat Neurosci
Biol Chem 277:43160–43167. https://doi.org/10.1074/ 10:1449–1457. https://doi.org/10.1038/nn2000
jbc.M205202200 Jerónimo-Santos A, Vaz SH, Parreira S et al (2015)
Halban PA, Irminger JC (1994) Sorting and processing of Dysregulation of TrkB receptors and BDNF function
secretory proteins. Biochem J 299(Pt 1):1–18 by amyloid-β peptide is mediated by calpain. Cereb
Hanson ND, Owens MJ, Nemeroff CB (2011) Depression, Cortex 25:3107–3121. https://doi.org/10.1093/cercor/
antidepressants, and neurogenesis: a critical reap- bhu105
praisal. Neuropsychopharmacology 36:2589–2602. Jha S, Dong B, Sakata K (2011) Enriched environment
https://doi.org/10.1038/npp.2011.220 treatment reverses depression-like behavior and
Henry RA, Hughes SM, Connor B (2007) AAV-mediated restores reduced hippocampal neurogenesis and pro-
delivery of BDNF augments neurogenesis in the nor- tein levels of brain-derived neurotrophic factor in mice
mal and quinolinic acid-lesioned adult rat brain. Eur J lacking its expression through promoter IV. Transl
Neurosci 25:3513–3525. https://doi.org/10.1111/j. Psychiatry 1:e40. https://doi.org/10.1038/tp.2011.33
1460-9568.2007.05625.x Jin J-J, Ko I-G, Kim S-E et al (2017) Age-dependent
Hernandez-Echeagaray E (2020) The role of the TrkB-T1 differences of treadmill exercise on spatial learning
receptor in the neurotrophin-4/5 antagonism of brain- ability between young- and adult-age rats. J Exerc
derived neurotrophic factor on corticostriatal synaptic Rehabil 13:381–386. https://doi.org/10.12965/jer.
transmission. Neural Regen Res 15:1973–1976. 1735070.535
https://doi.org/10.4103/1673-5374.282224 Katoh-Semba R, Takeuchi IK, Semba R, Kato K (1997)
Hock C, Heese K, Hulette C et al (2000) Region-specific Distribution of brain-derived neurotrophic factor in rats
neurotrophin imbalances in Alzheimer disease: and its changes with development in the brain. J
decreased levels of brain-derived neurotrophic factor Neurochem 69:34–42
and increased levels of nerve growth factor in hippo- Kim K, Sung Y-H, Seo J-H et al (2015) Effects of tread-
campus and cortical areas. Arch Neurol 57:846–851. mill exercise-intensity on short-term memory in the
https://doi.org/10.1001/archneur.57.6.846 rats born of the lipopolysaccharide-exposed maternal
Holt LM, Hernandez RD, Pacheco NL et al (2019) Astro- rats. J Exerc Rehabil 11:296–302. https://doi.org/10.
cyte morphogenesis is dependent on BDNF signaling 12965/jer.150264
112 F. F. Ribeiro and S. Xapelli

Kirschenbaum B, Goldman SA (1995) Brain-derived Commun 8:1668. https://doi.org/10.1038/s41467-


neurotrophic factor promotes the survival of neurons 017-01709-8
arising from the adult rat forebrain subependymal Maisonpierre PC, Belluscio L, Friedman B et al (1990)
zone. Proc Natl Acad Sci USA 92:210–214 NT-3, BDNF, and NGF in the developing rat nervous
Klein R, Conway D, Parada LF, Barbacid M (1990) The system: parallel as well as reciprocal patterns of
trkB tyrosine protein kinase gene codes for a second expression. Neuron 5:501–509. https://doi.org/10.
neurogenic receptor that lacks the catalytic kinase 1016/0896-6273(90)90089-x
domain. Cell 61:647–656. https://doi.org/10.1016/ Marlatt MW, Potter MC, Lucassen PJ, van Praag H (2012)
0092-8674(90)90476-U Running throughout middle-age improves memory
Kobilo T, Liu Q-R, Gandhi K et al (2011) Running is the function, hippocampal neurogenesis, and BDNF levels
neurogenic and neurotrophic stimulus in environmen- in female C57BL/6J mice. Dev Neurobiol 72:943–952.
tal enrichment. Learn Mem 18:605–609. https://doi. https://doi.org/10.1002/dneu.22009
org/10.1101/lm.2283011 Matyas JJ, O’Driscoll CM, Yu L et al (2017) Truncated
Kolbeck R, Jungbluth S, Barde Y-A (1994) TrkB.T1-mediated astrocyte dysfunction contributes to
Characterisation of neurotrophin dimers and impaired motor function and neuropathic pain after
monomers. Eur J Biochem 225:995–1003. https://doi. spinal cord injury. J Neurosci 37:3956–3971. https://
org/10.1111/j.1432-1033.1994.0995b.x doi.org/10.1523/JNEUROSCI.3353-16.2017
Kuzumaki N, Ikegami D, Tamura R et al (2011) Hippo- Maynard KR, Hobbs JW, Sukumar M et al (2017) Bdnf
campal epigenetic modification at the brain-derived mRNA splice variants differentially impact CA1 and
neurotrophic factor gene induced by an enriched envi- CA3 dendrite complexity and spine morphology in the
ronment. Hippocampus 21:127–132. https://doi.org/ hippocampus. Brain Struct Funct 222:3295–3307.
10.1002/hipo.20775 https://doi.org/10.1007/s00429-017-1405-3
Lau AG, Irier HA, Gu J et al (2010) Distinct 30 UTRs McDole B, Isgor C, Pare C, Guthrie K (2015) BDNF over-
differentially regulate activity-dependent translation expression increases olfactory bulb granule cell den-
of brain-derived neurotrophic factor (BDNF). Proc dritic spine density in vivo. Neuroscience
Natl Acad Sci USA 107:15945–15950. https://doi. 304:146–160. https://doi.org/10.1016/j.neuroscience.
org/10.1073/pnas.1002929107 2015.07.056
Lee R, Kermani P, Teng KK, Hempstead BL (2001) Reg- McDole B, Berger R, Guthrie K (2020) Genetic increases
ulation of cell survival by secreted proneurotrophins. in olfactory Bulb BDNF do not enhance survival of
Science 294:1945–1948. https://doi.org/10.1126/sci adult-born granule cells. Chem Senses 45:3–13. https://
ence.1065057 doi.org/10.1093/chemse/bjz058
Lee J, Seroogy KB, Mattson MP (2002) Dietary restriction Meeker R, Williams K (2014) Dynamic nature of the p75
enhances neurotrophin expression and neurogenesis in neurotrophin receptor in response to injury and disease.
the hippocampus of adult mice. J Neurochem J Neuroimmune Pharmacol 9:615–628. https://doi.org/
80:539–547 10.1007/s11481-014-9566-9
Lessmann V, Gottmann K, Malcangio M (2003) Middlemas DS, Lindberg RA, Hunter T (1991) trkB, a
Neurotrophin secretion: current facts and future neural receptor protein-tyrosine kinase: evidence for a
prospects. Prog Neurobiol 69:341–374 full-length and two truncated receptors. Mol Cell Biol
Li Y, Luikart BW, Birnbaum S et al (2008) TrkB regulates 11:143–153. https://doi.org/10.1128/MCB.11.1.143
hippocampal neurogenesis and governs sensitivity to Mizoguchi H, Nakade J, Tachibana M et al (2011) Matrix
antidepressive treatment. Neuron 59:399–412. https:// metalloproteinase-9 contributes to kindled seizure
doi.org/10.1016/j.neuron.2008.06.023 development in pentylenetetrazole-treated mice by
Linnarsson S, Willson CA, Ernfors P (2000) Cell death in converting pro-BDNF to mature BDNF in the hippo-
regenerating populations of neurons in BDNF mutant campus. J Neurosci 31:12963–12971. https://doi.org/
mice. Brain Res Mol Brain Res 75:61–69 10.1523/JNEUROSCI.3118-11.2011
Lu J-J, Yang M, Sun Y, Zhou X-F (2014) Synthesis, Mizui T et al (2017) BDNF pro-peptide: a novel synaptic
trafficking and release of BDNF. In: Kostrzewa RM modulator generated as an N-terminal fragment from
(ed) Handbook of neurotoxicity. Springer, New York, the BDNF precursor by proteolytic processing. Neural
pp 1955–1971 Regen Res 12(7):1024–1027. http://www.nrronline.
Luberg K, Wong J, Weickert CS, Timmusk T (2010) o r g / a r t i c l e . a s p ? i s s n ¼1 6 7 3 - 5 3 7 4 ; y e a r ¼2 0 1 7 ;
Human TrkB gene: novel alternative transcripts, pro- volume¼12;issue¼7;spage¼1024;epage¼1027;
tein isoforms and expression pattern in the prefrontal aulast¼Mizui. Accessed 9 Sept 2020
cerebral cortex during postnatal development. J Mowla SJ, Pareek S, Farhadi HF et al (1999) Differential
Neurochem 113:952–964. https://doi.org/10.1111/j. sorting of nerve growth factor and brain-derived
1471-4159.2010.06662.x neurotrophic factor in hippocampal neurons. J
Ma Z, Zang T, Birnbaum SG et al (2017) TrkB dependent Neurosci 19:2069–2080
adult hippocampal progenitor differentiation mediates Mowla SJ, Farhadi HF, Pareek S et al (2001) Biosynthesis
sustained ketamine antidepressant response. Nat and post-translational processing of the precursor to
brain-derived neurotrophic factor. J Biol Chem
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 113

276:12660–12666. https://doi.org/10.1074/jbc. Neurobiol Dis 77:220–227. https://doi.org/10.1016/j.


M008104200 nbd.2015.03.011
Nagappan G, Zaitsev E, Senatorov VV et al (2009) Con- Ribeiro FF (2017) Unveiling the trophic actions of adeno-
trol of extracellular cleavage of ProBDNF by high sine A2A receptors in neurite outgrowth and postnatal
frequency neuronal activity. Proc Natl Acad Sci USA neurogenesis: interaction with brain-derived
106:1267–1272. https://doi.org/10.1073/pnas. neurotrophic factor. PhD thesis. Universidade de
0807322106 Lisboa, Lisbon, Portugal
Neeper SA, Gómez-Pinilla F, Choi J, Cotman C (1995) Rose CR, Blum R, Pichler B et al (2003) Truncated TrkB-
Exercise and brain neurotrophins. Nature 373:109. T1 mediates neurotrophin-evoked calcium signalling
https://doi.org/10.1038/373109a0 in glia cells. Nature 426:74–78. https://doi.org/10.
Nibuya M, Morinobu S, Duman RS (1995) Regulation of 1038/nature01983
BDNF and trkB mRNA in rat brain by chronic electro- Rossi C, Angelucci A, Costantin L et al (2006) Brain-
convulsive seizure and antidepressant drug treatments. derived neurotrophic factor (BDNF) is required for
J Neurosci 15:7539–7547 the enhancement of hippocampal neurogenesis follow-
Nykjaer A, Lee R, Teng KK et al (2004) Sortilin is essen- ing environmental enrichment. Eur J Neurosci
tial for proNGF-induced neuronal cell death. Nature 24:1850–1856. https://doi.org/10.1111/j.1460-9568.
427:843–848. https://doi.org/10.1038/nature02319 2006.05059.x
Ohira K, Kumanogoh H, Sahara Y et al (2005) A truncated Rudge JS, Li Y, Pasnikowski EM et al (1994)
tropomyosin-related kinase B receptor, T1, regulates Neurotrophic factor receptors and their signal transduc-
glial cell morphology via Rho GDP dissociation inhib- tion capabilities in rat astrocytes. Eur J Neurosci
itor 1. J Neurosci 25:1343–1353. https://doi.org/10. 6:693–705
1523/JNEUROSCI.4436-04.2005 Russo-Neustadt AA, Beard RC, Huang YM, Cotman CW
Ortiz-López L, Vega-Rivera NM, Babu H, Ramírez- (2000) Physical activity and antidepressant treatment
Rodríguez GB (2017) Brain-derived neurotrophic fac- potentiate the expression of specific brain-derived
tor induces cell survival and the migration of murine neurotrophic factor transcripts in the rat hippocampus.
adult hippocampal precursor cells during differentia- Neuroscience 101:305–312
tion in vitro. Neurotox Res 31:122–135. https://doi. Sairanen M, Lucas G, Ernfors P et al (2005) Brain-derived
org/10.1007/s12640-016-9673-x neurotrophic factor and antidepressant drugs have dif-
Pang PT, Teng HK, Zaitsev E et al (2004) Cleavage of ferent but coordinated effects on neuronal turnover,
proBDNF by tPA/plasmin is essential for long-term proliferation, and survival in the adult dentate gyrus.
hippocampal plasticity. Science 306:487–491. https:// J Neurosci 25:1089–1094. https://doi.org/10.1523/
doi.org/10.1126/science.1100135 JNEUROSCI.3741-04.2005
Pencea V, Bingaman KD, Wiegand SJ, Luskin MB (2001) Sakata K, Woo NH, Martinowich K et al (2009) Critical
Infusion of brain-derived neurotrophic factor into the role of promoter IV-driven BDNF transcription in
lateral ventricle of the adult rat leads to new neurons in GABAergic transmission and synaptic plasticity in
the parenchyma of the striatum, septum, thalamus, and the prefrontal cortex. Proc Natl Acad Sci U S A
hypothalamus. J Neurosci 21:6706–6717 106:5942–5947. https://doi.org/10.1073/pnas.
Peng S, Wuu J, Mufson EJ, Fahnestock M (2004) 0811431106
Increased proNGF levels in subjects with mild cogni- Sarret P, Krzywkowski P, Segal L et al (2003) Distribution
tive impairment and mild Alzheimer disease. J of NTS3 receptor/sortilin mRNA and protein in the rat
Neuropathol Exp Neurol 63:641–649. https://doi.org/ central nervous system. J Comp Neurol 461:483–505.
10.1093/jnen/63.6.641 https://doi.org/10.1002/cne.10708
Pruunsild P, Kazantseva A, Aid T et al (2007) Dissecting Scharfman H, Goodman J, Macleod A et al (2005)
the human BDNF locus: bidirectional transcription, Increased neurogenesis and the ectopic granule cells
complex splicing, and multiple promoters. Genomics after intrahippocampal BDNF infusion in adult rats.
90:397–406. https://doi.org/10.1016/j.ygeno.2007.05. Exp Neurol 192:348–356. https://doi.org/10.1016/j.
004 expneurol.2004.11.016
Reichardt LF (2003) Neurobiology: signals that make Segawa M, Morinobu S, Matsumoto T et al (2013) Elec-
waves. Nature 426:25–26. https://doi.org/10.1038/ troconvulsive seizure, but not imipramine, rapidly
426025a up-regulates pro-BDNF and t-PA, leading to mature
Reichardt LF (2006) Neurotrophin-regulated signalling BDNF production, in the rat hippocampus. Int J
pathways. Philos Trans R Soc Lond, B, Biol Sci Neuropsychopharmacol 16:339–350. https://doi.org/
361:1545–1564. https://doi.org/10.1098/rstb.2006. 10.1017/S1461145712000053
1894 Seidah NG, Benjannet S, Pareek S et al (1996) Cellular
Reinhart V, Bove SE, Volfson D et al (2015) Evaluation of processing of the nerve growth factor precursor by the
TrkB and BDNF transcripts in prefrontal cortex, hip- mammalian pro-protein convertases. Biochem J 314
pocampus, and striatum from subjects with schizophre- (Pt 3):951–960
nia, bipolar disorder, and major depressive disorder. Shelly M, Cancedda L, Heilshorn S et al (2007) LKB1/
STRAD promotes axon initiation during neuronal
114 F. F. Ribeiro and S. Xapelli

polarization. Cell 129:565–577. https://doi.org/10. Torres-Cruz FM, Mendoza E, Vivar-Cortés IC et al (2019)


1016/j.cell.2007.04.012 Do BDNF and NT-4/5 exert synergistic or occlusive
Shimazu K, Zhao M, Sakata K et al (2006) NT-3 facilitates effects on corticostriatal transmission in a male mouse
hippocampal plasticity and learning and memory by model of Huntington’s disease? J Neurosci Res
regulating neurogenesis. Learn Mem 13:307–315. 97:1665–1677. https://doi.org/10.1002/jnr.24507
https://doi.org/10.1101/lm.76006 Vaegter CB, Jansen P, Fjorback AW et al (2011) Sortilin
Shirayama Y, Chen AC-H, Nakagawa S et al (2002) associates with Trk receptors to enhance anterograde
Brain-derived neurotrophic factor produces antidepres- transport and signaling by neurotrophins. Nat Neurosci
sant effects in behavioral models of depression. J 14. https://doi.org/10.1038/nn.2689
Neurosci 22:3251–3261. https://doi.org/10.1523/ van Praag H, Kempermann G, Gage FH (1999) Running
JNEUROSCI.22-08-03251.2002 increases cell proliferation and neurogenesis in the
Smith MA, Zhang LX, Lyons WE, Mamounas LA (1997) adult mouse dentate gyrus. Nat Neurosci 2:266–270.
Anterograde transport of endogenous brain-derived https://doi.org/10.1038/6368
neurotrophic factor in hippocampal mossy fibers. Vaynman S, Ying Z, Gomez-Pinilla F (2004) Hippocam-
Neuroreport 8:1829–1834. https://doi.org/10.1097/ pal BDNF mediates the efficacy of exercise on synaptic
00001756-199705260-00008 plasticity and cognition. Eur J Neurosci 20:2580–2590.
Snapyan M, Lemasson M, Brill MS et al (2009) Vascula- https://doi.org/10.1111/j.1460-9568.2004.03720.x
ture guides migrating neuronal precursors in the adult Vaynman SS, Ying Z, Yin D, Gomez-Pinilla F (2006)
mammalian forebrain via brain-derived neurotrophic Exercise differentially regulates synaptic proteins
factor signaling. J Neurosci 29:4172–4188. https:// associated to the function of BDNF. Brain Res
doi.org/10.1523/JNEUROSCI.4956-08.2009 1070:124–130. https://doi.org/10.1016/j.brainres.
Stoilov P, Castren E, Stamm S (2002) Analysis of the 2005.11.062
human TrkB gene genomic organization reveals Vaz SH, Jørgensen TN, Cristóvão-Ferreira S et al (2011)
novel TrkB isoforms, unusual gene length, and splic- Brain-derived neurotrophic factor (BDNF) enhances
ing mechanism. Biochem Biophys Res Commun GABA transport by modulating the trafficking of
290:1054–1065. https://doi.org/10.1006/bbrc.2001. GABA transporter-1 (GAT-1) from the plasma mem-
6301 brane of rat cortical astrocytes. J Biol Chem
Sun Y, Lim Y, Li F et al (2012) ProBDNF collapses 286:40464–40476. https://doi.org/10.1074/jbc.M111.
neurite outgrowth of primary neurons by activating 232009
RhoA. PLoS One 7:e35883. https://doi.org/10.1371/ Vilar M, Mira H (2016) Regulation of neurogenesis by
journal.pone.0035883 neurotrophins during adulthood: expected and unex-
Taliaz D, Stall N, Dar DE, Zangen A (2010) Knockdown pected roles. Front Neurosci 10:26. https://doi.org/10.
of brain-derived neurotrophic factor in specific brain 3389/fnins.2016.00026
sites precipitates behaviors associated with depression Wang L, Chang X, She L et al (2015) Autocrine action of
and reduces neurogenesis. Mol Psychiatry 15:80–92. BDNF on dendrite development of adult-born hippo-
https://doi.org/10.1038/mp.2009.67 campal neurons. J Neurosci 35:8384–8393. https://doi.
Tanqueiro SR, Ramalho RM, Rodrigues TM et al (2018) org/10.1523/JNEUROSCI.4682-14.2015
Inhibition of NMDA receptors prevents the loss of Waterhouse EG, An JJ, Orefice LL et al (2012) BDNF
BDNF function induced by amyloid β. Front promotes differentiation and maturation of adult-born
Pharmacol 9:237. https://doi.org/10.3389/fphar.2018. neurons through GABAergic transmission. J Neurosci
00237 32:14318–14330. https://doi.org/10.1523/
Teng HK, Teng KK, Lee R et al (2005) ProBDNF induces JNEUROSCI.0709-12.2012
neuronal apoptosis via activation of a receptor complex Webster MJ, Herman MM, Kleinman JE, Shannon
of p75NTR and sortilin. J Neurosci 25:5455–5463. Weickert C (2006) BDNF and trkB mRNA expression
https://doi.org/10.1523/JNEUROSCI.5123-04.2005 in the hippocampus and temporal cortex during the
Terry AV, Kutiyanawalla A, Pillai A (2011) human lifespan. Gene Expr Patterns 6:941–951.
Age-dependent alterations in nerve growth factor https://doi.org/10.1016/j.modgep.2006.03.009
(NGF)-related proteins, sortilin, and learning and Werry EL, Enjeti S, Halliday GM et al (2010) Effect of age
memory in rats. Physiol Behav 102:149–157. https:// on proliferation-regulating factors in human adult neu-
doi.org/10.1016/j.physbeh.2010.11.005 rogenic regions. J Neurochem 115:956–964. https://
Tervonen TA, Ajamian F, Wit JD et al (2006) doi.org/10.1111/j.1471-4159.2010.06992.x
Overexpression of a truncated TrkB isoform increases Woo NH, Teng HK, Siao C-J et al (2005) Activation of
the proliferation of neural progenitors. Eur J Neurosci p75NTR by proBDNF facilitates hippocampal long-
24:1277–1285. https://doi.org/10.1111/j.1460-9568. term depression. Nat Neurosci 8:1069–1077. https://
2006.05010.x doi.org/10.1038/nn1510
Tonchev AB (2011) Brain ischemia, neurogenesis, and Xu S-Y, Zhang Q-L, Zhang Q et al (2019) Regional and
neurotrophic receptor expression in primates. Arch cellular mapping of sortilin immunoreactivity in adult
Ital Biol 149:225–231 human brain. Front Neuroanat 13:31. https://doi.org/
10.3389/fnana.2019.00031
8 Intervention of Brain-Derived Neurotrophic Factor and Other Neurotrophins. . . 115

Yan Q, Radeke MJ, Matheson CR et al (1997) Immunocy- Zhang H, He X, Mei Y, Ling Q (2018) Ablation of ErbB4
tochemical localization of TrkB in the central nervous in parvalbumin-positive interneurons inhibits adult
system of the adult rat. J Comp Neurol 378:135–157 hippocampal neurogenesis through down-regulating
Yang J, Siao C-J, Nagappan G et al (2009) Neuronal BDNF/TrkB expression. J Comp Neurol
release of proBDNF. Nat Neurosci 12:113–115. 526:2482–2492. https://doi.org/10.1002/cne.24506
https://doi.org/10.1038/nn.2244 Zhu W, Cheng S, Xu G et al (2011) Intranasal nerve
Yang J, Harte-Hargrove LC, Siao C-J et al (2014) growth factor enhances striatal neurogenesis in adult
proBDNF negatively regulates neuronal remodeling, rats with focal cerebral ischemia. Drug Deliv
synaptic transmission, and synaptic plasticity in hippo- 18:338–343. https://doi.org/10.3109/10717544.2011.
campus. Cell Rep 7:796–806. https://doi.org/10.1016/ 557785
j.celrep.2014.03.040 Zigova T, Pencea V, Wiegand SJ, Luskin MB (1998)
Young KM, Merson TD, Sotthibundhu A et al (2007) p75 Intraventricular administration of BDNF increases the
neurotrophin receptor expression defines a population number of newly generated neurons in the adult olfac-
of BDNF-responsive neurogenic precursor cells. J tory bulb. Mol Cell Neurosci 11:234–245. https://doi.
Neurosci 27:5146–5155. https://doi.org/10.1523/ org/10.1006/mcne.1998.0684
JNEUROSCI.0654-07.2007 Zuccaro E, Bergami M, Vignoli B et al (2014) Polarized
Zagrebelsky M, Holz A, Dechant G et al (2005) The p75 expression of p75(NTR) specifies axons during devel-
neurotrophin receptor negatively modulates dendrite opment and adult neurogenesis. Cell Rep 7:138–152.
complexity and spine density in hippocampal neurons. https://doi.org/10.1016/j.celrep.2014.02.039
J Neurosci 25:9989–9999. https://doi.org/10.1523/
JNEUROSCI.2492-05.2005
Part III
Alzheimer and Central Nervous System
Rita Levi-Montalcini, NGF Metabolism
in Health and in the Alzheimer’s 9
Pathology

A. Claudio Cuello

My life has been enriched by excellent human relations, work, and interests. I have never
felt lonely.
Rita Levi-Montalcini

Abstract found enhanced protein levels and enzymatic


activity of the proteases responsible for the
This chapter relates biographic personal and
proteolytic degradation of mNGF. A biochem-
scientific interactions with Rita Levi-
ical prospect explaining the tropic factor vul-
Montalcini. It highlights research from our
nerability of the NGF-dependent basal
laboratory inspired by Rita’s fundamental dis-
forebrain cholinergic neurons and of their syn-
covery. This work from studies on potentially
aptic terminals. The NGF deregulation of this
neuro-reparative gangliosides, their
metabolic pathway is evident at preclinical
interactions with NGF, the role of exogenous
stages and reflected in body fluid particularly
NGF in the recovery of degenerating choliner-
in the cerebrospinal fluid (CSF). The findings
gic neurons of the basal forebrain to the evi-
of a deregulation of the NGF metabolic path-
dence that endogenous NGF maintains the
way and its reflection in plasma and CSF are
“day-to-day” cortical synaptic phenotype and
opening doors for the development of novel
the discovery of a novel CNS “NGF metabolic
biomarkers for preclinical detection of AD
pathway.” This brain pathway’s conceptual
pathology both in Alzheimer’s and in Down
platform allowed the investigation of its status
syndrome (DS) with “silent” AD pathology.
during the Alzheimer’s disease
(AD) pathology. This revealed a major com-
promise of the conversion of the NGF precur-
9.1 Preface
sor molecule (proNGF) into the most
biologically active molecule, mature NGF
I wish to begin this chapter by congratulating my
(mNGF). Furthermore, in this pathology, we
colleagues Luigi Aloe and Laura Calza for
keeping Rita’s legacy alive, by promoting excit-
A. C. Cuello (*) ing and enriching NGF-related meetings and by
Department of Pharmacology and Therapeutics, McGill allowing us to write reviews of our work with
University, Montreal, QC, Canada
commentaries in the first person regarding our
e-mail: claudio.cuello@mcgill.ca

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 119
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_9
120 A. C. Cuello

stories and interactions with Rita, stories that processes induced by “a protein factor” (Levi-
inspired our work for many subsequent years. Montalcini 1964).
The excellence in human relations, sharing Eduardo De Robertis was the son of Italian
research interests, as signaled by Rita’s quotation, immigrants with humble origins. He made signif-
is reflected in this account as are the icant contributions to cell and neurobiology
investigations on trophic responses in health and starting in the United States and became the
disease. Chair of the Department and Director of a Cell
Biology Research Institute at the Buenos Aires
University, where in 1962 the first isolation of
9.2 What I Knew About Rita During synaptic vesicles took place (De Robertis et al.
My Early Immersion 1962). As selected medical undergraduates, and
in the Neurosciences as Departmental TAs, we had the privilege of
having access to labs, making microscope
Rita’s quotation has guided my narrative; it is preparations and samples for histology and neu-
largely in the first person and it brings to forum roanatomy teachings, and assisting with access to
the significance of our friendship and the impact advanced techniques in the pioneering years of
of her discovery of NGF to Neurosciences. This electron microscopy. Near the completion of my
discovery led to the finding of a family of medical studies, in 1965, Maria Teresa Sabatini
neurotrophins and of their corresponding from the “De Robertis” Institute collaborated at a
receptors. Besides the neurotrophins, other tro- distance with Pietro (“Piero”) Angeletti, from
phic factors were found consequently, such as Rita’s research group, on the effects of NGF
the epidermal, fibroblast, and the ciliary trophic immunoneutralization, a work which was
factors, among others. published the following year in a non-very visible
Before getting to know Rita, I knew that she Italian journal (Sabatini et al. 1966).
was from the laboratory of Giuseppe Levi at It was only afterwards during my research
Torino University, a lab which trained scientists years in Cambridge and Oxford that I learnt
like Rita, Salvador Luria, and Renato Dulbecco— more details about the fascinating life Odyssey
all of whom received the Nobel Prize. My first of Rita and her further contributions. During my
mentor in Argentina, Eduardo De Robertis, dis- Cambridge period, first as a post-doctoral fellow,
coverer of the synaptic vesicles, was an admirer and later as an MRC scientific staff at Les
of Giuseppe Levi and his histology book was Iversen’s Unit, I followed Les’ pioneering
prominent in the library of our department at the collaborations with Hans Thoenen and Ian
Buenos Aires University Medical School. Hendry on the retrograde transport of NGF
During my initial lab training at medical (Hendry et al. 1974).
school and in later years, I transited through for- Like many neuroscience colleagues, I knew
mal research in neuroanatomy, electron micros- about the evolution of Rita’s seminal work with
copy, neuroendocrinology, catecholamines, novel Viktor Hamburger on the development of the
neuroactive peptides, the CNS cholinergic sys- chick embryo (Hamburger and Levi-Montalcini
tem, and ultimately Alzheimer’s pathology. 1949) which led to the discovery of NGF,
Throughout that route, I became aware of Rita’s initially identified as a “proteinaceous factor,” a
discovery of NGF and of the saga of Torino, saga well narrated by Luigi Aloe in 2004 (Aloe
Belgium, Rio de Janeiro, and St Louis (USA). 2004). In later years, well into my “auto-exile”
My first awareness of the NGF neurobiology to England and through the long-lasting and
came when I was still a medical student and a inspiring friendship with Giancarlo Pepeu
TA at the “De Robertis Institute” of the Univer- (Firenze) and with Ezio Giacobini (Illinois,
sity of Buenos Aires. By then, we all knew about USA and Geneva, Switzerland), I gained the
her spectacular 1964 paper in Science revealing full picture of Rita Levi-Montalcini, the persona
the classical “halo” of growing neuronal and the scientist.
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 121

During my chats with Ezio, Giancarlo, and wanted to collaborate with us given—in her
many Italian colleagues, I realized just how words—our well-recognized expertise in
“heroic” and unusual Rita’s story was. I came to immunohistochemistry.
know about Rita’s clandestine work in Italy, the After she had explained the project in detail, I
paper contradicting Hamburger’s interpretations told Rita that I found the project straightforward
on the influence of the limbs on the development and that I would be pleased to welcome to my lab
of the nervous system, “the axonal bridges” in one of her collaborators, to perform the work
particular. That paper was published with her past under my guidance at our Oxford laboratory.
mentor, Giuseppe Levi, “en francais,” in a rather She agreed and arranged for her close collabora-
obscure Belgian journal, in the middle of WWII tor Luigi Aloe to come to our lab. I then had the
(Levi-Montalcini 1997)! pleasure of having daily interactions with Luigi
The above publication, which could have eas- and practicing with him mi povero italiano. Luigi
ily been lost . . . provoked—after the war years— spent some time in Oxford doing his first hands-
Viktor Hamburger to invite Rita to come to his on experience in immunohistochemistry. He
lab in St. Louis to study the problem together to returned to Rome with a new expertise and with
resolve their differences. . . This was quite an a provision of reagents as well as recently devel-
extraordinary gesture, a gesture that nowadays oped monoclonal antibodies. A few months
would be rare to find. The move to St Louis elapsed when I received by mail a draft of a
offered Rita an excellent platform for applying paper intended for PNAS authored by Levi-
her talents to the outmost, establishing very pro- Montalcini, Luigi Aloe, and A. Claudio Cuello.
ductive and transformational collaborations with I made my corrections and edits to the paper,
Viktor Hamburger and Stanley Cohen. These crossing out my name in the authorship list and
studies built an entire new conceptual platform mailed it back. When the paper arrived back to
in the neurosciences. Rome, I received a call from Rita thanking me for
Knowing all the above, I never suspected that the corrections and she asked me why I had
Rita Levi-Montalcini, a person of my continued crossed out my name from the authorship list.
admiration, would ever become one of my close Somewhat irritated she asked if I did not like the
and personal friends. work (no te è piaciuto il lavoro). I responded that
it was a nice piece of work but that I felt I did not
have any intellectual participation in it, that my
9.3 My Encounters with Rita contribution was simply to instruct Luigi in
immunohistochemical procedures, and that it
My first encounter with Rita was most unusual. I was for me a privilege to assist her. A long silence
was, by then, in my Oxford lab, doing stereotactic followed broken by the sentence: Nothing like
surgery, when I heard a voice from behind me this has happened to me before.
saying: “Are you Dr Cuello?” Concentrated on The study with my modest “ghost” participa-
my task, I simple replied, “Yes,” without turning tion was ultimately published in PNAS 1985
around. The voice behind me continued, “I am (Levi-Montalcini and Aloe 1985). The above epi-
Rita Levi-Montalcini I wanted to consult with sode was the beginning of long and close friend-
you about a research project.” It goes without ship. Rita opened her heart to us and likewise my
saying that I interrupted my work, took my lab wife, Martha, and I shared with her all our dreams
coat out, and invited Rita to continue the conver- and expectations. She narrated to us the most
sation in my office. There, I indicated to her how significant aspects of her very rich, fascinating,
pleased I was in meeting her. I then asked Rita and inspiring life. We visited each other’s homes
about her interests. She told me that she wanted to in Rome and from our side her visitation in
study the effects of NGF on the phenotypic Oxford and later in Montreal. She knew our
changes during the development of the nervous daughters Paula and Karina well and we became
system in the Xenopus and expressed that she close to her sister Paola. Paola was an artist. She
122 A. C. Cuello

Fig. 9.1 Paola and Rita


posing for us in the balcony
of their apartment in Rome,
which overlooked
“Mussolini’s villa”
in Rome

was a very gentle and petite person who looked discussed literature, history, and politics. She
and dressed like a fashionable character from the wrote several non-scientific books in Italian and
late 1800s . On one occasion, she let us see her I have the entire collection at home with her
work. In the expectation of seeing delicate, mini- signature (Tempo di mutanti, Abbi il coraggio di
ature, paintings, we were most surprised to see conoscere, Tempo di azioni, and La Galassia
modern and forceful paintings and sculptures. mente). I learnt that she was a life-long close
Much later, I met her cousin Piera Levi- friend of Primo Levi, an author I admire and
Montalcini, and while visiting Rita in Rome, we from whom I have read and reread If This Is a
had the pleasure of meeting a more distant cousin Man and The Periodic Table. Rita was Primo
of hers, Dr. Sacerdote de Lustig, from Argentina. Levi’s confidant. In 1987, I received an unex-
Through many “at home” lunch and dinner pected call at home from Rita in tremendous
occasions, besides science, we discussed with anguish and she was nearly crying. This was to
Rita art, literature, history, and the state of the let me know that Primo Levi died by suicide. He
world. It was a well-known fact that the was one of her closest friends with whom she
Mussolini regime blunted her career in Torino; a shared transcendental moments while hiding
lesser-known irony was that the balcony of Rita’s from and resisting the fascistic Italian regime.
apartment in Rome overlooked the park, which She felt partially responsible for his death because
was part of Mussolini’s villa (see Fig. 9.1). she missed one of his calls asking for help. It was
The irony resides in the fact that Mussolini’s a long talk. I found myself in the estranged pre-
regime established the “Racial Laws” in Italy due dicament of comforting Rita; it was an intense
to which she was banned from Torino University emotional experience, which fortified our friend-
and it could have ended her career as well as her ship. Our encounters were always en famille with
life. This dramatic period of Rita’s life is well my wife Martha and while in Oxford also with my
reflected in her inspiring book In Praise of Imper- daughters Paula and Karina. We also often visited
fection (Levi-Montalcini 1988). our mutual labs (see Fig. 9.2).
Rita was a true “Renacentist” individual. Very Rita followed with interest my career evolu-
few were the subjects on which Rita did not have tion. She thought that I should not leave the
an opinion. She was a humanist and a defender of historically rich Oxford environment, my univer-
minority rights. I remember that over her desk sity position, and my fellowship with a Lincoln
there was a portrait of Dr. Martin Luther King. College, with late medieval traditions. Her initial
Besides science, during our encounters, we often resistance was converted in her unreserved
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 123

Fig. 9.2 I am visiting


Rita’s lab in Rome with the
slides for my talk at hand
and in the company of
Pietro Calissano, circa 1986

support when she understood about my enthusi- still regret that I missed this great occasion and
asm of taking a larger responsibility, across the most of all I regret not being able to share the
Atlantic, at McGill University. While at McGill, moment in which Rita received the highest inter-
she visited our lab and interacted for several days, national recognition for her scientific
in the most youthful fashion, with my new lab contributions.
trainees. Also, I had the satisfaction that my prop- Another memorable encounter with Rita was
osition of inviting Rita to give one of the first when I was invited by her to give one of the first
lectures for the Faculty of Medicine Series on lectures following the opening of the EBRI in
Foundations of Medical Sciences at McGill Uni- Rome and other lectures and interactions with
versity was enthusiastically accepted. This was an Rita and EBRI colleagues thereafter.
initiative organized by the very inspiring dean of With the passing of the years, I had the privi-
medicine, Richard Cruess, and a number of lege of participating in the symposium organized
McGill notables. These lectures included a good by Luigi Aloe and Italian colleagues in 2009, for
list of Nobel Laureates and other outstanding celebrating Rita’s hundredth anniversary and her
trailblazer scientists that were celebrated with a enormous legacy to science. I also attended the
formal black-tie dinner. . .a tradition that I helped more formal ceremony held in the Roman
to “transport” from Oxford to McGill. Palazzo Senatorial Capitol hill, where Rita gave
In a 1984 meeting in Italy, though I had no a magnificent speech next to the statue of Julius
idea she had already been nominated, I forecasted Cesar. Again, a most ironic distinction for a “non-
to Rita that she would win the Nobel Prize, to Arian” person once persecuted by the Race Laws
which she responded: “That will never happen, of fascistic Italy to give a speech next to the
Claudio, but if it happens, you will be the first to imposing statue of Julius Cesar.
be invited to the Nobel ceremony.” And so she While I was leading the McGill University
did. I received a phone call and a formal invita- “Brain@McGill” initiative, I proposed that the
tion. Unfortunately, the 1986 Nobel ceremony university recognize the exceptional
coincided with the celebration in Argentina of contributions to medicine and to society made
the 50th wedding anniversary of my parents. I by Rita by awarding her an honorary doctor of
124 A. C. Cuello

Fig. 9.3 Having the honor


of “hooding” Rita with the
gown representing the
McGill Science Doctorate,
with the participation of the
Rector of The Sapienza
University of Rome, on
February 23rd, 2011.
McGill Provost Anthony
Massi led the ceremony
with the endorsement of
McGill Principal Heather
Monroe-Bloom and with
the approval of the McGill
Senate

science degree. This was done with the collabora- a stroke-like type of cortical lesions (Sofroniew
tion of the Rector of the Sapienza Università di et al. 1983), a study led by Michael Sofroniew, by
Roma in 2011 and the McGill Provost Anthony then my Oxford graduate student. We
C. Masi. Rita was delighted with the distinction. I demonstrated that this retrograde atrophy of the
“hooded” Rita with the McGill robe upon com- n. basalis neurons was accompanied by a marked
pletion of the ceremony (see Fig. 9.3). This was a loss of its biochemical cholinergic phenotype,
very special and exceptional occasion, as it was i.e., loss of choline acetyltransferase activity
the first time in McGill’s 190-year history that the (Stephens et al. 1985). During our initial studies
University conferred an honorary doctorate on with that model, we demonstrated that the
foreign soil and the second time that such a shrunken n. basalis neurons could be salvaged
degree was awarded off campus. The first was in with the parenteral application of
1944, at the end of WWII, when Winston monogangliosides (Cuello et al. 1986; Sofroniew
Churchill and Franklin Delano Roosevelt were et al. 1986). The ganglioside GM1 was a com-
honored by McGill University in Quebec City. pound reported to have neuroprotective properties
That was my last encounter with Rita. and at the time applied clinically. Its
manufacturing process was proprietary of an Ital-
ian pharmaceutical company: Fidia. The funda-
mental biochemical aspects of ganglioside were
9.4 My First Interest in Neural
already revealed in the notable and extensive
Repair Leading to NGF
works of Tettamanti in Italy (Tettamanti 1988),
Research
Svennerholm in Sweden (Svennerholm 1980),
and Ledeen in the United States (Ledeen and Yu
My involvement in NGF research has the rooting
1982; Ledeen 1984).
in the development of an excellent model for the
My interest in the ganglioside GM1 was
study of the retrograde atrophy of cholinergic
aroused by a number of studies in the peripheral
neurons of the nucleus basalis in the rat following
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 125

nervous system, such as the demonstration of a whom I wrote more recently a testimonial honor-
GM1-induced improvement in the rate of recov- ing his long career in the field (Cuello 2012).
ery of the contractile responses of sympathetically Much of the interest in gangliosides research
denervated nictitating membranes (Ceccarelli plunged when the clinically applied GM1 was
et al. 1976), and the effects of brain gangliosides suspected of provoking Guillain-Barre syndrome.
on functional recovery in experimental regenera- One publication in 1994, based on a small patient
tion and re-innervation. The demonstration of the cohort, stated that the clinically applied GM1
neurorepair properties of GM1 on denervated could incite the Guillain-Barré syndrome (Landi
skeletal muscle was reported by Caccia and et al. 1994) and provoked much media coverage,
coworkers (Caccia et al. 1979) followed by com- which, regrettably, resulted in the suspension of
prehensive histological and functional studies. the GM1 therapy for peripheral neuropathies and
Gorio et al. (1980, 1983a, b) provided further in the suspension of the endorsement of ganglio-
support to ganglioside-induced axonal-synaptic side research by granting institutions. As such,
sprouting in models of compromised motor the fundamental neuropharmacology of
nerve innervation. gangliosides and their potential as reparative
Impressed by the above evidence, I had molecules, per se, or combined with trophic
discussions with colleagues at Fidia laboratory, factors were never fully developed. With the
and during my last years in Oxford and my initial passing of the years, it became quite clear that
years at McGill, we tested the hypothesis that there was no solid evidence of causality between
systemic administration of the GM1 ganglioside GM1 administration and the development of the
could have a reparative function in the CNS, Guillain-Barre syndrome, but rather there is solid
taking advantage of our model of retrograde atro- evidence to the contrary (Granieri et al. 1991;
phy of nucleus basalis neurons. To our amaze- Samson and Fiori 1994). The original paper of
ment, we found that the daily intraperitoneal Landi and coworkers was shown to be based on
application of 30 mg of GM1 for 30 days faulty epidemiological premises. However, the
provided a remarkable protection of the morphol- concern remained regarding the presence in the
ogy and neurochemistry of the forebrain n. basalis Guillain Barre syndrome of a diversity of anti-
cholinergic neurons (Cuello et al. 1986; ganglioside antibodies (including anti-GM1).
Sofroniew et al. 1986; Stephens et al. 1987). We Such concern has been currently nullified through
later observed that these neurotrophic-like effects numerous publications which demonstrated that
of GM1 gangliosides included improved in vivo antibodies against microbial glycolipids and
release of acetyl choline from the cerebral cortex glycoproteins present in the Guillain-Barre neu-
after extensive (Maysinger et al. 1988, 1990a, b), ropathy do cross-react with gangliosides. This
studies which were led by Dr. Maysinger with our aspect is nowadays interpreted as a cross-reaction
collaborators Herrera-Marschitz and Ungerstedt (“mimicry”) of likely causative anti-microbial
at the Karolinska Institute. These investigations antibodies detected in the syndrome
included the evidence that the monogangliosides (Wanleenuwat et al. 2020).
have reparative effects on GM1 in diminishing In the last period of our investigations of the
the ensuing deficits in memory-related tasks in neurotrophic-neuroreparative effects of GM1 in
animals bearing neocortical devascularizing experimental models of CNS cholinergic degen-
lesions (Elliott et al. 1989; Garofalo and Cuello eration and of the potential interactions with
1994). mature NGF, I was invited by Advances in Phar-
That period allowed me to have extensive macology to write a review on the neuropharma-
discussions with Rita about neural repair. It also cology of glycosphingolipids toward nerve
facilitated interactions with leading scientists in growth and repair (Cuello 1990) where I
ganglioside research and forming a long-lasting highlighted the state of the field and the possible
friendship with Richard Ledeen, in honor of application of GM1 alone and/or in combination
126 A. C. Cuello

with NGF. Ideas which were often discussed with higher CNS functions and in memory in particu-
Rita and which led to our lab entering fully lar (Drachman and Leavitt 1974; Bartus et al.
fledged NGF research. 1982).
Further to it, the histopathology of the nucleus
basalis with Nissl staining demonstrated the loss
9.5 NGF Functions in the Adult of “magnocellular” neurons, the cells responsible
Central Nervous System for the sole source of cholinergic innervation for
and Reparative Properties the human cerebral cortex (Whitehouse et al.
for Basal Forebrain Cholinergic 1982). This was a most influential scientific
Neurons report. However, the study overestimated the
loss of the nucleus basalis “magnocellular” cho-
The investigations on the reparative actions of linergic neurons; because the measurements were
gangliosides in halting the atrophy of basal fore- based on cell size, neuronal atrophy was
brain cholinergic neurons brought us to study the incorrectly counted as neuronal loss. The above
likely reparative effects of NGF in our lesion studies and a Science publication of Coyle and
model of retrograde degeneration of basal fore- collaborators (Coyle et al. 1983) promoted “the
brain cholinergic neurons (BFCN). At the time, it cholinergic hypothesis of AD,” with the implied
was already well established that NGF played a concept of a primary involvement of the basal
significant role in the survival and differentiation forebrain in the development of the pathology.
of primary sensory and autonomic neurons The resulting collective “cholinergic” studies
through the pioneering work or Rita. A similar had the merit of provoking the development of
NGF role on the maturation of the basal forebrain therapeutic anticholinesterases, with the objective
cholinergic neurons and their terminations in the of improving the brain’s “cholinergic tone” by
cerebral cortex and the hippocampus was blocking the extracellular degradation of acetyl
established (Mobley et al. 1986; Molnar et al. choline. This approach remains, even nowadays,
1998). the most successful symptomatic treatment in the
By then, it was assumed that BFCNs were AD pathology and its effects are most surprising,
completely dependent on NGF not only for their given the advanced brain pathology at the time of
developmental differentiation but as well for their clinical presentation. Since then, the field has
survival in the adult CNS. Such a concept was evolved considerably. In a review by the “Cho-
reinforced by the apparent death of the BFCN of linergic Working Group” (Hampel et al. 2018),
the medial septum, when deprived of “target- more contemporaneous ideas have been advanced
derived” NGF by the interruption of their connec- regarding the significance of BFCN in AAD.
tion with the hippocampus, by the axotomy of the In the early years of the above saga, with
fimbria-fornix projection, and their subsequent Michael Sofroniew, we proposed that the
rescue of such neuronal cholinergic set by the involvement of BFCN in the AD pathology was
timely application of exogenous mature NGF in a not a “primary” as initially thought but rather a
the ventricular space (Hefti 1986; Kromer 1987; “secondary” consequence to a primary cortical/
Williams et al. 1986; Gage et al. 1988). hippocampal pathology—a hypothesis which was
The above studies were significant in advanced in an invited TINS review (Cuello and
establishing the “proof of principle” of NGF Sofroniew 1984). Nowadays, it is largely
reparative properties in the CNS. They generated accepted that the presence of endogenous NGF
a great deal of anticipation in the field, since it had is not essential for BFCN survival, but rather for
already been established that this cholinergic sys- the maintenance of the cholinergic phenotype at
tem appeared most vulnerable in Alzheimer’s the cell body and axonal terminations. The first
disease (AD) (Bowen et al. 1976; Davies and and most unequivocal case was made by
Maloney 1976). This was of great interest as it Sofroniew and collaborators (Sofroniew et al.
was shown in humans and primates that the CNS 1990). In that report the case was made (quite
cholinergic system played a substantive role in convincingly) that, even after the excitotoxicity
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 127

elimination of most hippocampal neurons supply- et al. 1993). Such interpretation was later con-
ing “target-derived” NGF, the medial septum firmed experimentally by Rabin and Mochetti by
cholinergic neurons survive in a shrunken state. providing evidence of a direct GM1 activation of
This was in direct contradiction of the prevalent the TrkA receptors (Rabin and Mocchetti 1995),
concept of neuronal death following a drastic by Mutoh and collaborators showing that GM1
axotomy of the fimbria-fornix: an axotomy that tightly associates with the high-affinity tyrosine
was too close to the cell body of BFCN, thus kinase TrkA facilitating it (Mutoh et al. 1995),
leading to neuronal loss. In other words, the and by Farooqui and collaborators supporting a
reported cell death of the NGF-dependent cholin- GM1-induced dimerization of TrkA receptors
ergic neurons in the investigations applying the (Farooqui et al. 1997), both molecular alterations
interruption of the septal-hippocampal axonal signifying increased responses to NGF receptor
connection was not due to the deprivation of activation.
NGF supply but rather by the classical concept This line of work brought us to demonstrate
of neuronal cell death provoked by proximal that exogenous NGF alone and/or potentiated by
axotomy (Ramón y Cajal and May 1928). Subse- gangliosides, besides preventing the atrophy of
quently, we and others demonstrated that, in the nucleus basalis neurons NGF, had the capacity
human AD pathology, the predominant conse- of generating de novo cholinergic synaptogenesis
quence is the cellular atrophy, more than cell in the non-lesioned areas of the cerebral cortex of
death of BFCN of the nucleus basalis (Pearson adult rats, following stroke-like lesions (Garofalo
et al. 1983; Kordower et al. 1989). et al. 1992) (Fig. 9.4).
With the guiding hypothesis that cholinergic The ensuing comprehensive neurochemical
neurons of the nucleus basalis suffer a secondary, and pharmacological analysis of the NGF
retrograde, atrophy to a primary cortical lesion, neurotrophic effects in the CNS cholinergic sys-
we investigated the potential of exogenous NGF tem demonstrated correlative improvements in
for the recovery of degenerating NGF-dependent the choline uptake mechanisms and on the
cholinergic neurons of the nucleus basalis. Our in vivo microdialysis release of acetyl choline
initial studies on the retrograde atrophy of BFCN (Garofalo and Cuello 1995; Maysinger et al.
cell bodies and nerve terminals following cortical 1992), observations which are consistent with an
stroke-type lesions revealed the ganglioside NGF-induced generation of cortical cholinergic
potentiation of the NGF biochemical and struc- synapses in the adult and fully differentiated ner-
tural reparative effects (Cuello et al. 1989). With vous system. Such studies in our view fulfill the
Lorella Garofalo we did an exhaustive analysis predictions of Ramon y Cajal in that “...if experi-
comparing the therapeutic impact of NGF or the mental neurology is some day to supply artifi-
ganglioside GM1 alone or their therapeutic com- cially the deficiencies in question ... it must give
bination on cholinergic high-affinity choline to the sprouts (axonal sprouts), by means of ade-
uptake, receptor binding and choline acetyl trans- quate alimentation, a vigorous capacity for
ferase enzymatic activity in the cortical synaptic growth...” (Ramón y Cajal and May 1928).
area, and the micro-dissected n. basalis after A further demonstration of the fine-tuning
stroke-like cortical lesions on the retrograde cho- exerted by NGF on the maintenance of the den-
linergic atrophy model in the rat (Garofalo and sity of basal forebrain cholinergic synapses was
Cuello 1995). The study confirmed the coopera- provided by experiments in which the cortical
tive actions of NGF and GM1 in re-establishing endogenous NGF was mopped-out with the appli-
biochemical cholinergic markers after CNS cation of anti-NGF monoclonal antibodies or,
lesions and illustrated narrower “pseudotrophic” alternatively, with the injection of TrkA peptide
actions of GM1 (less potency and narrower thera- mimetic blockers. Thus, negating the NGF occur-
peutic window (Garofalo and Cuello 1995)). We rence or blocking its TrkA receptor activation was
further interpreted the GM1 potentiation of NGF sufficient to provoke the loss of pre-existing cor-
effects might be due as a facilitation of the NGF tical cholinergic synapses, in proportion to the
binding/activation of the TrkA receptor (Garofalo proximity of the site of injection (Debeir et al.
128

Fig. 9.4 Upper left-hand panel, the gray-shaded area represents the neocortical sites of such brain level. The aggrupation of cholinergic neurons in the inner portions of the
basal forebrain nerve terminals and site of the generation of NGF; the red area globus pallidus represents the nucleus basalis. On the right side the cholinergic (ChAT-
illustrates the unilateral experimental cortical stroke-like lesion. Upper right-hand immunoreactive) n. basalis neurons of control animals, middle micrograph
A. C. Cuello

panel on the left, the black dots represent the localization of cholinergic neurons at representing the retrograde atrophy of n. basalis cholinergic neurons following a
9

ä
Fig. 9.4 (continued) cortical lesion. The lower micrograph represents the recovery of presynaptic boutons in the NGF-treated cortically lesioned rats, indicative of
these n. basalis cholinergic neurons with the administration of exogenous NGF. Lower synaptogenesis. Lower right-hand panel illustrates the representative ultrastructural
left-hand panel illustrates micrographs and histograms of the density of the varicosities images of the shrinkage of cortical cholinergic presynaptic in the lesioned rats and
(presynaptic sites) of cholinergic terminals in the remaining, non-lesioned, cerebral the hypertrophy and large area of contact of cholinergic varicosities in the lesioned
cortex in control rats, control plus NGF, adjacent stroke-type lesions without NGF and NGF-treated rats. As per Garofalo et al., PNAS, 1992
after NGF treatment, respectively. Note the above average density of cholinergic
Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology
129
130 A. C. Cuello

Fig. 9.5 The upper


drawing represents
schematically the
experimental approach
utilized to investigate the
impact of the cerebral
cortex endogenous mature
NGF in the maintenance of
the NGF-dependent
cholinergic synapses
originated from nerve
terminals of the n. basalis of
the rat. The occurring
mature NGF was
immunoneutralized either
by the application of anti-
NGF monoclonal
antibodies or by blocking
their activation of the TrkA
receptor by applying a
mimetic peptide. As
represented in the
histogram at the bottom,
both procedures resulted in
the significant loss of the
density of pre-existing
cholinergic presynaptic
boutons (VAChT-IR:
Vesicular Acetyl Choline
Transporter-
Immunoreactive sites), as in
Debeir et al., PNAS, 1999

1999). This was a finding indicating that the “day- dependent manner. A failure in a release mecha-
to-day” density of cortical cholinergic neurons is nism could have provided an explanation for tro-
entirely dependent on the continued supply of phic disconnection of basal forebrain cholinergic
minute amounts of endogenous NGF. This con- neurons in aging or neurodegenerative
cept is well in line with the classical Hebbian conditions. The situation of the NGF sustenance
principle “of synaptic growth following neuronal of NGF-dependent neurons in the case of AD was
activity” (Hebb 1949) (Fig. 9.5). particularly challenging. In early years, authorita-
tive publications suggested a causative role of
NGF failure for the retrograde atrophy death of
9.6 A Novel NGF Metabolic FBCN (Stewart and Appel 1988; Hefti and
Pathway Weiner 1986). However, such assumptions of an
NGF trophic disconnect explaining the atrophy of
9.6.1 The Background basal cholinergic neurons and the loss of cortical
and hippocampal synapses in AD was not
We came across the evidence that proNGF and supported by the neurochemical evidence. It was
not mature NGF (mNGF) was the molecular form shown that in AD brains the NGF synthesis
of this neurotrophin being released in an activity- remained unchanged (Goedert et al. 1986; Jette
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 131

et al. 1994; Fahnestock et al. 1996) and that the with Martin Bruno, by then my graduate student,
offer of the NGF precursor molecule, proNGF, in deciphering how NGF was actually released in
contrary to expectations, was not diminished but an activity-dependent fashion in the fully matured
supranormally elevated (Fahnestock et al. 2001, central nervous system. The preceding work, as
2004; Peng et al. 2005; Pedraza et al. 2005). stimulating it was at the time, had some signifi-
Such a situation created an AD NGF paradox cant drawback.
regarding the apparent healthy status of the tro- The preceding studies, in fact, supported the
phic support in the occurrence of a progressive decades-long prevalent idea that the molecule
atrophy of BFCN. The first explanation has been released in an activity-dependent manner was
the diminished TrkA-immunoreactive sites in AD mature NGF. This assumption was based on
brains (Mufson et al. 1997; Hock et al. 1998). in vitro studies on dissociated hippocampal cells
Another explanation has been that in the AD or tissue slices transfected with either proNGF or
pathology there is an Aβ amyloid-induced mature NGF cDNA constructs and analyzing the
impairment in the axonal retrograde transport supernatants resulting from the application of
responsible for NGF mobilization from nerve high molarity of KCl (Blochl and Thoenen
terminals to neuronal soma to affect the trophic 1996; Hoener 2000; Mowla et al. 1999). In
functions (Salehi et al. 2006). Both the these historical studies, the resulting supernatants
impairment of axonal transport and the losses of were assayed with the available anti-NGF
the cognate high-affinity receptor could be plau- antibodies in an ELISA format in the assumption
sible scenarios. On the other hand, the presence of that the signals thus obtained represented the
mature NGF is fundamentally responsible for actual mature NGF. However, it is nowadays
regulating the expression of all housekeeping clear that these antibodies, and current ones at
genes, including those related to axonal transport, large, do not differentiate mature NGF from its
transmitter-specific proteins, i.e., NGF-dependent precursor, the proNGF molecule.
neurons. This is also the case for the expression of To avoid the above experimental drawbacks,
TrkA protein, the high-affinity NGF receptor, we decided to go back to “first principles.” We
since NGF has been shown in in vivo conditions resorted to use ex vivo rat cerebral cortex tissue
where the TrkA receptor expression is controlled blocks, i.e., neurons preserving their contacts to a
by its own ligand: NGF (Figueiredo et al. 1995; great extent and in a conventional milieu, as
Venero et al. 1994). opposed to dissociated neurons or tissue slices,
The more challenging aspect is to resolve genetically manipulated to express mature NGF
whether the reported tau and Aβ intraneuronal nor proNGF. The cerebral cortex blocks were in
pathologies reported in the AD pathology are contrast taken from intact tissue. In consequence,
the cause of the atrophy of the BFCN or if it is a the experiments reflected the activity-dependent
pre-existing NGF trophic disconnect which release of endogenous neurotrophins in physio-
makes these neurons vulnerable to the propaga- logical conditions. These cortical tissue blocks
tion of toxic tau and Aβ amyloid proteins. We were continuously superfused with a balanced
propose that in AD there is progressive loss of and oxygenated buffer, obtaining time-dependent
mature NGF, which is best explained by the exis- fractions of superfusates after neuronal stimula-
tence of a NGF metabolic pathway, as described tion with high KCl, carbachol, or glutamate. An
below. approach not very far from the Otto Loewi’s
classical “vagusstoff” experiments leading to the
discovery of chemical nerve transmission and
9.6.2 The Discovery of the NGF eventually the identification of acetyl choline
Metabolic Pathway (Loewi 1921). For that, Martin Bruno was able
to reproduce the tissue superfusion system which
Keeping in mind the above background regarding I learnt at the MRC Cambridge Iversen’s Unit in
the pathophysiology of NGF, we were interested Neurochemical Pharmacology. In the knowledge
132 A. C. Cuello

Fig. 9.6 On top, a schematic representation of ex vivo indicating that only proNGF was readily found in fractions
system utilized to investigate the nature of endogenous collected immediately after any type of tissue-stimulation.
neurotrophins released by neurotransmission or high NGF indicates a control band from loading with 5 ng of
molarity K+ stimulation. Small rat cortical tissue blocks mNGF. (b) and (c) illustrate the superfusate fractions
were placed into a chamber and constantly superfused with collected at 5-min intervals. In (b), the peak in the graph
a modified Hanks’ buffer equilibrated with 95% O2 and represents the increase in proNGF released after carbachol.
5% CO2 with a slow flow rate. After 30 min of equilibra- A second carbachol stimulation was blocked by the Ca++
tion of 0.25 ml/min at 37  C, the tissue was subjected to intracellular chelator AM BAPTA, while in (c) the block-
stimulations with carbachol (100 nM), glutamate (60 μM), ade of extracellular Ca++ with BAPTA failed to prevent
or KCl (50 mM) applied over a 5-min period. (a) A typical the transmitter-stimulated release
Western blot analysis of the detected neurotrophins,

that ELISA methods do not properly distinguish The finding of a stimulus-dependent proNGF
mature NGF from ProNGF, to investigate the release created a problem. How was the mature
superfusate fractions following tissue stimulation, NGF generated and how was it going to reach its
we resorted to apply a Western blot approach. To cognate receptors to exert its neurotrophic
our great surprise, the superfusate fractions actions? It took us about 2 years to discover the
emerging after stimulation with either KCl, car- occurrence of a complex metabolic cascade
bachol, or glutamate inexorably revealed that the responsible for the conversion of proNGF to
activity-dependent released neurotrophin in phys- mNGF and of the degradation in the extracellular
iological conditions was not mNGF, as the dogma space of the remnants of mNGF which were not
had established, but rather its precursor, proNGF internalized and transported retrogradely, i.e., fol-
(Fig. 9.6). lowing their binding TrkA/p75 receptor
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 133

Fig. 9.7 The top panels demonstrate the immunoreactive simultaneously in the extracellular space upon transmitter
sites within large pyramidal neurons of layer V of the or membrane-depolarizing KCl stimuli (image from video
cerebral cortex of the rat. Scale bar-xxx (from Bruno and animation of the NGF metabolic pathway: https://
Cuello, PNAS, 2006). The other panel illustrates the meetings.ami.org/2018/project/ngf-metabolism-in-
cohort of activity-dependent molecules released alzheimers-disease/)

complexes. These proteins responsible for the and biochemical phenotype: a process well
NGF metabolism were plasminogen, tPA (tissue illustrated by Mobley, Schwab, and others (Seiler
plasminogen activator), neuroserpin, and matrix and Schwab 1984; Grimes et al. 1996). The
metalloproteinases. We found this mix of zymo- remaining mNGF in the extracellular space is
gen, convertases, and endogenous inhibitors to ultimately degraded by activated Matrix Mettallo-
co-release along with proNGF. We found that proteases (MMP9) and also, more recently
the conversion of proNGF to mNGF is made by identified also by the actions of MMP3 (Pentz
the plasmin, which is generated by the action of et al. 2020). The activity of the MMPs is also
(tPA over the zymogen plasminogen. The actions regulated by the endogenous inhibitor Timp-1.
of tPA are, in turn, regulated by neuroserpin The key proteins encompassing the NGF meta-
inhibiting the extent of tPA activity (Bruno and bolic pathway have been co-localized along
Cuello 2006). The newly formed mNGF is there- proNGF in cortical pyramidal neurons, more
fore readily available to bound the cognate recep- prominently in lamina V (Bruno and Cuello
tor, internalized and retrogradely transported 2006), as illustrated in Fig. 9.7.
axonally, to the cell soma of the NGF-dependent The description of this metabolic pathway was
neurons to activate the generation of all proteins published in PNAS in 2006 (Bruno and Cuello
required for the maintenance of the anatomical 2006) with the approval of the elected Editor,
134 A. C. Cuello

Fig. 9.8 Schematic


representations of events
leading to proNGF
conversion into mNGF and
its degradation. Neuronally
stored proNGF,
plasminogen, tPA,
neuroserpin, proMMP-9,
andTIMP-1would be
released into the
extracellular space upon
neuronal stimulation.
Released tPA would induce
the conversion of
plasminogen into plasmin.
tPA activity will be tightly
regulated by the released
neuroserpin. The generated
plasmin would convert
proNGF into mNGF.
mNGF would interact with
its cognate receptors (TrkA
and p75 neurotrophin
receptor) and internalized
by synapses of
NGF-dependent neurons.
The remaining mNGF will
be ultimately degraded by
active MMP-9. From Bruno
and Cuello, PNAS, 2006

Hans Thoenen, who made towering contributions a spectacular buildup of proNGF. Contrary to it,
to our understanding of fundamental aspects of the application of an MMP inhibitor protects
the biochemistry and physiology of mNGF from its proteolytic degradation by
neurotrophins. Sending the submission for review MM9, with the consequential elevation of brain
and accepting its publication was an exemplary levels of mNGF, which otherwise are close to
gesture of scientific gentlemanship as the path- undetectable at physiological conditions
way contradicted his original ideas of intracellular (Fahnestock et al. 2001) (Fig. 9.9).
conversion of proNGF to mNGF and its release as Further to the pharmacological validation of
the mature neurotrophin. A schematic summary the NGF pathway, with Simon Allard further we
of the pathway is represented in Fig. 9.8. demonstrated that this pathway is downstream
This pathway was validated pharmaco- responsible for the maintenance of the cholinergic
logically in the rat brain initially by the applica- synaptic phenotype at the cerebral cortex level.
tion of exogenous neuroserpin blocking tPA By blocking the proNGF conversion to mNGF
activity and, therefore, inhibiting the plasmin for- with local administration of α2-antiplasmin in the
mation and leading to splicing the proNGF, thus rat prefrontal cortex, we observed, as expected, a
preventing the formation of mNGF. The result is diminution of the local cortical levels of
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 135

Fig. 9.9 Histogragrams of pharmacological interventions mNGF as compared with the control levels from cortical
on the NGF metabolic pathway. On the left, note the tissues receiving the inactive isomer GM(-) and thus dra-
remarkable increase in the cortical levels of proNGF in matically inverted the proNGF to mNGF physiological
neuroserpin-treated animals after 72 h post-injection ratio. The mNGF cortical levels were raised highly signif-
(mean +/– SEM; P  0.001; t test). The right-hand histo- icantly (P  0.001) having a consequential decrease of
gram illustrates that the cortical application of the MMP proNGF (P  0.01) as compared to basal levels
inhibitor GM6001 results in an over tenfold increment of

endogenous NGF and elevation of proNGF. Such bodies of n. basalis BFCN. This was
a scenario resulted in the significant loss or accompanied by a marked downregulation of the
pre-existing NGF-dependent cholinergic NGF p75 neurotrophin receptor and of TrkA
pre-synaptic boutons, without affecting the den- receptor as well as a reduction in choline
sity of glutamatergic, GABAergic, or catechol- acetyltransferase protein expression. These
aminergic synapses (Allard et al. 2012). In changes were restricted to the cholinergic
opposition, applying in the prefrontal cortex an n. basalis which were marked with the retro-
MMP inhibitor with the same experimental gradely transported FluoroGold (Allard et al.
approach resulted in an increment in the density 2018)—see Fig. 9.10.
of cholinergic synaptic boutons. This study provided strong evidence
At a later stage, we investigated with Simon demonstrating the cortical compromise of the
whether the phenotypic regulatory influence of extracellular maturation of proNGF sufficient to
the NGF metabolic pathway was extensive to cause a somatodendritic retrograde degeneration
the status of the cell bodies of the n. basalis of the cell bodies of the NGF-dependent BFCN of
BFCN. For those studies we used a combination the n. basalis. Such experimental evidence would
of marking the n. basalis neurons receiving the also give credence to the postulated idea that the
plasmin inhibitor at cortical synaptic level and cholinergic basal forebrain atrophy observed in
investigating the pharmacological effects on neu- AD and in DS is due to a deregulation of the NGF
ronal size and classical transmitter and receptor metabolic pathway, as discussed below.
markers.
To achieve that, we applied α2-antiplasmin in
a restricted area of the neocortex injecting simul-
9.7 The Deregulation of the NGF
taneously FluoroGold, a classical axonal retro-
Metabolic Pathway
grade transport marker. In that manner we could
in Alzheimer’s Disease
identify in the n. basalis which choline acetylase
and in Down Syndrome
(ChAT)-immunoreactive neurons were, or were
not, exposed to α2-antiplasmin. As such we found
The discovery of the NGF metabolic pathway
that the cortical blockade of the proNGF matura-
allowed us to test the hypothesis of its possible
tion into mNGF resulted in an atrophy of the cell
deregulation in the AD pathology as a plausible
136 A. C. Cuello

Fig. 9.10 Schematic


representation illustrating
the experimental models
and indicating that only the
n. basalis neurons exposed
to α2-antiplasmin (white-
brown ovals) at their
cerebral cortex suffered
retrograde degeneration
(cell body atrophy)
resulting from the inhibition
of plasmin activity
converting proNGF into
mNGF, while neurons
retrogradely transported
fluorogold particles (
yellow spots) were not
affected. The histograms
illustrate the degree of
neuronal cell shrinkage of
NGF-dependent cholinergic
BCFN after 2 or 4 weeks of
cortical application of α2-
antiplasmin and the rapid
loss of the expression of the
TrkA receptors in those
cells at the same time
intervals. This is an
observation reinforcing the
concept that in the
cholinergic neurons the
expression of TrkA receptor
proteins, as well as that of
all the neuronal
housekeeping proteins, is
dependent on the
continuous supply of
endogenous NGF. From
Allard et al, Neurobiology
of Aging, 2018, with the
publisher’s permission

outcome. That hypothesis, if proven, could (previously unexplained) elevation of proNGF


resolve the prevalent “NGF-cholinergic paradox” content. This compromise in the proNGF conver-
of cholinergic atrophy in the apparent trophic sion was accompanied by increased levels of the
factor supply. This we did initially with Martin mNGF-degrading MMP9 protein (Bruno et al.
Bruno. We applied the biochemical paradigm of 2009b). In other words, we found in AD brains
the NGF metabolic pathway by the investigation a scenario of “double jeopardy,” i.e., diminished
of the status of the pathway in control and AD mNGF production and augmented mNGF prote-
postmortem brain samples from the Netherlands olysis. Further to it, in collaboration with Elliott
brain bank. This revealed the existence of a com- Mufson, in cortical postmortem samples from the
promise in the conversion of proNGF to the bio- brain bank of the Religious Order Study, we
logically active mNGF, with the consequential found a notable accumulation of both the protein
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 137

and the zymogenic activity of MMP9 in samples of APP and Aβ proteins in cerebral cortex post-
of individual identified as MCI (mild cognitive mortem samples of DS individuals. More signifi-
impaired) (Bruno et al. 2009a). More importantly cantly, these studies eloquently illustrated that
that study demonstrated a correlation between the there was indeed a compromise of the NGF met-
degree of MMP9 content and the degree of cog- abolic pathway responsible for the conversion of
nitive losses (Bruno et al. 2009b). These findings proNGF to mNGF in DS brains bearing AD
were highly suggestive that the NGF deregulation pathology. This was shown by lower levels of
could start at preclinical levels of the AD plasminogen and a reduced tPA mRNA as well
pathology. as an increment in neuroserpin expression and
With the arrival of Florencia Iulita to our lab, also an increase in brain proNGF levels (Iulita
we embarked on a study of the status of the NGF et al. 2014). These conditions eloquently signaled
metabolic pathway in Down syndrome (DS). We a marked impairment in the maturation of
hypothesized that DS individuals should have an proNGF to mNGF, providing a rationale for the
equally deregulated NGF metabolic pathway as known DS cholinergic atrophy (Yates et al.
they develop inexorably an AD pathology, given 1980). As is in the AD brains, we observed in
that the trisomy of the chromosome 21 results in DS brains elevated zymogenic activity of MMP9,
triplication of the gene expressing APP (amyloid which would exacerbate the degradation of the
precursor protein), which is provoking the pathol- already diminished levels of the biologically
ogy (Lott 1982; Wisniewski and Wrzolek 1988; active mNGF. Interestingly the genetic model of
Hardy et al. 1989; Head et al. 2016; Lott and DS and the cultures of human fetal cortical cells
Head 2019). As such, DS individuals should also demonstrated an age-related increment of
also display a deregulated NGF brain metabolism, proNGF. Importantly, the levels of proNGF in
an aspect which would be consistent with the DS brains correlated with the Aβ42 amyloid con-
known atrophy of the basal forebrain cholinergic tent; see Fig. 9.11 (Iulita et al. 2014).
system. As the AD in DS individuals develops silently
The investigation on the status of the NGF over decades, we thought that the analysis of body
metabolic pathway in DS was led by Florencia; fluid samples in adults at stages prior to the presen-
and it was facilitated through an excellent collab- tation of clinical AD could provide clues or poten-
oration with Sonia Do Carmo, Lotta Granholm, tial biomarkers signaling the evolving, silent, AD
Jorge Busciglio, and Thomas Wisniewski. As brain pathology in DS. For our first study we had
such we confirmed the expected high expression the good fortune of interacting with Filippo Caraci

Fig. 9.11 Scattergrams illustrating a positive correlation P ¼ 0.001) cortices of post-mortem brain samples of DS
between proNGF and amyloid-β42 in (a) the temporal individuals. From Iulita et al, Brain 2014 with the
(r ¼ 0.626, P ¼ 0.017) and in (b) the frontal (r ¼ 0.629, Publisher’s permission
138 A. C. Cuello

who, with his collaborators, was following longitu- 9.8 The NGF Metabolic Pathway
dinally a mature DS cohort performing cognitive in Health and Disease
assessments and extracting blood samples. and Future Challenges
Through a collaborative work led by Florencia
Iulita, we investigated the plasma levels of Aß It is of interest that the Alzheimer’s-like amyloid
amyloid peptide, proNGF, tPA, neuroserpin, pathology in a rat transgenic model
metalloproteases in 31 individuals with DS (with overexpressing the human app gene bearing AD
and without dementia) and in 31 healthy controls. mutations is sufficient to deregulate the brain
The examination of associations between NGF metabolic pathway. The obvious conclusion
biomarkers and cognitive decline demonstrated is that the sole occurrence of just one of hallmarks
foremost that declining plasma Aß peptides and of the AD pathology like the abnormal brain
increasing proNGF levels correlated with cognitive accumulation of Aβ amyloid peptides can elicit
decline (Iulita et al. 2016b). These findings support alterations of this pathway similar to those
the notion that incremental levels of proNGF could observed in human brains’ postmortem samples
be of value in identifying individuals who are at from individuals with sporadic AD. In the
preclinical AD stages. This is being an unmet need McGill-APP transgenic rat the brain neurochemi-
in the field. Figure 9.12 illustrates the changes in cal analysis revealed, as in AD, a differential
proNGF levels combined with “self-to-self” longi- dysregulation of NGF and BDNF transcripts and
tudinal analysis that are strategies of great promise protein expression as the amyloid pathology
(Fig. 9.13).

Fig. 9.12 Illustration of time-dependent relationship squares represent individuals whose plasma proNGF
between changes in blood proNGF with cognitive decline levels did not change significantly. Regression lines are
in DS individuals. Graphical representation of cognitive modeling those individuals whose proNGF levels
score drop in the Test of Severe Impairment (TSI) (left increased over 12 months. From Iulita et al. 2016
graph) and in the Modified Mini Mental (3-MS) (right reproduced with the Publisher’s permission. The deregu-
hand graph) scores associated with longitudinal change lation of the NGF metabolic pathway has been discussed
(d) in proNGF as analyzed by linear regression analysis. in extent in a number of reviews (Iulita and Cuello 2014,
The upward triangles represent non-demented DS 2016; Iulita et al. 2016a; Cuello et al. 2019) and the overall
individuals whose plasma levels of proNGF increased ideas discussed above are schematically represented in
over 12 months and downward triangles depict DS Fig. 9.13
individuals whose levels of proNGF decreased. Black
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 139

Fig. 9.13 Left, schematic representation of the biochem- production will diminish the generation of all proteins in
ical alterations of the NGF metabolic pathway in AD and NGF-dependent BFCN, including the expression of TrkA
DS with AD pathology. Deficits in tPA diminish plasmin- receptors. That will result in the progressive atrophy of
ogen conversion into plasmin, therefore leading to these neurons and therefore in the expression of the ace-
impaired proNGF conversion with consequent proNGF tylcholine (Ach) synthesizing enzyme (choline acetyl
accumulation. The low amounts of mNGF are further transferase: (ChAT) and the reduced release of its neuro-
compromised by higher MMP-9 activity. transmitter Ach. That will diminish the so-called “cholin-
Pro-inflammatory mediators as well as other metallo- ergic tone” at their cortical presynaptic sites. A diminished
proteases like MMP-3 would increase MMP-9 activation cholinergic tone would cease to facilitate the activation of
in the AD pathology. From Iulita and Cuello (2016). the “non-amyloidogenic” APP pathway (Nitsch et al.
Right, scheme indicating that the abnormal brain accumu- 1992, 1993) in the cerebral cortex, i.e., facilitating instead
lation of toxic Aβ peptides per se or conjointly with the the APP “amyloidogenic” pathway generating more Aβ
early AD inflammation will provoke the impaired NGF amyloid peptides and, consequently, creating a pathologi-
metabolism as represented on the left. The reduced mNGF cal vicious cycle

progresses. Briefly, BDNF mRNA levels start to proNGF molecules accompanied at late stages
diminish significantly at very early stages of the with an increased expression of MMP9, the
amyloid pathology, even before the occurrence of NGF-degrading protease. Again, this experimen-
the amyloid plaques (Iulita et al. 2017). Loss of tal evidence would favor a direct or indirect caus-
BDNF expression is characteristic of AD pathol- ative role of Aβ amyloid on the AD pathology
ogy (Holsinger et al. 2000). Contrary to that, no NGF metabolic deregulation. Whether amyloid
changes were observed in NGF mRNA expres- per se or indirectly remains to be established;
sion at any stage of the amyloid pathology in the however, an amyloid-induced CNS inflammation
transgenic rats, while the levels of proNGF were could contribute to the NGF metabolic deregula-
increasingly elevated paralleling the progress of tion. This has been insinuated by the
the amyloid pathology (Iulita et al. 2017) simi- demonstrating that blocking in vivo with
larly to the case in AD pathology, as discussed in minocycline the inflammatory effects elicited Aβ
the preceding sections. As in the human condi- amyloid oligomers also blunted the Aβ peptide
tion, the transgenic rat exhibiting progressive oligomers rise of proNGF levels (Bruno et al.
amyloid pathology displayed the same AD-like 2009b)
compromise of the conversion of proNGF into While we have strong experimental evidence
mature NGF resulting in a brain build-up of regarding an Aβ amyloid causality for the NGF
140 A. C. Cuello

metabolic deregulation of the NGF metabolic individuals already show AD-like alterations in
pathway in AD, the likely added participation of the tissue levels of key molecules participants on
tau pathology to this metabolic deregulation has the NGF pathway. In consequence, it would be
not yet been explored. reasonable to assume that body fluids, in particu-
One of the questions often made to us is lar CSF, should reflect some of these brain molec-
“whether the pharmacological manipulation of ular changes.
this pathway could reestablish the NGF ‘trophic
tone’ in AD and therefore reestablish the dimin- Acknowledgments The work narrated above was made
ished ‘cholinergic tone,’” which has a definitive historically possible by several UK, Canada, and US
research-granting institutions, notably the UK MRC and
role in cognitive functions. The assumption being
Wellcome Trust, from Canada the MRC, CIHR, and the
that a timely and effective therapeutic strategy Alzheimer Society of Canada, and from the United States
reestablishing the normality of the endogenous the Alzheimer Association and the NIH. Most importantly,
NGF supply, at its physiological sites, would be the research narrated here would not have been possible
without the creativity and dedication of my outstanding lab
of undeniable value. This would be a great prom-
members or without the contributions from my outstand-
ise as even in AD brains, as Eriksdotter and ing external collaborators.
collaborators have shown, the application of
exogenous NGF elicits a reparative improvement Dedication I would like to dedicate these reflections
in the brain’s glucose metabolism (Ferreira et al. about trophic responses not only to the memory of Rita
2015) along with a beneficial effect on CSF cho- Levi Montalcini and her stellar career, but also as a tribute
to Mario Bunge’s legacy. He was one of the world’s
linergic markers (Karami et al. 2015).
greatest philosophers of his generation. A philosopher
The pharmacological reestablishment of a with formal and deep knowledge of the physical and
“healthy” NGF metabolism is a desirable thera- biological sciences. A philosopher who understood synap-
peutic objective. It is also a most challenging tic plasticity and who strongly made the case that mind,
consciousness, and all psychological responses were
endeavor, given that the molecular members of
strictly governed by neuronal connections and biochemi-
the protease-convertase cascade are also involved cal brain events. I had the privilege of sharing with Mario
in other fundamental aspects of the CNS bio- many interminable scientific discussions and with him and
chemistry and physiology. Notwithstanding such his family many memorable events. I will sorely miss his
loss as a dear friend and as an inspiring McGill colleague.
obstacles, our lab is investigating a potential,
demanding, experimental avenue.
A most immediately promising route would be References
to further explore the likelihood that the
pathology-induced deregulation of the NGF met- Allard S, Leon WC, Pakavathkumar P, Bruno MA,
Ribeiro-Da-Silva A, Cuello AC (2012) Impact of the
abolic pathway could render predictable NGF maturation and degradation pathway on the cor-
biomarkers of the preclinical AD pathology. tical cholinergic system phenotype. J Neurosci
This is a very likely scenario. As discussed 32:2002–2012
above, progressive raise of blood proNGF levels Allard S, Jacobs ML, Do Carmo S, Cuello AC (2018)
Compromise of cortical proNGF maturation causes
are observed in DS in their transition to DS plus selective retrograde atrophy in cholinergic nucleus
clinical AD. Preliminary work of our lab with the basalis neurons. Neurobiol Aging 67:10–20
lab of Juan Fortea (Sant Pau Memory Unit, Aloe L (2004) Rita Levi-Montalcini: the discovery of
Barcelona would support that an even better nerve growth factor and modern neurobiology. Trends
Cell Biol 14:395–399
reflection of the NGF deregulation in DS is Bartus RT, Dean RL 3rd, Beer B, Lippa AS (1982) The
found in CSF material (Pentz et al. 2019). cholinergic hypothesis of geriatric memory dysfunc-
Another encouraging information is emerging tion. Science 217:408–414
from an ongoing study in our lab, led by Rowan Blochl A, Thoenen H (1996) Localization of cellular stor-
age compartments and sites of constitutive and
Pentz in collaboration with David Bennett. In activity-dependent release of nerve growth factor
these advanced studies there is a clear indication (NGF) in primary cultures of hippocampal neurons.
that in postmortem brain samples of NCI Mol Cell Neurosci 7:173–190
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 141

Bowen DM, Smith CB, White P, Davison AN (1976) Elliott PJ, Garofalo L, Cuello AC (1989) Limited neocor-
Neurotransmitter-related enzymes and indices of hyp- tical devascularizing lesions causing deficits in mem-
oxia in senile dementia and other abiotrophies. Brain ory retention and choline acetyltransferase activity—
99:459–496 effects of the monosialoganglioside GM1. Neurosci-
Bruno MA, Cuello AC (2006) Activity-dependent release ence 31:63–76
of precursor nerve growth factor, conversion to mature Fahnestock M, Scott SA, Jette N, Weingartner JA,
nerve growth factor, and its degradation by a protease Crutcher KA (1996) Nerve growth factor mRNA and
cascade. Proc Natl Acad Sci U S A 103:6735–6740 protein levels measured in the same tissue from normal
Bruno MA, Leon WC, Fragoso G, Mushynski WE, and Alzheimer’s disease parietal cortex. Brain Res Mol
Almazan G, Cuello AC (2009a) Amyloid beta-induced Brain Res 42:175–178
nerve growth factor dysmetabolism in Alzheimer dis- Fahnestock M, Michalski B, Xu B, Coughlin MD (2001)
ease. J Neuropathol Exp Neurol 68:857–869 The precursor pro-nerve growth factor is the predomi-
Bruno MA, Mufson EJ, Wuu J, Cuello AC (2009b) nant form of nerve growth factor in brain and is
Increased matrix metalloproteinase 9 activity in mild increased in Alzheimer’s disease. Mol Cell Neurosci
cognitive impairment. J Neuropathol Exp Neurol 18:210–220
68:1309–1318 Fahnestock M, Yu G, Michalski B, Mathew S,
Caccia M, Meola G, Cerri C, Frattola L, Scarlato G, Aporti Colquhoun A, Ross GM, Coughlin MD (2004) The
FJM (1979) Treatment of denervated muscle by nerve growth factor precursor proNGF exhibits
gangliosides. Muscle Nerve 2:382–389 neurotrophic activity but is less active than mature
Ceccarelli B, Aporti F, Finesso M (1976) Effects of brain nerve growth factor. J Neurochem 89:581–592
gangliosides on functional recovery in experimental Farooqui T, Franklin T, Pearl DK, Yates AJ (1997) Gan-
regeneration and reinnervation. Adv Exp Med Biol glioside GM1 enhances induction by nerve growth
71:275–293 factor of a putative dimer of TrkA. J Neurochem
Coyle JT, Price DL, Delong MR (1983) Alzheimer’s dis- 68:2348–2355
ease: a disorder of cortical cholinergic innervation. Ferreira D, Westman E, Eyjolfsdottir H, Almqvist P,
Science 219:1184–1190 Lind G, Linderoth B, Seiger A, Blennow K,
Cuello AC (1990) Glycosphingolipids that can regulate Karami A, Darreh-Shori T, Wiberg M, Simmons A,
nerve growth and repair. Adv Pharmacol 21:1–50 Wahlund LO, Wahlberg L, Eriksdotter M (2015) Brain
Cuello AC (2012) Gangliosides, NGF, brain aging and changes in Alzheimer’s disease patients with
disease: a mini-review with personal reflections. implanted encapsulated cells releasing nerve growth
Neurochem Res 37:1256–1260 factor. J Alzheimers Dis 43:1059–1072
Cuello AC, Sofroniew MV (1984) The anatomy of the Figueiredo BC, Skup M, Bedard AM, Tetzlaff W, Cuello
CNS cholinergic neurons. Trends Neurosci 7:74–78 AC (1995) Differential expression of p140trk,
Cuello AC, Stephens PH, Tagari PC, Sofroniew MV, p75NGFR and growth-associated phosphoprotein-43
Pearson RC (1986) Retrograde changes in the nucleus genes in nucleus basalis magnocellularis, thalamus
basalis of the rat, caused by cortical damage, are and adjacent cortex following neocortical infarction
prevented by exogenous ganglioside GM1. Brain Res and nerve growth factor treatment. Neuroscience
376:373–377 68:29–45
Cuello AC, Garofalo L, Kenigsberg RL, Maysinger D Gage FH, Armstrong DM, Williams LR, Varon S (1988)
(1989) Gangliosides potentiate in vivo and in vitro Morphological response of axotomized septal neurons
effects of nerve growth factor on central cholinergic to nerve growth factor. J Comp Neurol 269:147–155
neurons. Proc Natl Acad Sci U S A 86:2056–2060 Garofalo L, Cuello AC (1994) Nerve growth factor and the
Cuello AC, Pentz R, Hall H (2019) The brain NGF meta- monosialoganglioside GM1: analogous and different
bolic pathway in health and in Alzheimer’s pathology. in vivo effects on biochemical, morphological, and
Front Neurosci 13:62 behavioral parameters of adult cortically lesioned rats.
Davies P, Maloney AJ (1976) Selective loss of central Exp Neurol 125:195–217
cholinergic neurons in Alzheimer’s disease. Lancet Garofalo L, Cuello AC (1995) Pharmacological character-
2:1403 ization of nerve growth factor and/or monosialogan-
Debeir T, Saragovi HU, Cuello AC (1999) A nerve growth glioside GM1 effects on cholinergic markers in the
factor mimetic TrkA antagonist causes withdrawal of adult lesioned brain. J Pharmacol Exp Ther
cortical cholinergic boutons in the adult rat. Proc Natl 272:527–545
Acad Sci U S A 96:4067–4072 Garofalo L, Ribeiro-Da-Silva A, Cuello AC (1992) Nerve
De Robertis E, De Lores Arnaiz GR, De Iraldi AP (1962) growth factor-induced synaptogenesis and hypertrophy
Isolation of synaptic vesicles from nerve endings of the of cortical cholinergic terminals. Proc Natl Acad Sci U
rat brain. Nature 194:794–795 S A 89:2639–2643
Drachman DA, Leavitt J (1974) Human memory and the Garofalo L, Ribeiro-Da-Silva A, Cuello AC (1993) Poten-
cholinergic system. A relationship to aging? Arch tiation of nerve growth factor-induced alterations in
Neurol 30:113–121 cholinergic fibre length and presynaptic terminal size
142 A. C. Cuello

in cortex of lesioned rats by the monosialoganglioside receptor expression in the parietal cortex of patients
GM1. Neuroscience 57:21–40 with Alzheimer’s disease. Neurosci Lett 241(2):151–
Goedert M, Fine A, Hunt SP, Ullrich A (1986) Nerve 154. https://doi.org/10.1016/S0304-3940(98)00019-6
growth factor mRNA in peripheral and central rat Hoener MC (2000) Role played by sodium in activity-
tissues and in the human central nervous system: lesion dependent secretion of neurotrophins—revisited. Eur
effects in the rat brain and levels in Alzheimer’s dis- J Neurosci 12:3096–3106
ease. Brain Res 387:85–92 Holsinger RM, Schnarr J, Henry P, Castelo VT,
Gorio A, Carmignoto G, Facci L, Finesso M (1980) Motor Fahnestock M (2000) Quantitation of BDNF mRNA
nerve sprouting induced by ganglioside treatment. Pos- in human parietal cortex by competitive reverse
sible implications for gangliosides on neuronal growth. transcription-polymerase chain reaction: decreased
Brain Res 197:236–241 levels in Alzheimer’s disease. Brain Res Mol Brain
Gorio A, Carmignoto G, Finesso M, Polato P, Nunzi MG Res 76:347–354
(1983a) Muscle reinnervation—II. Sprouting, synapse Iulita MF, Cuello AC (2014) Nerve growth factor meta-
formation and repression. Neuroscience 8:403–IN1 bolic dysfunction in Alzheimer’s disease and Down
Gorio A, Marini P, Zanoni R (1983b) Muscle reinnerva- syndrome. Trends Pharmacol Sci 35:338–348
tion—III. Motoneuron sprouting capacity, enhance- Iulita MF, Cuello AC (2016) The NGF metabolic pathway
ment by exogenous gangliosides. Neuroscience in the CNS and its dysregulation in down syndrome
8:417–429 and Alzheimer’s disease. Curr Alzheimer Res
Granieri E, Casetta I, Govoni V, Tola MR, Paolino E, 13:53–67
Rocca WA (1991) Ganglioside therapy and Guillain- Iulita MF, Do Carmo S, Ower AK, Fortress AM, Flores
Barre syndrome. A historical cohort study in Ferrara, Aguilar L, Hanna M, Wisniewski T, Granholm A-C,
Italy, fails to demonstrate an association. Neuroepi- Buhusi M, Busciglio J, Cuello AC (2014) Nerve
demiology 10:161–169 growth factor metabolic dysfunction in Down’s syn-
Grimes ML, Zhou J, Beattie EC, Yuen EC, Hall DE, drome brains. Brain 137:860–872
Valletta JS, Topp KS, Lavail JH, Bunnett NW, Mobley Iulita MF, Caraci F, Cuello AC (2016a) A link between
WC (1996) Endocytosis of activated TrkA: evidence nerve growth factor metabolic deregulation and
that nerve growth factor induces formation of signaling amyloid-beta-driven inflammation in Down syndrome.
endosomes. J Neurosci 16:7950–7964 CNS Neurol Disord Drug Targets 15:434–447
Hamburger V, Levi-Montalcini R (1949) Proliferation, Iulita MF, Ower A, Barone C, Pentz R, Gubert P,
differentiation and degeneration in the spinal ganglia Romano C, Cantarella RA, Elia F, Buono S,
of the chick embryo under normal and experimental Recupero M, Romano C, Castellano S, Bosco P, DI
conditions. J Exp Zool 111:457–501 Nuovo S, Drago F, Caraci F, Cuello AC (2016b) An
Hampel H, Mesulam MM, Cuello AC, Farlow MR, inflammatory and trophic disconnect biomarker profile
Giacobini E, Grossberg GT, Khachaturian AS, revealed in Down syndrome plasma: relation to cogni-
Vergallo A, Cavedo E, Snyder PJ, Khachaturian ZS tive decline and longitudinal evaluation. Alzheimers
(2018) The cholinergic system in the pathophysiology Dement 12:1132–1148
and treatment of Alzheimer’s disease. Brain Iulita MF, Bistue Millon MB, Pentz R, Aguilar LF, Do
141:1917–1933 Carmo S, Allard S, Michalski B, Wilson EN,
Hardy JA, Owen MJ, Goate AM, James LA, Haynes AR, Ducatenzeiler A, Bruno MA, Fahnestock M, Cuello
Rossor MN, Roques P, Mullan MJ (1989) Molecular AC (2017) Differential deregulation of NGF and
genetics of Alzheimer’s disease. Biochem Soc Trans BDNF neurotrophins in a transgenic rat model of
17:75–76 Alzheimer’s disease. Neurobiol Dis 108:307–323
Head E, Lott IT, Wilcock DM, Lemere CA (2016) Aging Jette N, Cole MS, Fahnestock M (1994) NGF mRNA is
in Down syndrome and the development of not decreased in frontal cortex from Alzheimer’s dis-
Alzheimer’s disease neuropathology. Curr Alzheimer ease patients. Brain Res Mol Brain Res 25:242–250
Res 13:18–29 Karami A, Eyjolfsdottir H, Vijayaraghavan S, Lind G,
Hebb DO (1949) The organization of behavior: a neuro- Almqvist P, Kadir A, Linderoth B, Andreasen N,
psychological theory. Wiley, Chapman & Hall, Blennow K, Wall A, Westman E, Ferreira D,
New York Kristoffersen Wiberg M, Wahlund LO, Seiger A,
Hefti F (1986) Nerve growth factor promotes survival of Nordberg A, Wahlberg L, DARREH-Shori T,
septal cholinergic neurons after fimbrial transections. J Eriksdotter M (2015) Changes in CSF cholinergic
Neurosci 6:2155–2162 biomarkers in response to cell therapy with NGF in
Hefti F, Weiner WJ (1986) Nerve growth factor and patients with Alzheimer’s disease. Alzheimers Dement
Alzheimer’s disease. Ann Neurol 20:275–281 11:1316–1328
Hendry IA, Stöckel K, Thoenen H, Iversen LL (1974) The Kordower JH, Gash DM, Bothwell M, Hersh L, Mufson
retrograde axonal transport of nerve growth factor. EJ (1989) Nerve growth factor receptor and choline
Brain Res 68:103–121 acetyltransferase remain colocalized in the nucleus
Hock C, Heese K, Müller-Spahn F, Hulette C, Rosenberg basalis (Ch4) of Alzheimer’s patients. Neurobiol
C, Otten U (1998) Decreased trkA neurotrophin Aging 10:67–74
9 Rita Levi-Montalcini, NGF Metabolism in Health and in the Alzheimer’s Pathology 143

Kromer LF (1987) Nerve growth factor treatment after increases choline acetyltransferase activity in develop-
brain injury prevents neuronal death. Science ing basal forebrain neurons. Mol Brain Res 1:53–62
235:214–216 Molnar M, Tongiorgi E, Avignone E, Gonfloni S,
Landi G, Ciccone A, D’alessandro R, Dossi BC, Ricci S, Ruberti F, Domenici L, Cattaneo A (1998) The effects
Simone IL (1994) Gangliosides and the Guillain-Barre of anti-nerve growth factor monoclonal antibodies on
syndrome. BMJ 308:1638 developing basal forebrain neurons are transient and
Ledeen RW (1984) Biology of gangliosides: neuritogenic reversible. Eur J Neurosci 10:3127–3140
and neuronotrophic properties. J Neurosci Res Mowla SJ, Pareek S, Farhadi HF, Petrecca K, Fawcett JP,
12:147–159 Seidah NG, Morris SJ, Sossin WS, Murphy RA (1999)
Ledeen RW, Yu RK (1982) Gangliosides: structure, isola- Differential sorting of nerve growth factor and brain-
tion, and analysis. Methods Enzymol 83:139–191 derived neurotrophic factor in hippocampal neurons. J
Levi-Montalcini R (1964) Growth control of nerve cells by Neurosci 19:2069–2080
a protein factor and its antiserum: discovery of this Mufson EJ, Lavine N, Jaffar S, Kordower JH, Quirion R,
factor may provide new leads to understanding of Saragovi HU (1997) Reduction in p140-TrkA receptor
some neurogenetic processes. Science 143:105–110 protein within the nucleus basalis and cortex in
Levi-Montalcini R (1988) In praise of imperfection : my Alzheimer’s Disease. Exp Neurol 146(1):91–103.
life and work. Basic Books, New York https://doi.org/10.1006/EXNR.1997.6504
Levi-Montalcini R (1997) Les conséquences de la destruc- Mutoh T, Tokuda A, Miyadai T, Hamaguchi M, Fujiki N
tion d’un territoire d’innervation périphérique sur le (1995) Ganglioside GM1 binds to the Trk protein and
développement des centres nerveux correspondants regulates receptor function. Proc Natl Acad Sci U S A
dans l’embryon de poulet. In: The Saga of the nerve 92:5087–5091
growth factor: preliminary studies, discovery, further Nitsch RM, Slack BE, Wurtman RJ, Growdon JH (1992)
development. World Scientific Release of Alzheimer amyloid precursor derivatives
Levi-Montalcini R, Aloe L (1985) Differentiating effects stimulated by activation of muscarinic acetylcholine
of murine nerve growth factor in the peripheral and receptors. Science 258:304–307
central nervous systems of Xenopus laevis tadpoles. Nitsch RM, Farber SA, Growdon JH, Wurtman RJ (1993)
Proc Natl Acad Sci U S A 82:7111–7115 Release of amyloid beta-protein precursor derivatives
Loewi O (1921) Über humorale übertragbarkeit der by electrical depolarization of rat hippocampal slices.
Herznervenwirkung. Pflüger’s Archiv für die gesamte Proc Natl Acad Sci U S A 90:5191–5193
Physiologie des Menschen und der Tiere 189:239–242 Pearson RC, Sofroniew MV, Cuello AC, Powell TP,
Lott IT (1982) Down’s syndrome, aging, and Alzheimer’s Eckenstein F, Esiri MM, Wilcock GK (1983) Persis-
disease: a clinical review. Ann N Y Acad Sci tence of cholinergic neurons in the basal nucleus in a
396:15–27 brain with senile dementia of the Alzheimer’s type
Lott IT, Head E (2019) Dementia in Down syndrome: demonstrated by immunohistochemical staining for
unique insights for Alzheimer disease research. Nat choline acetyltransferase. Brain Res 289:375–379
Rev Neurol 15:135–147 Pedraza CE, Podlesniy P, Vidal N, Arevalo JC, Lee R,
Maysinger D, Herrera-Marschitz M, Carlsson A, Hempstead B, Ferrer I, Iglesias M, Espinet C (2005)
Garofalo L, Cuello AC, Ungerstedt U (1988) Striatal Pro-NGF isolated from the human brain affected by
and cortical acetylcholine release in vivo in rats with Alzheimer’s disease induces neuronal apoptosis
unilateral decortication: effects of treatment with mediated by p75NTR. Am J Pathol 166:533–543
monosialoganglioside GM1. Brain Res 461:355–360 Peng S, Wuu J, Mufson EJ, Fahnestock M (2005) Precur-
Maysinger D, Herrera-Marschitz M, Ungerstedt U, Cuello sor form of brain-derived neurotrophic factor and
AC (1990a) Acetylcholine release in vivo: effects of mature brain-derived neurotrophic factor are decreased
chronic treatment with monosialoganglioside GM1. in the pre-clinical stages of Alzheimer’s disease. J
Neuropharmacology 29:151–159 Neurochem 93:1412–1421
Maysinger D, Herrera-Marschitz M, Ungerstedt U, Cuello Pentz R, Iulita MF, Juan Fortea A, Cuello C (2019)
AC (1990b) Effects of treatment with Identifying Alzheimer’s disease in Down syndrome
microencapsulated monosialoganglioside GM1 on cor- with NGF metabolism: hope for better treatment and
tical and striatal acetylcholine release in rats with cor- diagnosis? T21RS Sci Soc Bull 1–3
tical devascularizing lesions. Neurosci Lett Pentz R, Iulita MF, Ducatenzeiler A, Bennett DA, Cuello
118:252–256 AC (2020) The human brain NGF metabolic pathway
Maysinger D, Herrera-Marschitz M, Goiny M, is impaired in the pre-clinical and clinical continuum of
Ungerstedt U, Claudio Cuello, A. (1992) Effects of Alzheimers disease. Mol Psychiatry:1–15. https://doi.
nerve growth factor on cortical and striatal acetylcho- org/10.1038/S41380-020-0797-2
line and dopamine release in rats with cortical Rabin SJ, Mocchetti I (1995) GM1 ganglioside activates
devascularizing lesions. Brain Research 577:300–305 the high-affinity nerve growth factor receptor trkA. J
Mobley WC, Rutkowski JL, Tennekoon GI, Gemski J, Neurochem 65:347–354
Buchanan K, Johnston MV (1986) Nerve growth factor
144 A. C. Cuello

Ramón y Cajal S, May RM (1928) Degeneration & regen- and other areas of the rat brain. J Neurochem
eration of the nervous system. Oxford University 45:1021–1026
Press, Humphrey Milford, London Stephens PH, Tagari PC, Garofalo L, Maysinger D,
Sabatini MT, Levi-Montalcini R, Angeletti PU (1966) Piotte M, Cuello AC (1987) Neural plasticity of basal
[Early effects of anti-nerve growth factor serum on forebrain cholinergic neurons: effects of gangliosides.
the nerve cell]. Ann Ist Super Sanita 2:349–355 Neurosci Lett 80:80–84
Salehi A, Delcroix JD, Belichenko PV, Zhan K, Wu C, Stewart SS, Appel SH (1988) Trophic factors in neuro-
Valletta JS, Takimoto-Kimura R, Kleschevnikov AM, logic disease. Annu Rev Med 39:193–201
Sambamurti K, Chung PP, Xia W, Villar A, Campbell Svennerholm L (1980) Ganglioside designation. In: Struc-
WA, Kulnane LS, Nixon RA, Lamb BT, Epstein CJ, ture and function of gangliosides. Springer
Stokin GB, Goldstein LS, Mobley WC (2006) Tettamanti G (1988) Towards the understanding of the
Increased App expression in a mouse model of physiological role of gangliosides. J New Trends Gan-
Down’s syndrome disrupts NGF transport and causes glioside Res Neurochem Neuroregenerative Aspects
cholinergic neuron degeneration. Neuron 51:29–42 14:625–646
Samson J, Fiori MJB (1994) Gangliosides and the Venero JL, Knusel B, Beck KD, Hefti F (1994) Expression
Guillain-Barre syndrome. BMJ Clin Res 308 of neurotrophin and trk receptor genes in adult rats
(6944):1638 with fimbria transections: effect of intraventricular
Seiler M, Schwab ME (1984) Specific retrograde transport nerve growth factor and brain-derived neurotrophic
of nerve growth factor (NGF) from neocortex to factor administration. Neuroscience 59:797–815
nucleus basalis in the rat. Brain Res 300:33–39 Wanleenuwat P, Iwanowski P, Kozubski W (2020)
Sofroniew MV, Pearson RC, Eckenstein F, Cuello AC, Antiganglioside antibodies in neurological diseases. J
Powell TP (1983) Retrograde changes in cholinergic Neurol Sci 408:116576
neurons in the basal forebrain of the rat following Whitehouse PJ, Price DL, Struble RG, Clark AW, Coyle
cortical damage. Brain Res 289:370–374 JT, Delon MR (1982) Alzheimer’s disease and senile
Sofroniew MV, Pearson RC, Cuello AC, Tagari PC, dementia: loss of neurons in the basal forebrain. Sci-
Stephens PH (1986) Parenterally administered GM1 ence 215:1237–1239
ganglioside prevents retrograde degeneration of cho- Williams LR, Varon S, Peterson GM, Wictorin K,
linergic cells of the rat basal forebrain. Brain Res Fischer W, Bjorklund A, Gage FH (1986) Continuous
398:393–396 infusion of nerve growth factor prevents basal fore-
Sofroniew MV, Galletly NP, Isacson O, Svendsen CN brain neuronal death after fimbria fornix transection.
(1990) Survival of adult basal forebrain cholinergic Proc Natl Acad Sci U S A 83:9231–9235
neurons after loss of target neurons. Science Wisniewski HM, Wrzolek M (1988) Pathogenesis of amy-
247:338–342 loid formation in Alzheimer’s disease, Down’s syn-
Stephens PH, Cuello AC, Sofroniew MV, Pearson RC, drome and scrapie. Ciba Found Symp 135:224–238
Tagari P (1985) Effect of unilateral decortication on Yates CM, Simpson J, Maloney AF, Gordon A, Reid AH
choline acetyltransferase activity in the nucleus basalis (1980) Alzheimer-like cholinergic deficiency in Down
syndrome. Lancet 2:979
NGF and the Amyloid Precursor Protein
in Alzheimer’s Disease: From Molecular 10
Players to Neuronal Circuits

Viviana Triaca, Francesca Ruberti, and Nadia Canu

Abstract (MRI) and positron emission tomography


Alzheimer’s disease (AD), one of the most (PET) studies in mild cognitive impairment
common causes of dementia in elderly people, (MCI) and AD patients highlight the early
is characterized by progressive impairment in involvement of BFCNs in MCI and the early
cognitive function, early degeneration of basal phase of AD. These data support the choliner-
forebrain cholinergic neurons (BFCNs), gic and neurotrophic hypotheses of AD and
abnormal metabolism of the amyloid precursor suggest new targets for AD therapy.
protein (APP), amyloid beta-peptide (Aβ) Different mechanisms account for selective
depositions, and neurofibrillary tangles. vulnerability of BFCNs to AD pathology, with
According to the cholinergic hypothesis, dys- regard to altered metabolism of APP and tau.
function of acetylcholine-containing neurons In this review, we provide a general overview
in the basal forebrain contributes markedly to of the current knowledge of NGF and APP
the cognitive decline observed in AD. In addi- interplay, focusing on the role of APP in
tion, the neurotrophic factor hypothesis posits regulating NGF receptors trafficking/signal-
that the loss nerve growth factor (NGF) signal- ling and on the involvement of NGF in
ling in AD may account for the vulnerability to modulating phosphorylation of APP, which
atrophy of BFCNs and consequent impairment in turn controls APP intracellular trafficking
of cholinergic functions. Though acetylcholin- and processing. Moreover, we highlight the
esterase inhibitors provide only partial and consequences of APP interaction with
symptomatic relief to AD patients, emerging p75NTR and TrkA receptor, which share the
data from in vivo magnetic resonance imaging same binding site within the APP juxta-
membrane domain. We underline the impor-
tance of insulin dysmetabolism in AD pathol-
V. Triaca · F. Ruberti
Institute of Biochemistry and Cell Biology (IBBC), ogy, in the light of our recent data showing that
National Research Council (CNR), Campus A. Buzzati- overlapping intracellular signalling pathways
Traverso, Monterotondo, RM, Italy stimulated by NGF or insulin can be compen-
e-mail: viviana.triaca@cnr.it; francesca.ruberti@cnr.it
satory. In particular, NGF-based signalling is
N. Canu (*) able to ameliorates deficiencies in insulin sig-
Institute of Biochemistry and Cell Biology (IBBC),
nalling in the medial septum of 3Tg-AD
National Research Council (CNR), Campus A. Buzzati-
Traverso, Monterotondo, RM, Italy mice. Finally, we present an overview of
NGF-regulated microRNAs (miRNAs).
Department of System Medicine, Section of Physiology,
University of Rome “Tor Vergata”, Rome, Italy These small non-coding RNAs are involved
e-mail: nadia.canu@uniroma2.it in post-transcriptional regulation of gene
# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 145
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_10
146 V. Triaca et al.

expression, and we focus on a subset that are abnormal extracellular accumulation and deposi-
specifically deregulated in AD and thus poten- tion in the brain and the cerebral vessel [cerebral
tially contribute to its pathology. amyloid angiopathy] of the amyloid-β peptide
(Aβ) with 40 or 42 amino acids (Aβ 40 and Aβ
42). These peptides are the products of the physi-
ological metabolism of the APP after its sequen-
10.1 Alzheimer’s Disease Pathology tial cleavage by the aspartyl protease β-site APP
cleaving enzyme 1 (BACE1, also termed
In 1906, the German clinical psychiatrist and β-secretase) and the γ-secretase complex (includ-
neuroanatomist Alois Alzheimer first identified ing PS1 and PS2) in neurons. While Aβ 40 is the
and described a ‘peculiar severe disease process most abundant form, neurotoxicity is mainly
of the cerebral cortex’ in Auguste Deter, a mediated by Aβ 42. NFTs are essentially made
51-year-old woman, who rapidly developed of paired helical filaments (PHFs), which are
dementia typical of older individuals. This dis- fibrils of different morphology mainly composed
ease process, which would become known as of the abnormal misfolded and hyperpho-
Alzheimer’s disease (AD), is today the most com- sphorylated microtubule-associated protein tau.
mon cause of dementia among elderly people. Invariably accompanying NFTs are the neuropil
AD can be either sporadic or familial. Sporadic threads (i.e., abnormal neurites containing, con-
AD (SAD) usually occurs after age 65 and is the trary to normal neurites, straight and paired heli-
most common form of AD. Familial AD (FAD), cal filaments composed of tau and ubiquitin;
which typically presents in middle age, is a very Serrano-Pozo et al. 2011).
rare genetic condition caused by a mutation in the Glial responses have come to assume a pivotal
type 1 transmembrane protein APP, Presenilin role in both clinical expression and progression of
1 (PS1) and 2 (PS2) genes, whose protein cognitive decline in AD. Indeed, reactive
products regulate APP processing (Dorszewska astrocytes and activated microglial cells normally
et al. 2016), and increase copy number for APP decorate amyloid plaques, suggesting that Aβ is a
gene, as in Down syndrome (DS). major trigger. However, glial responses show a
Clinical manifestations of AD include spatial positive relationship also with NFT and contrib-
navigation, memory, thinking, comprehension, ute to the ongoing neurodegeneration (Serrano-
learning capacity loss, as well as significant dete- Pozo et al. 2011).
rioration of cognitive function as compared to Neuronal loss is the cause of pathological cor-
normal aging-related changes. The weakening of tical atrophy as revealed by Nissl or NeuN
cognitive function is generally associated with, staining of brain sections, computed tomography
and sometimes precedes, deterioration in emo- (CT) scan and MRI analysis in vivo. Its regional
tional control, social behaviour, or motivation. and laminar distribution meets that of NFTs. Not-
Thus, patients progress from normal brain ageing withstanding, within the same region neuronal
to mild cognitive impairment (MCI) followed by loss overcomes the numbers of NFTs, and
increasing dementia severity (i.e., mild, moderate, neurons bearing NFT may be still alive. These
and severe) in SAD. findings led to considering the neuronal loss a
better correlate of cognitive deficits than the num-
Anatomopathological Hallmarks: APP, Tau, ber of NFTs. Synapse loss is another causative
Glial Response, Neuronal agent of cortical atrophy in AD. Synapse degen-
and Synaptic Loss The main neuropathological eration is responsible for the loss of structural
features of AD include amyloid plaques, neurofi- plasticity and inefficiency of the neuronal net-
brillary tangles (NFT), glial responses, and neu- work in AD, as shown post mortem by
ronal and synaptic loss (Serrano-Pozo et al. immunohistochemistry using antibodies against
2011). The amyloid plaques result from the pre- or post-synaptic proteins, by electron
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 147

microscopy, and by 18F-fluorodeoxyglucose pos- Neurotrophic Hypothesis Intimately connected


itron emission tomography ([18F] FDG PET) in to the cholinergic hypothesis is the neurotrophic
human subjects (Kashyap et al. 2019). Interest- hypothesis (Appel 1981; Chen et al. 2018).
ingly, network activity seems to be disproportion- According to this, loss of a neurotrophic factor
ately weakened in the early stages of AD, [in particular NGF (although today we refer to the
possibly due to the regional specific effects of loss of NGF-signalling)] in the AD brain may
altered metabolism of APP and tau on synapses account for the dysfunction of cholinergic system.
(Spires-Jones and Hyman 2014). This hypothesis has been recently reinforced by
data from single-cell neuron-specific gene expres-
sion profiles and functional network analysis that
have predicted a strong association of the
10.2 Past and Current
neurotrophin signalling pathway with neuronal
Aetiopathological Hypotheses
vulnerability to AD (Roussarie et al. 2020).
in Sporadic Alzheimer’s
Disease (SAD)
Aβ and Aβ-Oligomer (AβO) Cascade
Hypothesis In 1992, it was proposed that the
There are several hypotheses regarding the causes
accumulation of fibrillar Aβ peptides in the
of SAD.
brain—as result of an imbalance between Aβ
production and Aβ clearance—is a crucial step
Cholinergic Hypothesis In the late 1970s,
in AD pathogenesis and that NFT, cell loss, vas-
researchers enunciated the ‘cholinergic hypothe-
cular damage, and dementia all result from Aβ
sis of age-associated memory dysfunction’ that
deposition (Hardy and Higgings 1992). This
was extended also to AD. According to this
hypothesis relied on evidence that people carry-
hypothesis, AD dementia was attributed to a fail-
ing mutations in three genes involved in Aβ pro-
ure of the cholinergic system (Davies and
duction (APP, PS1, PS2) and patients with DS
Maloney 1976). One class of symptom-
(overexpressing APP due to a triplication the
alleviating medications, such as the cholinester-
gene coding for APP, which is located on chro-
ase inhibitors, are in common use today. The
mosome 21) experienced memory loss and neu-
cholinergic hypothesis implied that the altered
rological changes characteristic of
physiology of basal forebrain cholinergic neurons
AD. Moreover, the generation of different APP
(BFCNs) contributed to AD pathology. A sugges-
or APP/PS1 transgenic mouse models developing
tion later confirmed by finding that a massive
some of the main features of FAD (Puzzo et al.
neurons reduction in the nucleus basalis of
2014) reinforced the Aβ hypothesis. However, the
Meynert (NBM) (Whitehouse et al. 1981, 1982)
evidence that Aβ causes SAD is complex and
preceded by years the cholinergic impaiments
hazy, thus prompting criticisms about the central
characteristic of AD (Cuello et al. 2007; Schliebs
role of the Aβ cascade in AD pathogenesis.
and Arendt 2011).
Among these are the weak temporal and
Recently this concept has been questioned by
anatomical correlation between Aβ deposition,
the only modest and transient cognitive relief
neuronal death, and clinical symptoms in SAD,
provided by cholinesterase inhibitors, possibly
and the finding that all attempts to develop A-
because of late administration to severe AD
β-targeting drugs to treat AD have been mostly
patients. On the other hand, novel drugs targeting
unsuccessful so far.
the cholinergic system, like muscarinic receptor
In 1998, the Aβ cascade evolved into the AβO
agonists (Fisher 2012; Verma et al. 2018), as well
hypothesis. According to this hypothesis, soluble
as deep brain stimulation (DBS) of basal fore-
and oligomeric forms of Aβ trigger the cascade of
brain nuclei (Mankin and Fried 2020), are
events leading to AD (Cline et al. 2018). The
among the most promising approaches for early
main issue related to the AβO hypothesis is the
phase of AD therapy.
148 V. Triaca et al.

time of appearance and exact identity of the more to-cell spreading of tau pathology (Takahashi
toxic AβO species. This aspect deserves further et al. 2015).
investigation to develop Aβ-oriented therapeutics
against the correct target at the desired time. An Integrated Hypothesis for AD An emerging
Moreover, Aβ monomer homeostasis is essential point of view sustains that AD pathogenesis ‘is
for the synaptic function and it should be taken not simply a neuron-centric, linear cascade
into consideration since the Aβ ‘loss-of-function’ initiated by Aβ/AβO and leading to dementia,
in neurons is a key determinant in AD pathophys- but rather a long, complex cellular phase
iology (Giuffrida et al. 2009). consisting of feedback and feedforward responses
of astrocytes, microglia, and vasculature’
Tau Hypothesis In contrast to plaque deposition, (De Strooper and Karran 2016). A series of events
tau pathology correlates more closely with neuro- elicit reactive gliosis and neuroinflammation.
nal loss and with the severity, and duration of Among these are ROS activation, impaired astro-
cognitive deficits (Arriagada et al. 1992; Bierer cytic clearance of Aβ [associated or not to induc-
et al. 1995; Giannakopoulos et al. 2003; Bejanin tion of microglial phagocytosis (Batarseh et al.
et al. 2017). Tau pathology [due to an imbalance 2016; Nielsen et al. 2010)], AβO-mediated switch
of tau isoforms for splicing deficits and/or aber- in microglial phenotype to a pro-inflammatory
rant post-translational modifications (hyperpho- phenotype leading to improper tumour necrosis
sphorylation, glycosylation, and truncation)] factor secretion (El-Shimy et al. 2015). In turn,
disrupts neuronal homeostasis by affecting the reactive microglia and astrocytes secrete numer-
transport system (of the neurotrophic receptors, ous pro-inflammatory cytokines, disturb the sig-
APP, etc.), the cytoskeletal system, signalling nalling of insulin, alter astrocytic insulin
system, and mitochondrial integrity, and secretion, contribute to glucose hypometabolism,
culminates in aberrant cell death even in the impair neurotrophic supply, impact the expres-
absence of NFTs (Iqbal et al. 2016). Moreover, sion of excitatory amino acid transporters 1 and
tau pathology usually first appears in discrete and 2, thus impairing synaptic plasticity. This point of
specific areas to later spread trans-synaptically, to view has flowed into a more integrated hypothesis
other brain regions, eventually affecting circuit for SAD (De Felice 2013). According to this
functional connectivity and physiology (Frost hypothesis, SAD results from the accumulation
et al. 2009). For several years, Aβ and tau deposi- over the time of the insults to the brain and
tion have been seen as two distinct pathogno- peripheral organs that cause an increase of AβO
monic signs and causative agents of levels.
AD. However, a huge amount of experimental
evidence has shown a mutual control between
Aβ and tau (Selkoe 2011). Indeed, the high affin-
10.3 BFCNs’ Vulnerability in AD
ity between Aβ and tau induces the assembly of a
Target Circuits
‘soluble complex that may promote self-
aggregation of both into the insoluble forms’
Aβ and tau pathologies impair the circuits
(Guo et al. 2006). Moreover, AβO seeds the for-
involved in spatial orientation, learning, and
mation of highly toxic tau oligomers, redistributes
memory. Nevertheless showing different spatial/
tau to dendrites, and drives the synaptic spread of
temporal distributions. Aβ deposition begins
tau (Bilousova et al. 2016). Also, the extracellular
within the cortex (frontal, temporal, and occipital)
region of APP, with or without FAD mutations,
and later attacks the hippocampus, the EC, insular
forms a complex with tau fibrils. This complex
and cingulate cortices, and amygdala. Then, it
promotes the uptake of tau fibrils into the cells
colonizes several subcortical regions, including
and causes the seed-dependent intracellular tau
BFCNs, and later the substantia nigra, brainstem
aggregation. This latter, in turn, favours the cell-
nuclei, and also the molecular layer of the
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 149

cerebellum (Thal et al. 2002). NFT deposition is In line with the cholinergic and neurotrophic
quite different from that for Aβ plaques. NFT hypotheses, BFCNs are selectively vulnerable to
deposition exhibits the identical regional pattern Aβ and tau oligomers due to their dependence on
as neurodegeneration and occurs in an exceed- NGF, though the underlying mechanism is not
ingly predictable, stereotypical spatiotemporal well known. If the availability of NGF is ham-
sequence of six stages (Stages I–VI) (Braak and pered due to an imbalance in maturation/degrada-
Braak 1991). tion of NGF, and/or a concomitant increase in
In line with the foremost recent clinical precursor pro-NGF, BFCNs undergo progressive
findings, current experimental approaches focus atrophy (Cuello et al. 2007). Further they may
on specific neuronal vulnerability and network become more vulnerable (Allen et al. 2011) or
deficit as the main driver of MCI to AD progres- even contribute to the emergence of Aβ and tau
sion and AD pathology occurrence, replacing the pathology (Calissano et al. 2010; Capsoni et al.
old view of AD as a single-molecule single-cell 2000), finally leading to dementia exacerbation
neurodegenerative disease. Recently, and/or anticipation. Recently, BFCN vulnerabil-
neurodegeneration in AD has been demonstrated ity has been demonstrated to occur also because
to specifically affect the circuits of the basal fore- of the higher metabolic activity required by their
brain in a predictable manner (Schmitz et al. recruitment in reinforced feedback, during rapid
2018). reconfiguration of cortical state and plasticity
BFCNs include the medial septum (Ch1), the (Hangya et al. 2015). Moreover, their requirement
vertical and horizontal diagonal Bands of Broca of acetyl-CoA for the biosynthesis of both ATP
(VDB or Ch2 and HDB or Ch3, respectively), the and acetylcholine may concur to their higher sus-
substantia innominata, and the NBM (Ch4). As ceptibility to AD pathology (Szutowicz et al.
described by anterograde, retrograde, and trans- 2013; Wurtman 1992).
synaptic labellings, primates’ cholinergic inputs BFCN atrophy was also reported by the AD
to the hippocampus arise from the medial septum neuroimaging initiative (ADNI), demonstrating
and vertical limb of the diagonal band of Broca that BF abnormalities precede and predict cortical
(DbB) innervates the hippocampal target neurons degeneration in AD subjects, as also
within the CA1 and dentate gyrus, both through demonstrated by cortical thinning as detected by
the fornix and a non-fornical route (reviewed in PET in MCI patients, but not in cognitively nor-
Ballinger et al. 2016). mal people (Schmitz et al. 2018) and by increased
Due to their intrinsic anatomy, biochemistry, Aβ burden in the prefrontal cortex (Kerbler et al.
and neurotransmitter release, BFCNs have higher 2015). Interestingly, DbB and frontal cortex dam-
propensity to accumulate disease-related proteins age correlate well with cognitive decline, with
(Aβ and tau specifically). In particular, early Meynert’s nucleus deficits being more associated
accumulation of intraneuronal Aβ in adult life with impaired delayed recall in MCI subjects
and Aβ oligomerization during the aging process (Grothe et al. 2010), particularly in women
makes BFCNs more prone to AD degeneration (Cantero et al. 2016). Also, secondary BFCN
(Baker-Nigh et al. 2015). atrophy occurs in a late stage as a result of neuron
Alteration of cortical cholinergic neurons and shrinkage in their cortical target circuits (Pearson
tau accumulation in the NBM are observed in et al. 1983; Vogels et al. 1990). Of note, the
patients > 60 years and found increased in elderly global cognitive deterioration rate can be
people, particularly those affected by AD (Geula predicted based on the volume of basal choliner-
et al. 2008; Sassin et al. 2000). Accordingly, gic nuclei, with higher sensitivity than that
in vivo MRI findings confirmed the reduced observed in hippocampal neurons (Teipel et al.
BFCNs’ volume in MCI and AD (Kilimann 2005).
et al. 2014; Teipel et al. 2005, 2011; Grothe Moreover, the medial septum is also able to
et al. 2010). reconfigure circuit function restoring the residual
150 V. Triaca et al.

memory in case of a fornical deficit (Zatorre et al. APP is a type-I transmembrane protein
2012), further pinpointing BFCNs as a promising concentrated in the synapse, synthesized in the
target for AD therapy. The medial septum net- endoplasmic reticulum (ER), transported to the
work has been shown to modulate hippocampal Golgi/trans-Golgi network (TGN), and inserted
memory via regulated theta oscillations by low in the plasma membrane (Caporaso et al. 1994).
firing neurons (Petersen and Buzsáki 2020; Cell surface APP can be internalized for
Tsanov 2017; Patel et al. 2012) and gamma endosomal recycling to the cell surface, for late
oscillations (Leung and Ma 2018), and to inhibit endosomal/lysosomal degradation, or for trans-
the occurrence of an abnormal excitatory status port from endosomes back to the TGN (reviewed
(Colom et al. 2006). In line, wave induction by in Eggert et al. 2018). By regulating APP phos-
DBS resulted in ameliorated cognition (Jeong phorylation at specific sites, NGF may dictate
et al. 2014; Stypulkowski et al. 2017), and distur- APP trafficking and processing in neurons
bance in the theta and gamma network is pro- (Matrone et al. 2011; Triaca et al. 2016). It is
posed as an early AD biomarker (Kitchigina well known that APP is cleaved in the
2018). Taken together, these results support the amyloidogenic pathway by BACE1 at an extra-
hypothesis that BFCNs dysfunction is an early cellular site adjacent to the plasma membrane to
event, significantly contributing to the AD con- yield the βCTF fragment (transmembrane
tinuum, and that re-activation of BF circuits may β-carboxyterminal), which is further cleaved by
lead to a significant improvement of synaptic γ-secretase complex [a complex comprising
activity and cognition. presenilin, nicastrin, anterior pharynx defective
1 (APH-1), and presenilin enhancer 2 (PEN-2)]
to release the soluble APP intracellular cytoplas-
10.4 APP and NGF Interplay mic domain (AICD) fragment, Aβ 40, and Aβ
42 peptides. APP can also be cleaved though the
As already mentioned, the viability and physiol- non-amyloidogenic pathway by cell surface
ogy of BFCNs rely on NGF, which is initially α-secretase (remarkably ADAM10). Cleavage
produced as precursor pro-NGF by BFCN targets by α-secretase results in the release of the
in the hippocampus and cortical tissues. NGF, ectodomain of APP-soluble fragment (sAPPα),
here produced, binds to its high-affinity TrkA and the simultaneous generation of a membrane
and the low-affinity (p75NTR) receptors present retained C-terminal fragment (APP-CTF). This
at the distal end of BFCNs axons and, once latter can be further processed by γ-secretase
internalized, it is retrogradely transported to the (reviewed by Zhang et al. 2011). Recent studies
cell body. Internalization and transport of the have indicated that, while APP α-secretases
NGF-receptors complex to the cell body are cleavage occurs at the plasma membrane, proteo-
required for acetylcholine neurotransmitter lytic processing by BACE1, and then Aβ produc-
release to initiate responses in the BFCN targets. tion, requires APP internalization from the cell
In AD cholinergic neurons, the NGF/NGF recep- surface to intracellular, acidic, endocytic
tor complex is affected at many levels including compartments (Koo and Squazzo 1994; Selkoe
the following: (1) unbalanced NGF maturation et al. 1996; Haass et al. 2012).
with an increase in the pro-NGF; (2) reduced
TrkA/p75NTR ratio; (3) increased pro-apoptotic
activity of p75NTR via its interaction with 10.4.1 APP Control of the NGF
pro-NGF; (4) stimulation of p75NTR by A- Receptor Trafficking
β-peptide binding; (5) loss of TrkA receptor that and Signalling
increases the affinity of pro-NGF for p75NTR,
thus favouring the pro-apoptotic activity of Several pieces of evidence demonstrated a mutual
p75NTR in cooperation with sortilin (reviewed influence between trafficking and signalling of
by Mufson et al. 2019; Ioannou and Fahnestock NGF receptors with the trafficking and signalling
2017). of APP. Besides being the precursor of Aβ
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 151

peptides, APP is a protein whose physiological endosomal compartment and that Rab5 increased
functions are likely lost in AD; thus, the loss- activation plays a paramount role in perturbing
function of APP contributes to AD pathogenesis. NGF trafficking and signalling in AD and DS,
APP has been implicated in neuronal plasticity, thus leading to BFCN atrophy. Since APP
synapse formation, iron and calcium metabolism, interacts with both p75NTR and TrkA
cell adhesion, and also the regulation of traffick- (Fombonne et al. 2009; Matrone et al. 2011),
ing of BACE1, PS1 (Kamal et al. 2001; Liu et al. Zang and collaborators asked whether the mecha-
2009), and choline transporter (Wang et al. 2007). nism of APP-mediated impairment of NGF retro-
Beyond its own metabolism, APP somehow grade transport relied on altered trafficking of its
interacts with the retrograde axonal transport TrkA and p75NTR receptors. Thus, they
mechanism in the BFCNs that are among the investigated the effects of APP on the p75NTR
earliest neurons to degenerate in and TrkA trafficking and the downstream signal-
AD. Particularly it has been reported that ling under NGF stimulation. Interestingly, they
increased APP gene dosage [triplicated in Down found that APP regulates cell surface levels of
syndrome (DS)] and overexpression of APP strik- the two NGF receptors. In particular, down-
ingly affect NGF retrograde transport and cause regulation and over-expression of APP resulted
degeneration of BFCNs in a DS mouse model. in increased or decreased NGF/NGF receptors’
Also other transmembrane APP_CTF may likely endocytosis, respectively (Zhang et al. 2013).
negatively impact NGF retrograde transport Moreover, they showed that APP deficiency, by
(Salehi et al. 2006) by perturbing the endocytic- enhancing the NGF-mediated PI3K/Akt and
lysosomal pathway. Further investigations to MAPK, pathways, resulted in an increased neu-
identify the mechanism by which APP and its ronal differentiation and survival in response to
various products impair the endocytic-lysosomal NGF. It follows that the down-regulation of these
pathway were performed in PC12 cells, cultured NGF stimulated pathways, as occurring when
rat BFCNs, and primary BFCNs fromTs65Dn APP is overexpressed, may result in early BFCN
mice, a DS mouse model. It has been found that suffering and eventually cell death.
Rab5 (major regulator of endosome biogenesis)
positive endosomes were enlarged in cultured
BFCNs from Ts65Dn mice, and endosomal size 10.4.2 Bad and Good Interactions
was restored by APP RNAi, indicating that APP of APP with NGF Receptors
is directly involved in such event. Moreover,
overexpression of full-length wild-type and As already mentioned, the APP can interact with
mutant forms of APP, as well as of the β-CTF p75NTR and TrkA NGF-receptors. However, the
fragment, induced the activation of Rab5 and effect of such different interactions regarding sur-
enlargement of Rab5 endosomes in cultured vival and APP metabolism is different.
PC12 and rat BFCNs (Xu et al. 2016). Recently, Fombonne et al. (2009) showed that APP and
super-resolution structured illumination micros- p75NTR interact directly, using different
copy (SR-SIM) and transmission electron micros- techniques (including the yeast two-hybrid
copy (TEM) have shown that endosome had a assay, bioluminescence, resonance, energy trans-
normal size but appeared clustered in fibroblasts fer, confocal microscopy, and coimmunopreci-
and IPCS-derived neurons from individual DS, pitation in vitro and in vivo). By deletion
and BFCN cultures from DS human subjects, studies, they found that β-CTF (C99) (APP597-
suggesting that APP overexpression causes a 695) but not C83 fragment (APP613-695) is suf-
‘traffic jam’ in the endosomal compartment ficient for this interaction, indicating that the Aβ
(Botté et al. 2020). region of APP is required for APP/p75NTR inter-
Altogether, these findings suggest that action. Moreover, they found that the
overexpression of APP perturbs the Rab5 positive APP/p75NTR complex is negatively modulated
152 V. Triaca et al.

by NGF and Aβ peptide, both in vitro and in vivo. complex. We have ruled out strong candidates
Indeed, colocalization of APP and p75NTR was such as p75NTR, ShcC, and Mint-2 in bridging
reduced in a mouse model of AD expressing a APP and TrkA, since their co-expression with
high level of Aβ (PDAPP, J20 line). The authors APP and TrkA BIFC plasmids did not lead to an
suggested that the APP/p75NTR complex is increased BIFC signal (Canu et al. 2017a, b).
blocked in AD as a consequence of the likely Likewise, we suggested that a potential partner
more favourite binding of Aβ to APP. Interest- may be sortilin. Sortilin is a member of the
ingly, they demonstrated that NGF and Aβ pep- Vps10p-domain receptor family that acts as a
tide by preventing this association block its receptor of neurotrophic factors, mediates traf-
downstream effects. In detail, they demonstrated ficking from the secretory pathway to endosomes,
that the interaction between APP and p75NTR and retrograde transport to the TGN (Nielsen
‘shifts the effect of APP from a trophic one, et al. 2001). Particularly, sortilin binds TrkA,
with the production of sAPPα and the Fe65- enables TrkA anterograde axonal transport,
dependent transcriptional effects, to an anti- enhances neurotrophin signalling (Vaegter et al.
trophic one, with the reduction in sAPPα and 2011), and interacts with APP to influence its
the increase in Aβ peptide production’ production and cellular uptake (Gustafsen et al.
(Fombonne et al. 2009). The negative impact of 2013). Despite this physiological role, sortilin
the APP/p75NTR interaction was further acts as a co-receptor with p75NTR to mediate
supported by the finding that co-expression of Pro-NGF-induced pro-death signalling (Nykjaer
APP and p75NTR in neuroblastoma cells line et al. 2004). APP/TrkA complex is confined to the
‘enhanced caspase-3 dependent cells death over plasma membrane, ER, Golgi, and endocytic
that induced by either APP or p75NTR alone’ vesicles, as evidenced by the colocalization of
(Fombonne et al. 2009). Equally, the APP may the APP/TrkA complex with compartment-
also disturb p75NTR processing and signalling specific markers by both BiFC analysis
by jamming the endocytic pathway (see above). (in HEK-293 cells expressing exogenous APP
Regarding APP/TrkA interaction (Matrone and TrkA) and PLA assay in rat septal neuronal
et al. 2011; Zhang et al. 2013; Triaca et al. culture. Interestingly, drugs that perturb cellular
2016), we recently investigated this issue by trafficking block this interaction consistently with
bimolecular fluorescence complementation the known physiological transport of APP and
(BiFC), proximity ligation assay (PLA), and TrkA in these organelles. Our findings suggest
co-immunoprecipitation of full-length and that the reciprocal physiological and pathological
selected APP and TrkA fragments in transfected interplay between APP and TrkA might occur
HEK293 cells and rat primary septal cultures within these compartments where both proteins
(Canu et al. 2017a, b). Interestingly, we found form homodimers in the ER/Golgi before
that, as for APP/p75NTR interaction (Fombonne travelling to the cell surface.
et al. 2009), the APP juxta-membrane region Moreover, we found that in primary septal
encompassing β- and α-secretase cleavage sites neurons, NGF increases the number of TrkA/
is sufficient for its interaction with TrkA. On the APP complexes in every compartment, including
other side, TrkA binds APP using its own juxta- the ER and Golgi apparatus within 1 h of NGF
transmembrane, as well as its intracellular treatment. This effect is likely dependent on
domain. The latter TrkA domain is likely crucial NGF-mediated changes in APP phosphorylation
for modulating the phosphorylation state of APP, (Triaca et al. 2016; Matrone et al. 2011) since
known to affect in turn the interaction with TrkA TrkA and APP levels did not change under NGF
(Triaca et al. 2016). Although BiFC and PLA are stimulation (Canu et al. 2017a). Conversely,
able to detect the interaction among proteins in TrkA/APP complexes decrease in number with-
close proximity (distance range 7–15 nm), they out any apparent decrease of TrkA or APP and
do not exclude the involvement of a third partner before the loss of cell viability following pro-
in favouring the assembly of a multimolecular death-inducing stimuli like NGF deprivation, Aβ
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 153

peptide, staurosporine, and rapamycin treatments BACE, respectively; Lee et al. 2003) and to its
(Canu et al. 2017a). Such agents, via different subsequent trafficking and processing. Particu-
mechanisms, activate the amyloidogenic APP larly, the phosphorylation of APP at Thr668
processing [as observed in NGF-deprived pri- (T6889 in the YENPT668Y motif of the APP
mary hippocampal neurons (Matrone et al. C-tail has been described to control its intracellu-
2008)] by disentangling the physiological TrkA/ lar trafficking to abnormal Rab5 positive early
APP complex. Since APP/APP oligomerization endosomes, favouring Aβ fragments generation
promotes the amyloidogenic APP processing, by BACE1, thus contributing to
we hypothesize that TrkA/APP reduces the num- neurodegeneration (Barbagallo et al. 2010;
ber of APP homodimers in physiological Chang et al. 2006; Cataldo et al. 2004). In line,
conditions and under NGF stimulation and that, hyperphosphorylation of APP is observed in
during cell death (Canu et al. 2017a) and AD human AD brain (Lee et al. 2003; Schettini
neurodegeneration (Triaca et al. 2016), the disso- et al. 2010) and it is mediated by a number of
ciation of APP/TrkA complex allows the assem- ser/thr kinases, including JNKs 1-3, GSK3β,
bly of APP homodimers, which are more Cdk5, and ROCK1, all recognized key patholog-
susceptible to processing by β- and γ-secretase. ical players in Aβ-driven synaptic dysfunctions,
axonal swelling, and neuronal death (Hu et al.
2019). Accordingly, specifically targeting APP
10.4.3 NGF Regulates APP Trafficking phosphorylation was hypothesized as a promising
and Processing in BFCNs AD therapy (Lee et al. 2003).
Recently, it has been demonstrated that NGF
Regardless of the disease aetiology (sporadic or modulates APP phosphorylation at T668 by
familial), protein toxicity seems to be a main down-regulating the JNK kinase in BFCNs
hallmark of several neurodegenerative diseases in vitro, as well as in intact septo-hippocampal
(including AD, Parkinson’s disease, Huntington’s slices ex vivo, and in AD animal models. The
disease, frontotemporal dementia, and NGF-TrkA signalling pathway stimulates ShcC-
amyotrophic lateral sclerosis) and involves accu- mediated PI3K inhibition of JNK, in particular of
mulation, mislocalization, and/or aggregation of the 54kDa isoform JNK(p54), thus reducing
the disease-specific proteins. In neurodegenera- APPpT668 level in adult BFCNs (Triaca et al.
tive diseases, protein toxicity is often 2016). Activation of ShcC, the adult brain iso-
phosphorylation-dependent and leads to cellular form of the early TrkA adaptor Shc, represents a
defects such as transcriptional alteration, mito- key signalling event in NGF-driven
chondrial dysfunction, and an impaired protein/ neuroprotection of BFCNs and related cognition.
RNA quality control system. Each of these events Indeed, the ShcC genetic ablation is able per se to
critically contributes to the initiation and progres- increase JNK activation (Triaca et al. 2016, 2018)
sion of cognitive/motor deficits (Chung et al. by releasing the Shc-PI3K control over the initia-
2018). Protein phosphorylation of APP in AD, tion of the apoptotic route (Molton et al. 2003),
of tau in AD and PD, and of alpha-synuclein in thus specifically fostering β40 and βCTF genera-
Lewy bodies of PD and synucleinopathies, is tion in the hippocampus of shcC-KO mice, and
responsible for initiating and sustaining the affecting related cognitive performance in the
degenerative process in vulnerable BFCNs, visual object recognition test (vORT; Triaca
substantia nigra, and neurons, respectively et al. 2018). In line, the aberrant activation of
(Oueslati 2016; Tenreiro et al. 2014; Spillantini the alternative TrkA-PLCγ pathway has been
et al. 1998; Goedert et al. 1992). In the case of suggested to promote p75NTR-dependent
APP, phosphorylation at specific tyrosine (tyr) or amyloidogenesis and apoptosis in hippocampal
serine/threonine (ser/thr) residues at its intracellu- neurons (Matrone et al. 2008, 2009).
lar tail (C-terminal) is key to alternative cleavage Upon NGF-elicited autophosphorylation at
pathways by α or β secretases (ADAM10/17 and Tyr490 (Y490), TrkApY490 binds APP and
154 V. Triaca et al.

moves to the Golgi system, where APP exposure promising tools to restore the physiological
to BACE1 is irrelevant, possibly because of the NGF-TrkA system control over APP
prevailing TrkA competitive binding to APP and processing—the final goal being the restoration
of reduced physiological BACE1 level in the of normal cholinergic activity in the basal fore-
compartment. The reduced APP/BACE1 interac- brain of early AD patients and possibly MCI.
tion directly results in the down-regulation of
amyloidogenic APP cleavage, and βCTF and Aβ
levels (Triaca et al. 2016). 10.5 BFCN Insulin Resistance
Following T668 phosphorylation, APPpT668 as a Risk Factor for AD.
is not able to bind TrkA, as observed by Improvement by NGF
co-immunoprecipitation and colocalization stud-
ies. Released by TrkA, APP is now free to be The brain has been long time thought to be
trafficked to the BACE1-enriched endo-lyso- insulin-independent. Instead, recent work has
somal compartment, where more Aβ is produced demonstrated the expression of the insulin recep-
(Triaca et al. 2016). Similarly, the T668 phos- tor (IR), insulin receptor substrate (IRS), and
phorylation is known to cause the detachment of glucose transporters 1-8 (Glut1-8) in the BF, and
another well-known APP interactor, Fe65, lead- highlighted the key insulin’s action on brain glu-
ing to neuronal death (Chang et al. 2006). On the cose homeostasis, memory, and mood (reviewed
other hand, the neuroprotective interaction of in de la Monte 2012). In particular, insulin has
APP with TrkA is affected in the cortex and been reported to affect the expression of N-
hippocampus of AD patients and of the 3xTg- methyl-D-aspartate (NMDA) receptors (Skeberdis
AD mouse model, where the APPpT668 level is et al. 2001), modulate levels of the
augmented. Thus, in addition to favouring the neurotransmitters, like acetylcholine (Fishwick
amyloidogenic APP-APP interaction by competi- and Rylett 2015), and influence long-term poten-
tion (Canu et al. 2017a, b), the lack of APP-TrkA tiation (LTP; Zhao and Alkon 2001). In turn, the
binding directly promotes Aβ generation via the deficiency/lack of insulin signalling in the brain,
BACE1 cleavage of APP (Triaca et al. 2016). also known as neuronal insulin resistance, leads
Noteworthy, the role of TrkA as APP shuttling to changes in PI3K, Akt, and GSK3β activities, as
protein was hypothesized a decade ago (Heese well as Tau hyperphosphorylation, and Aβ pro-
et al. 2004). Further evidence of a role for the duction in Neuronal Insulin Receptor KO mice
NGF pathway in the modulation of APP phos- (NIRKO; Schubert et al. 2004).
phorylation came from the observation that Chronic exposure to an excess of insulin
transactivated TrkA induces Y682 phosphoryla- determines insulin resistance via feedback inhibi-
tion at the Y682NPTY motif of the APP C-tail tion of the IR by the Ser306 phosphorylation of
in vitro and subsequent binding of the APP intra- the insulin receptor substrate 1 (IRS1), the insulin
cellular domain (AID) with Shc (Tarr et al. 2002). signalling hub, hampering subsequent metabolic
Opposite, APP-TrkA interaction is lost in APPYG stimulation of the insulin pathway (De Felice
transgenic mice lacking the Y682 residue et al. 2014). Age-related hyperinsulinemia,
(Matrone et al. 2011), resulting in reduced APP which is know to induce a decrease in cell sensi-
degradation and cholinergic demise (La Rosa tivity to a subsequent glucose challange,
et al. 2015; Matrone et al. 2012). quadriplicates the risk of developing AD (Chow
Conclusively, these findings show the signal- et al., 2019).
ling and molecular substrates, and the intracellu- In line, systemic hyperinsulinemia, type 2 dia-
lar mechanisms related to the anti-amyloidogenic betes mellitus (T2D), obesity, and metabolic
NGF action in BFCNs of the septo-hippocampal conditions may trigger tissue ‘desensitization’ to
pathway in physio-pathological conditions. insulin, increasing the risk to develop AD (Kellar
Promoting APP-TrkA interaction and/or pro- and Craft 2020; Ferreira et al. 2018). Detection of
moting the ShcC-PI3K pathway are envisaged as cerebral Aβ with 18-F fluorbetan amyloid PET,
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 155

brain FDG-PET, and blood HOMA-IR index 3xTg-AD mice rescued insulin resistance in the
(homeostasis model assessment of insulin resis- medial septum at the pre-symptomatic stage, sev-
tance (HOMA-IR) has correlated central eral months before its detection in the neocortex
hypoglycaemia with peripheral insulin resistance and hippocampus (Sposato et al. 2019).
in AD patients, and associated central Since insulin resistance correlates with Aβ
dysmetabolism with cortical memory deficits burden, even before the AD onset of memory
(Willette et al. 2015). Consistently, a diabetes decline (Wang et al. 2019a, b), the ability of
diagnosis is a good predictor of cognitive decline NGF to reactivate the insulin pathway and control
and mortality over 10 years in the elderly popula- Aβ production and related cognition further
tion (Exalto et al. 2013; Tilvis et al. 2004). Now- pinpoints its use for insulin resistance, glucose
adays, AD is considered now as type 3 diabetes, dysmetabolism, and synaptic derangement in
because of the well-characterized status reminis- early AD.
cent of insulin resistance in the brain promoting
amyloidogenesis and neurodegeneration (Arnold
et al. 2018; De Felice et al. 2014). Central meta- 10.6 Interplay Between MicroRNAs,
bolic dysfunction is considered a well-established APP, and NGF in Alzheimer’s
feature of AD, and brain glucose hypometabolism Disease
is associated with AD dementia (Whitmer et al.
2007; Craft 2007) even before early AD manifes- MicroRNAs and AD MicroRNAs (miRNAs)
tation (Sperling et al. 2011). are evolutionarily conserved small non-coding
Accordingly, metabolic derangement and con- RNAs around 22 nucleotides in length. They
current early synaptic failure in BFCNs have been bind messenger RNA (mRNA) of target genes
reported to parallel memory deficits in MCI and mainly within the 30 untranslated region (UTR)
AD, and proposed as a good predictor of MCI and regulate, at the post-transcriptional level,
progression towards AD (Chen and Mobley gene expression through transcript destabilization
2019), although the underlying mechanisms are or translation inhibition. In humans and
still under debate. mammals, each miRNA can potentially modulate
To investigate these mechanisms, we devel- the expression of several target genes and in turn
oped an in vitro model of insulin resistance in regulate many cellular processes. Brain-
rat cholinergic neurons by means of expressed miRNAs play a role in neuronal devel-
hyperinsulinemic culture conditions. BFCNs opment, synaptic transmission, synaptic plastic-
were shown to develop insulin resistance as ity, and behaviour. Furthermore, specific
shown by reduced activation of both insulin miRNAs are highly enriched in several neuronal
receptor (IR) and insulin substrate receptor cell types (reviewed in Kiltschewskij and Cairns
1 (IRS1). Further, insulin-resistant neurons 2019). Alteration of miRNA expression profiles
expressed a higher level of serine phosphorylated has been described in AD human brain tissues.
IRS1, a well-known hallmark of insulin resis- Dysregulation of miRNAs is implicated in APP
tance, and showed reduced glucose transporter expression, Aβ production/clearance, and tau
2 (Glut2) translocation to the plasma membrane phosphorylation, as well as in insulin signalling,
resulting in lower glucose uptake. Also, neuronal and neuroinflammation; and it has been shown to
activity was repressed, as demonstrated by contribute to AD pathology (reviewed in Wang
decreased nuclear c-Fos expression (Sposato et al. 2019a; Improta-Caria et al. 2020).
et al. 2019). The interplay between APP and NGF
Significantly, NGF was shown to improve metabolisms could also be modulated by
insulin resistance by TrkA-driven tyrosine phos- miRNAs. MiRNAs regulating APP expression
phorylation and activation of IRS1, both in the and, in turn, amyloidogenesis (i.e. miR-16,
in vitro model and in 3xTg-AD mice. Interest- miR-20 family, miR-101, miR-153 miR-346,
ingly enough, nasal administration of NGF to etc) are down-regulated in AD mouse models
156 V. Triaca et al.

and AD human brain tissues (reviewed in as times and doses of NGF treatment, were used,
Amakiri et al. 2019). It has been shown that the the NGF-regulated miRNAs identified were often
modulation of these microRNAs has functional different in the various studies, and not always
effects on relevant hallmarks of AD pathology. associated with the same function. For example,
For example, the brain delivery of miR-16 in up-regulation of miR-221 in PC12 cells has been
mice reduces APP, BACE1, and tau in a region- found after 48 h of NGF treatment and it has been
specific manner (Parsi et al. 2015). Conversely, associated with cell survival and activation of
inhibition of miR-101 in mouse hippocampus not extracellular signal-regulated kinase (ERK)
only up-regulates AD-related genes but also neg- (Terasawa et al. 2009). In another study
atively affects learning and memory (Barbato up-regulation of miR-221 after 48 h and 8 days
et al. 2020). It might be relevant to investigate of NGF exposure was associated with cell differ-
whether the down-regulation of these miRNAs entiation (Hamada et al. 2012; Pandey et al.
leading to APP over-expression could also inter- 2015).
fere with NGF trafficking, as observed in BFCNs Montalban et al. (2014) profiled miRNA
of the DS mouse model (see Sect. 10.4.1). expression in PC12 cells not only upon NGF
treatment but also upon NGF deprivation of ter-
NGF-Modulated miRNAs and Their minally differentiated cells. The expression of a
Dysregulation in AD Few data are available small number of miRNAs was up-regulated by
on the miRNA-mediated regulation of NGF and NGF, and their induction was lost upon 24 h NGF
NGF receptors in the nervous system. Interest- deprivation. Among the regulated miRNAs, they
ingly, it has been shown that Let-7 miRNAs found the miR-212/132 cluster, already known to
induce peripheral nerve regeneration by be modulated by BDNF, and including miR-21,
regulating NGF expression (Li et al. 2015). Evi- miR-29c, miR-30c miR-93, miR-103, miR-207,
dence of NGF receptors modulation by miRNAs miR-691, and miR-709. Interestingly, a few of
has been observed in an in vivo mouse model of these miRNAs have also been associated with
brain focal ischemia, where TrkA and p75NTR AD. Among them, noteworthy is miR-132 that
receptors exert protective and deleterious effects, is involved in several CNS functions and it is one
respectively. After cerebral ischemia, a decrease of the most down-regulated miRNAs in the inter-
in the level of miR-592 induces an increase of mediate and late Braak stages of AD. Several
p75NTR that is associated also to an increase of in vitro and in vivo studies have shown that
pro-NGF and, in turn, cell death (Irmady et al. miR-132 depletion may aggravate AD pathology
2014). However, at this time there is no evidence acting on both Aβ and tau pathways, altering
that miRNA-mediated regulation of NGF and/or several mechanisms of cellular homeostasis,
TrkA/p75NTR genes may occur in AD vulnera- inducing dysfunction in synaptic plasticity and
ble neuronal circuitry. memory processes (reviewed by Salta and De
Instead, the effects of NGF on miRNAs Strooper 2017). To correlate miRNAs deregula-
expression have been more extensively tion to failure of NGF signalling is relevant to
investigated. Several expression profiles have highlight that the expression of miR-132 has been
been mainly performed in NGF-differentiated studied over the course of AD in relation to Aβ,
PC12 cells to identify NGF-regulated miRNAs tau, and ChAT expression in post-mortem NBM
(Terasawa et al. 2009; Hamada et al. 2012; human samples. It was shown that miR-132
Montalban et al. 2014; Pandey et al. 2015). expression levels were quite stable during the
These studies described miRNAs involved in early stage of AD and decreased significantly
NGF-related cell survival and differentiation, but during the late stages, showing a positive correla-
also neuronal degeneration due to NGF depriva- tion with ChAT (Zhu et al. 2016). Thus, it is
tion. As different experimental conditions, such tempting to speculate that loss of NGF supply,
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 157

in the course of AD, may contribute to miR-132 regulate axonal elongation without changing
down-regulation and lead to the vulnerability of miRNA expression. Indeed, Map1b and Calm1
NBM in the late phase of AD. mRNA were released from the Fragile X Mental
Another NGF-regulated miRNA in PC12 cells retardation protein (FMRP) and miR-181d-
potentially associated with AD is miR-21. repressing granules (Wang et al. 2015).
Montalban et al. (2014) showed that miR-21 is Overall, a deep investigation of the role of
highly up-regulated from 6 h to 10 days of NGF NGF on miRNAs regulation and their effects on
treatment and participates in NGF signalling. It specific targets in TrkA/p75NTR cells of the CNS
enhances MAPK and Akt signalling and induces is required. These studies in control and AD dis-
neuronal differentiation by increasing the neurite ease models may lead to the discovery of
length. Notably, over-expression of this miRNA NGF-regulated miRNAs potentially associated
protects differentiated-PC12 cells from degenera- with disease progression.
tion upon NGF deprivation. More recently, it has
been shown that the level of miR-21 in cortical
and hippocampal neurons decreases significantly 10.7 Conclusions
after Aβ exposure (Chatterjee et al. 2016). More-
over, miR-21 over-expression in the hippocam- The current view of AD aetiopathology posits
pus of a mouse model of AD rescued memory that Aβ and tau depositions display a well-defined
deficits and decreased both Aβ deposition and spatial and temporal pattern of appearance. One
proinflammatory factors (Cui et al. 2018). take-home lesson from unsuccessful clinical trials
Whether miR-21 down-regulation in AD is centred on Aβ is that once Aβ has been deposited
strictly associated with the loss of NGF signalling in vulnerable BF circuits, the application of A-
needs to be further investigated. β-targeting antibodies and/or BACE inhibitors
Altered expression of few miRNAs in AD fails to improve the neuronal deficits or to induce
might also be part of a protective/adaptative even partial recovery of cognition. The most plau-
response to the pathology. A recent study showed sible reasons for the failure of these therapeutical
that miR-200c, a miRNA gradually induced in approaches for AD include insufficiently early
NGF treated PC12 cells, is also up-regulated in intervention, but also inaccurate choice of treat-
a mouse model of AD- and Aβ-treated neuronal ment targets, improper drug dosages, loss of the
cells (Wu et al. 2016). The authors found that physiological function of Aβ peptides, and an
miR-200c increase supports the survival and dif- inadequate understanding of the complex patho-
ferentiation of cultured neurons by PTEN down- physiology of AD.
regulation. Moreover, they also suggested that Aβ-centred clinical trials have stimulated the
only TrkA positive cholinergic neurons are sensi- search for a more integrated view of AD patho-
tive to miR-200c-mediated neurite outgrowth. genesis, which has helped to improve AD diag-
Thus, the Aβ-miR-200c-PTEN pathway could nosis, development of new disease-specific
protect cholinergic neurons at the early stage of tracking markers, and prediction of disease pro-
AD. Besides changes of miRNA expression, gression. In line with this, AD aetiopathology has
NGF also induces PC12 neuronal differentiation been re-conceptualized as a neurodegenerative
by a reduction of let-7a miRNA activity. In par- continuum, with three different phases reflecting
ticular, the uncoupling of miRNA from Ago2 the progressive cognitive decline (Jack et al.
protein (Patranabis and Bhattacharyya 2016), the 2018). In addition to considering the clinically
main component of the RNA-induced silencing defined syndromal phases (MCI, early AD, severe
complex implicated in miRNA-mediated post- AD), AD staging takes into account the unique
transcriptional regulation of mRNA targets, has combination of biological features within the A
been observed. Likewise, it has been shown that (Aβ) - T (tau) - N (Neurodegeneration) system,
NGF signalling in dorsal root ganglia may where N is a measure of FDG-PET
158 V. Triaca et al.

Fig. 10.1 A schematic model illustrating the control of trafficking of APP to the Golgi compartment and to the
APP metabolism by the NGF/TrkA signalling, the early plasma-membrane, and (3) preferential physiological
insulin pathway and miR-132 level in healthy (up) and AD cleavage by ADAM10/17, and sAPPα level increase.
(bottom) BFCNs. Healthy brain: The NGF pathway and Upon NGF stimulation, pTrkAY490 is also able to induce
the anti-amyloidogenic route are shown. Upon binding to the insulin/IR pathway by tyrosine transphosphorylation
its ligand NGF, TrkA autophosphorylates at the Y490 of the IRS1, promoting glucose uptake mainly through
residue, thus acting as a docking site for ShcC. Once Glut4 and sustaining cholinergic neurons activity, and
bound and activated, ShcC hinders the JNK-mediated metabolism. In healthy brain, NGF signalling in choliner-
phosphorylation of APP at the T668 site. Since TrkA gic neurons may modulate the expression of miR-132
does not bind APP if phosphorylated at T668, the reduc- implicated in the homeostasis of Aβ and tau phosphoryla-
tion of NGF-mediated APPpT668 promotes: (1) the tion. Tau, not within the focus of this review, is not
APP-TrkA binding, (2) the subsequent TrkA-mediated hyperphosphorylated, and is in equilibrium between
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 159

hypometabolism and MRI-detected atrophy International (CC BY 4.0). A special thanks to


(Ebenau et al. 2020). Dr. V. Sposato for her contribution to the
experimental work.
As reported in AD Drug Development Pipe- The authors apologize for the scientific publications not
line 2020, several new drugs are in clinical trials acknowledged in this manuscript because of space
(phases 1–3) for the treatment of AD. Among restrictions.
these are drugs for improving synaptic plasticity/ The authors sincerely apologize to all those colleagues
whose important work was not cited in this paper owing
neuroprotection (24%) and drugs targeting to space limitations.
metabolism (12%), such as insulin sensitizers.
The neurotrophin NGF, with its clear Conflict of Interest Statement The authors declare that
neuroprotective activity over BFCNs, has been they have no actual or potential conflicts of interest and
demonstrated to exert double-edged effects. It that these data are not published elsewhere. In addition all
acts as an Aβ-modulating molecule under authors approve the study described in this report.
physio-pathological conditions (Triaca et al.
2016; Matrone et al. 2008; Canu et al. References
2017a, b), but also as an insulin sensitizer by
activating IRS1, a central metabolic hub for neu- Allen SJ, Watson JJ, Dawbarn D (2011) The neurotrophins
ronal metabolism and synaptic activity in the and their role in Alzheimer’s disease. Curr
Neuropharmacol 9:559–573
normal and AD brain (Sposato et al. 2019). Amakiri N, Kubosumi A, Tran J, Reddy PH (2019) Amy-
In light of recent findings, effective AD treat- loid beta and microRNAs in Alzheimer’s disease.
ment may require a combination of treatments Front Neurosci 13:430
rather than a monotherapy. Target-specific non-- Appel SH (1981) A unifying hypothesis for the cause of
amyotrophic lateral sclerosis, parkinsonism, and
invasive delivery of a combination of classical Alzheimer disease. Ann Neurol 10:499–505
APP/tau-targeted molecules, together with novel Arnold SE, Arvanitakis Z, Macauley-Rambach SL et al
compounds that sustain neuronal trophism, (2018) Brain insulin resistance in type 2 diabetes and
recover neuronal metabolism and improve synap- Alzheimer disease: concepts and conundrums. Nat Rev
Neurol 14:168–181
tic plasticity and are likely candidates as thera- Arriagada PV, Growdon JH, Hedley-Whyte ET, Hyman
peutic interventions to AD dementia. Useful tools BT (1992) Neurofibrillary tangles but not senile
may come from NGF-based and/or miRNA- plaques parallel duration and severity of Alzheimer’s
based approaches (Fig. 10.1). disease. Neurology 42(3 Pt 1):631–639
Baker-Nigh A, Vahedi S, Davis EG, Weintraub S, Bigio
EH, Klein WL, Geula C (2015) Neuronal amyloid-β
Acknowledgements This work was supported by accumulation within cholinergic basal forebrain in age-
Finanziamento delle Attività Base di Ricerca FFABR to ing and Alzheimer’s disease. Brain 138:1722–1737
NC. Graphic elaboration of the illustration by VT was Ballinger EC, Ananth M, Talmage DA, Role LW (2016)
partially realized with vectorial images from ‘FreeVector. Basal forebrain cholinergic circuits and signaling in
com’, under Creative Common Attribution 4.0 cognition and cognitive decline. Neuron
91:1199–1218

ä
Fig. 10.1 (continued) binding and detaching from CTFβ and Aβ. In AD, miR-132 expression is reduced and
microtubles. Alzheimer’s brain: The impairment of contributes to the increased level of Aβ and tau phosphor-
NGF/TrkA signalling and the amyloidogenic route. ylation in AD. Abnormal tau phosphorylation causes its
Reduced availability of mature NGF, and impaired expres- aggregation. Concomitantly, ser/thr kinases, like JNK,
sion levels or activation of TrkA affects APP metabolism phosphorylate IRS1 at key serine residues, hampering its
in BFCN. In fact, disturbances in the NGF/TrkA-ShcC further activation and leading to neuronal insulin resis-
signalling pathway and the alternative assembly of the tance, a condition characterized by insulin insensitivity,
pro-apototic p75NTR/sortilin complex grant the activation reduced glucose uptake, and deficits in synaptic activity
of JNK. This latter results in augmented APPpT668 levels and memory. Grey and white syringes indicate the classi-
and disentanglement of the APP/TrkA complex. APPpT668 cal and suggested AD targets, respectively, for a combined
is then trafficked to the endosomal-lysosomal therapeutical approach to this devastating pathology
compartments where it is cleaved by BACE1, to generate
160 V. Triaca et al.

Barbagallo AP, Weldon R, Tamayev R, Zhou D, Caporaso GL, Takei K, Gandy SE, Matteoli M,
Giliberto L, Foreman O, D’Adamio L (2010) Tyr Mundigl O, Greengard P, De Camilli P (1994) Mor-
(682) in the intracellular domain of APP regulates phologic and biochemical analysis of the intracellular
amyloidogenic APP processing in vivo. PLoS One 5 trafficking of the Alzheimer beta/A4 amyloid precursor
(11):e15503 protein. Neurosci 14:3122–3138
Barbato C, Giacovazzo G, Albiero F, Scardigli R, Capsoni S, Ugolini G, Comparini A, Ruberti F, Berardi N,
Scopa C, Ciotti MT, Strimpakos G, Coccurello R, Cattaneo A (2000) Alzheimer-like neurodegeneration
Ruberti F (2020) Cognitive decline and modulation of in aged antinerve growth factor transgenic mice. Proc
Alzheimer’s disease-related genes after inhibition of Natl Acad Sci USA 97:6826–6831
microRNA-101 in mouse hippocampal neurons. Mol Cataldo AM, Petanceska S, Terio NB, Peterhoff CM,
Neurobiol 57(7):3183–3194 Durham R, Mercken M, Mehta PD, Buxbaum J,
Batarseh YS, Duong QV, Mousa YM, Al Rihani SB, Haroutunian V, Nixon RA (2004) Abeta localization
Elfakhri K, Kaddoumi A (2016) Amyloid-beta and in abnormal endosomes: association with earliest
astrocytes interplay in amyloid-beta related disorders. Abeta elevations in AD and Down syndrome.
Int J Mol Sci 17:338 Neurobiol Aging 25:1263–1272
Bejanin A, Schonhaut DR, La Joie R, Krame JH, Baker Chang KA, Kim HS, Ha TY, Ha JW, Shin KY, Jeong YH,
SL, Sosa N et al (2017) Tau pathology and Lee JP, Park CH, Kim S, Baik TK, Suh YH (2006)
neurodegeneration contribute to cognitive impairment Phosphorylation of amyloid precursor protein (APP) at
in Alzheimer’s disease. Brain 140:3286–3300 Thr668 regulates the nuclear translocation of the APP
Bierer LM, Haroutunian V, Gabriel S, Knott PJ, Carlin LS, intracellular domain and induces neurodegeneration.
Purohit DP, Perl DP, Schmeidler J, Kanof P, Davis KL Mol Cell Biol 26:4327–4338
(1995) Neurochemical correlates of dementia severity Chatterjee N, Sanphui P, Kemeny S, Greene LA, Biswas
in Alzheimer’s disease: relative importance of the cho- SC (2016) Role and regulation of Cdc25A phosphatase
linergic deficits. J Neurochem 64:749–760 in neuron death induced by NGF deprivation or
Bilousova T, Miller CA, Poon WW, Vinters HV, β-amyloid. Cell Death Discov 2:16083
Corrada M, Kawas C, Hayden EY, Teplow DB, Chen X-Q, Mobley WC (2019) Exploring the pathogene-
Glabe C, Albay R 3rd, Cole GM, Teng E, Gylys KH sis of Alzheimer disease in basal forebrain cholinergic
(2016) Synaptic amyloid-beta oligomers precede neurons: converging insights from alternative
p-Tau and differentiate high pathology control cases. hypotheses. Front Neurosci 13:446
Am J Pathol 186:185–198 Chen Q, Sawa M, Mobley WC (2018) Dysregulation of
Botté A, Lainé J, Xicota L, Heiligenstein X, Fontaine G, neurotrophin signaling in the pathogenesis of
Kasri A, Rivals I, Goh P, Faklaris O, Cossec JC, Alzheimer disease and of Alzheimer disease in Down
Morel E, Rebillat AS, Nizetic D, Raposo G, Potier syndrome. Free Radic Biol Med 114:52–61
MC (2020) Ultrastructural and dynamic studies of the Chow H-M, Shi M, Cheng A, Gao Y, Chen G, Song X, So
endosomal compartment in Down syndrome. Acta RWL, Zhang J, Herrup K (2019) Age-related
Neuropathol Commun 8(1):89 hyperinsulinemia leads to insulin resistance in neurons
Braak H, Braak E (1991) Neuropathological stageing of and cell-cycle-induced senescence. Nat Neurosci
Alzheimer-related changes. Acta Neuropathol 22:1806–1819
82:239–259 Chung CG, Lee H, Lee SB (2018) Mechanisms of protein
Calissano P, Amadoro G, Matrone C, Ciaffrè S, toxicity in neurodegenerative diseases. Cell Mol Life
Corsetti V, Marolda R, Ciotti MT, DiLuzio A, Sci 75:3159–3180
Severini C, Mercanti D, Provenzano C, Canu N Cline EN, Bicca MA, Viola KL, Klein WL (2018) The
(2010) Does the term “NGF” actually means anti- amyloid-β oligomer hypothesis: beginning of the third
amyloidogenic ? The case of NGF. Cell Death Diff decade. J Alzheimer Dis 64(S1):S567–S610
17(7):1126–1133 Colom LV, García-Hernández A, Castañeda MT, Perez-
Cantero JL, Zaborszky L, Atienza M (2016) Volume loss Cordova MG, Garrido-Sanabria ER (2006) Septo-
of the nucleus basalis of meynert is associated with hippocampal networks in chronically epileptic rats:
atrophy of innervated regions in mild cognitive potential antiepileptic effects of theta rhythm genera-
impairment. Cereb Cortex 27(8):3881–3889 tion. J Neurophysiol 95:3645–3653
Canu N, Pagano I, LaRosa LR, Pellegrino M, Ciotti MT, Craft S (2007) Insulin resistance and Alzheimer’s disease
Mercanti D, Moretti F, Sposato V, Triaca V, Petrella C, pathogenesis: potential mechanisms and implications
Maruyama IN, Levi A, Calissano P (2017a) Associa- for treatment. Curr Alzheimer Res 4(2):147–152
tion of TrkA and APP is promoted by NGF and Cuello AC, Bruno MA, Bell KFS (2007) NGF-cholinergic
reduced by cell death-promoting agents. Front Mol dependency in brain aging, MCI and Alzheimer’s dis-
Neurosci 10:15 ease. Curr Alzheimer Res 4:351–358
Canu N, Amadoro G, Triaca V, Latina V, Sposato V, Cui GH, Wu J, Mou FF, Xie WH, Wang FB, Wang QL,
Corsetti V, Severini C, Ciotti MT, Calissano P Fang J, Xu YW, Dong YR, Liu JR, Guo HD (2018)
(2017b) The intersection of NGF/TrkA signaling and Exosomes derived from hypoxia-preconditioned mes-
amyloid precursor protein processing in Alzheimer’s enchymal stromal cells ameliorate cognitive decline by
disease neuropathology. Int J Mol Sci 18(6):1319 rescuing synaptic dysfunction and regulating
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 161

inflammatory responses in APP/PS1 mice. FASEB J Giannakopoulos P, Herrmann FR, Bussière T, Bouras C,
32(2):654–668 Kövari E, Perl DP, Morrison JH, Gold G, Hof PR
Davies P, Maloney AJ (1976) Selective loss of central (2003) Tangle and neuron numbers, but not amyloid
cholinergic neurons in Alzheimer’s disease. Lancet load, predict cognitive status in Alzheimer’s disease.
2:1403 Neurology 60:1495–1500
De Felice FG (2013) Alzheimer’s disease and insulin Giuffrida ML, Caraci F, Pignataro B, Cataldo S, De
resistance: translating basic science into clinical Bona P, Bruno V, Molinaro G, Pappalardo G,
applications. J Clin Invest 123:531–539 Messina A, Palmigiano A, Garozzo D, Nicoletti F,
De Felice FG, Lourenco MV, Ferreira ST (2014) How Rizzarelli E, Copani A (2009) Beta-amyloid
does brain insulin resistance develop in Alzheimer’s monomers are neuroprotective. J Neurosci
disease? Alzheimer Dement 10:S26–S32 29:10582–10587
de la Monte SM (2012) Brain insulin resistance and defi- Goedert M, Spillantini MG, Cairns NJ, Crowther RA
ciency as therapeutic targets in Alzheimers disease. (1992) Tau proteins of alzheimer paired helical
Curr Alzheimer Res 9(1):35–66 filaments: abnormal phosphorylation of all six brain
De Strooper B, Karran E (2016) The cellular phase of isoforms. Neuron 8:159–168
Alzheimer’s disease. Cell 164:603–615 Grothe M, Zaborszky L, Atienza M, Gil-Neciga E,
Dorszewska J, Prendecki M, Oczkowska A, Dezor M, Rodriguez-Romero R, Teipel SJ, Amunts K, Suarez-
Kozubski W (2016) Molecular basis of familial and Gonzalez A, Cantero JL (2010) Reduction of basal
sporadic Alzheimer’s disease. Curr Alzheimer Res forebrain cholinergic system parallels cognitive
13:952–963 impairment in patients at high risk of developing
Ebenau JL, Timmers T, Wesselman LMP et al (2020) Alzheimer’s disease. Cereb Cortex 20:1685–1695
ATN classification and clinical progression in subjec- Guo JP, Arai T, Miklossy J, McGeer PL (2006) Aβ and tau
tive cognitive decline. Neurology 95:e46–e58 form soluble complexes that may promote self aggre-
Eggert S, Thomas C, Kins S, Hermey G (2018) Trafficking gation of both into the insoluble forms observed in
in Alzheimer’s disease: modulation of APP transport Alzheimer’s disease. Proc Natl Acad Sci USA
and processing by the transmembrane proteins LRP1, 103:1953–1958
SorLA, SorCS1c, Sortilin, and Calsyntenin. Mol Gustafsen C, Glerup S, Pallesen LT, Olsen D, Andersen
Neurobiol 55(7):5809–5829 OM, Nykjær A, Madsen P, Petersen CM (2013)
El-Shimy IA, Heikal OA, Hamdi N (2015) Minocycline Sortilin and SorLA display distinct roles in processing
attenuates Abeta oligomers-induced pro-inflammatory and trafficking of amyloid precursor protein. J
phenotype in primary microglia while enhancing Abeta Neurosci 33:64–71
fibrils phagocytosis. Neurosci Lett 609:36–41 Haass C, Kaether C, Thinakaran G, Sisodia S (2012)
Exalto LG, Biessels GJ, Karter AJ, Huang ES, Katon WJ, Trafficking and proteolytic processing of APP. Cold
Minkoff JR, Whitmer RA (2013) Risk score for pre- Spring Harb Perspect Med 2(5):a006270
diction of 10 year dementia risk in individuals with Hamada N, Fujita Y, Kojima T, Kitamoto A, Akao Y,
type 2 diabetes: a cohort study. Lancet Diabetes Nozawa Y, Ito M (2012) MicroRNA expression
Endocrinol 1:183–190 profiling of NGF-treated PC12 cells revealed a critical
Ferreira LSS, Fernandes CS, Vieira MNN, de Felice FG role for miR-221 in neuronal differentiation.
(2018) Insulin resistance in Alzheimer’s disease. Front Neurochem Int 60:743–750
Neurosci 12:830 Hangya B, Ranade SP, Lorenc M, Kepecs A (2015) Cen-
Fisher A (2012) Cholinergic modulation of amyloid pre- tral cholinergic neurons are rapidly recruited by rein-
cursor protein processing with emphasis on M1 mus- forcement feedback. Cell 162:1155–1168
carinic receptor: perspectives and challenges in Hardy JA, Higgings GA (1992) Alzheimer’s disease: the
treatment of Alzheimer’s disease. J Neurochem 120 amyloid cascade hypothesis. Science 256:184–185
(Suppl 1):22–33 Heese K, Inoue N, Sawada T (2004) APP, NGF & the
Fishwick KJ, Rylett RJ (2015) Insulin regulates the activ- ‘Sunday-driver’ in a Trolley on the Road. Resto Neurol
ity of the high-affinity choline transporter CHT. PLoS Neurosci 22(2):131–136
One 10:e0132934 Hu YB, Ren RJ, Zhang YF, Huang Y, Cui HL, Ma C, Qiu
Fombonne J, Rabizadeh S, Banwait S, Mehlen P, WY, Wang H, Cui PJ, Chen HZ, Wang G (2019)
Bredesen DE (2009) Selective vulnerability in Rho-associated coiled-coil kinase 1 activation
Alzheimer’s disease: amyloid precursor protein and mediates amyloid precursor protein site-specific
p75(NTR) interaction. Ann Neurol 65(3):294–303 Ser655 phosphorylation and triggers amyloid pathol-
Frost B, Jacks RL, Diamond MI (2009) Propagation of tau ogy. Aging Cell 18(5):e13001
misfolding from the outside to the inside of a cell. J Improta-Caria AC, Nonaka CKV, Cavalcante BRR, De
Biol Chem 284:12845–12852 Sousa RAL, Aras Júnior R, Souza BSF (2020) Modu-
Geula C, Nagykery N, Nicholas A, Wu C-K (2008) Cho- lation of microRNAs as a potential molecular mecha-
linergic neuronal and axonal abnormalities are present nism involved in the beneficial actions of physical
early in aging and in Alzheimer disease. J Neuropathol exercise in Alzheimer disease. Int J Mol Sci 21
Exp Neurol 67:309–318 (14):4977
162 V. Triaca et al.

Ioannou MS, Fahnestock M (2017) ProNGF, but Not amyloid precursor protein promotes endo-lysosomal
NGF, switches from neurotrophic to apoptotic activity dysfunction by disrupting APP-SorLA interaction.
in response to reductions in TrkA receptor levels. Int J Front Cell Neurosci 9:109
Mol Sci 18:599 Lee MS, Kao SC, Lemere CA, Xia W, Tseng HC, Zhou Y,
Iqbal K, Liu F, Gong CX (2016) Tau and neurodegenera- Neve R, Ahlijanian MK, Tsai LH (2003) APP
tive disease: the story so far. Nat Rev Neurol 12 processing is regulated by cytoplasmic phosphoryla-
(1):15–27 tion. J Cell Biol 163:83–95
Irmady K, Jackman KA, Padow VA, Shahani N, Martin Leung LS, Ma J (2018) Medial septum modulates hippo-
LA, Cerchietti L, Unsicker K, Iadecola C, Hempstead campal gamma activity and prepulse inhibition in an
BL (2014) Mir-592 regulates the induction and cell N-methyl-d-aspartate receptor antagonist model of
death-promoting activity of p75NTR in neuronal ische- schizophrenia. Schizoph Res 198:36–44
mic injury. J Neurosci 34:3419–3428 Li S, Wang X, Gu Y, Chen C, Wang Y, Liu J, Hu W,
Jack CR Jr, Bennett DA, Blennow K, Carrillo MC, Yu B, Wang Y, Ding F, Liu Y, Gu X (2015) Let-7
Dunn B, Haeberlein SB, Holtzman DM, Jagust W, microRNAs regenerate peripheral nerve regeneration
Jessen F, Karlawish J, Liu E, Molinuevo JL, by targeting nerve growth factor. Mol Ther 23
Montine T, Phelps C, Rankin KP, Rowe CC, (3):423–433
Scheltens P, Siemers E, Snyder HM, Sperling R, Liu Y, Zhang YW, Wang X, Zhang H, You X, Liao FF,
Contributors (2018) NIA-AA research framework: Xu HJ (2009) Intracellular trafficking of presenilin 1 is
toward a biological definition of Alzheimer’s disease. regulated by beta-amyloid precursor protein and phos-
Alzheimerd Dement 14(4):535–562 pholipase D1. Biol Chem 284:12145–12152
Jeong DU, Lee JE, Lee SE, Chang WS, Kim SJ, Chang JW Mankin EA, Fried I (2020) Modulation of human memory
(2014) Improvements in memory after medial septum by deep brain stimulation of the entorhinal-
stimulation are associated with changes in hippocam- hippocampal circuitry. Neuron 106(2):218–235
pal cholinergic activity and neurogenesis. BioMed Res Matrone C, Ciotti MT, Mercanti D, Marolda R, Calissano
Int 2014:1–10 P (2008) NGF and BDNF signaling control
Kamal A, Almenar-Queralt A, LeBlanc JF, Roberts EA, amyloidogenic route and a production in hippocampal
Goldstein LS (2001) Kinesin-mediated axonal trans- neurons. Proc Natl Acad Sci USA 105:13139–13144
port of a membrane compartment containing beta- Matrone C, Marolda R, Ciafrè S, Ciotti MT, Mercanti D,
secretase and presenilin-1 requires APP. Nature 414 Calissano P (2009) Tyrosine kinase nerve growth fac-
(6864):643–648 tor receptor switches from prosurvival to proapoptotic
Kashyap G, Bapat D, Das D, Gowaikar R, Amritkar RE, activity via Abeta-mediated phosphorylation. Proc
Rangarajan G, Ravindranath V, Ambika G (2019) Natl Acad Sci U S A 106(27):11358–11363
Synapse loss and progress of Alzheimer’s disease—a Matrone C, Barbagallo AP, La Rosa LR, Florenzano F,
network model. Sci Rep 9:6555 Ciotti MT, Mercanti D, Chao MV, Calissano P,
Kellar D, Craft S (2020) Brain insulin resistance in D’Adamio L (2011) APP is phosphorylated by TrkA
Alzheimer’s disease and related disorders: mechanisms and regulates NGF/TrkA signaling. J Neurosci
and therapeutic approaches. Lancet Neurol 31:11756–11761
19:758–766 Matrone C, Luvisetto S, La Rosa LR, Tamayev R,
Kerbler GM, Fripp J, Rowe CC, Villemagne VL, Pignataro A, Canu N, Yang L, Barbagallo AP,
Salvado O, Rose S, Coulson EJ (2015) Basal forebrain Biundo F, Lombino F, Zheng H, Ammassari-Teule M,
atrophy correlates with amyloid β burden in D’Adamio L (2012) Tyr682 in the Aβ-precursor pro-
Alzheimer’s disease. NeuroImage Clin 7:105–113 tein intracellular domain regulates synaptic connectiv-
Kilimann I, Grothe M, Heinsen H, Alho EJ, Grinberg L, ity, cholinergic function, and cognitive performance.
Amaro E Jr, Dos Santos GA, da Silva RE, Mitchell AJ, Aging Cell 6:1084–1093
Frisoni GB, Bokde AL, Fellgiebel A, Filippi M, Molton SA, Todd DE, Cook SJ (2003) Selective activation
Hampel H, Klöppel S, Teipel SJ (2014) Subregional of the c-Jun N-terminal kinase (JNK) pathway fails to
basal forebrain atrophy in Alzheimer’s disease: a mul- elicit Bax activation or apoptosis unless the phosphoi-
ticenter study. J Alzheimers Dis 40:687–700 nositide 30 ‐kinase (PI3K) pathway is inhibited. Onco-
Kiltschewskij D, Cairns MJ (2019) Temporospatial guid- gene 22:4690–4701
ance of activity-dependent gene expression by Montalban E, Mattugini N, Ciarapica R, Provenzano C,
microRNA: mechanisms and functional implications Savino M, Scagnoli F, Prosperini G, Carissimi C,
for neural plasticity. Nucleic Acids Res 47:533–545 Fulci V, Matrone C, Calissano P, Nasi S (2014)
Kitchigina VF (2018) Alterations of coherent theta and MiR-21 is an NGF-modulated microRNA that
gamma network oscillations as an early biomarker of supports NGF signaling and regulates neuronal degen-
temporal lobe epilepsy and Alzheimer’s disease. Front eration in PC12 cells. Neuromolecular Med
Integr Neurosci 12:36 16:415–430
Koo EH, Squazzo SL (1994) Evidence that production and Mufson EJ, Counts SE, Ginsberg SD, Mahady L, Perez
release of amyloid beta-protein involves the endocytic SE, Massa SM, Longo FM, Ikonomovic MD (2019)
pathway. J Biol Chem 269:17386–17389 Nerve growth factor pathobiology during the progres-
La Rosa LR, Perrone L, Nielsen MS, Calissano P, sion of Alzheimer’s disease. Front Neurosci 13:533
Andersen OM, Matrone C (2015) Y682G mutation of
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 163

Nielsen MS, Madsen P, Christensen EI, Nykjær A, Salta E, De Strooper B (2017) microRNA-132: a key
Gliemann J, Kasper D, Pohlmann R, Petersen CM noncoding RNA operating in the cellular phase of
(2001) The sortilin cytoplasmic tail conveys Golgi- Alzheimer’s disease. FASEB J 31:424–433
endosome transport and binds the VHS domain of the Sassin I, Schultz C, Thal DR, Rüb U, Arai K, Braak E,
GGA2 sorting protein. EMBO J 20:2180–2190 Braak H (2000) Evolution of Alzheimer’s disease-
Nielsen HM, Mulder SD, Belien JA, Musters RJ, related cytoskeletal changes in the basal nucleus of
Eikelenboom P, Veerhuis R (2010) Astrocytic A beta Meynert. Acta Neuropathol 100:259–269
1-42 uptake is determined by A beta-aggregation state Schettini G, Govoni S, Racchi M, Rodriguez G (2010)
and the presence of amyloid-associated proteins. Glia Phosphorylation of APP-CTF-AICD domains and
58:1235–1246 interaction with adaptor proteins: signal transduction
Nykjaer A, Lee R, Teng KK, Jansen P, Madsen P, Nielsen and/or transcriptional role—relevance for Alzheimer
MS, Jacobsen C, Kliemannel M, Schwarz E, Willnow pathology. J Neurochem 115:1299–1308
TE, Hempstead BL, Petersen CM (2004) Sortilin is Schliebs R, Arendt T (2011) The cholinergic system in
essential for proNGF-induced neuronal cell death. aging and neuronal degeneration. Behav Brain Res
Nature 427(6977):843–848 221:555–563
Oueslati AJ (2016) Implication of alpha-synuclein phos- Schmitz TW, Mur M, Aghourian M, Bedard M-A, Spreng
phorylation at S129 in synucleinopathies: what have RN, Alzheimer’s Disease Neuroimaging Initiative
we learned in the last decade? Parkinsons Dis 26 (2018) Longitudinal Alzheimer’s degeneration reflects
(1):39–51 the spatial topography of cholinergic basal forebrain
Pandey A, Singh P, Jauhari A, Singh T, Khan F, Pant AB, projections. Cell Rep 24:38–46
Parmar D, Yadav S (2015) Critical role of the miR-200 Schubert M, Gautam D, Surjo D et al (2004) Role for
family in regulating differentiation and proliferation of neuronal insulin resistance in neurodegenerative
neurons. J Neurochem 133:640–652 diseases. Proc Natl Acad Sci U S A 101:3100–3105
Parsi S, Smith PY, Goupil C, Dorval V, Hébert SS (2015) Selkoe DJ (2011) Resolving controversies on the path to
Preclinical evaluation of miR-15/107 family members Alzheimer’s therapeutics. Nat Med 17:1060–1065
as multifactorial drug targets for Alzheimer’s disease. Selkoe DJ, Yamazaki T, Citron M, Podlisny MB, Koo EH,
Mol Ther Nucleic Acids 4(10):e256 Teplow DB, Haass C (1996) The role of APP
Patel J, Fujisawa S, Berényi A, Royer S, Buzsáki G (2012) processing and trafficking pathways in the formation
Traveling theta waves along the entire septotemporal of amyloid beta-protein. Ann N Y Acad Sci 777:57–64
axis of the hippocampus. Neuron 75:410–417 Serrano-Pozo A, Frosch MP, Masliah E, Hyman BT
Patranabis S, Bhattacharyya SN (2016) Phosphorylation of (2011) Neuropathological alterations in Alzheimer dis-
Ago2 and subsequent inactivation of let-7a ease. Cold Spring Harb Perspect Med 1:a006189
RNP-specific microRNAs control differentiation of Skeberdis VA, Lan J, Zheng X, Zukin RS, Bennett MVL
mammalian sympathetic neurons. Mol Cell Biol (2001) Insulin promotes rapid delivery of N-methyl-D-
36:1260–1271 aspartate receptors to the cell surface by exocytosis.
Pearson RCA, Gatter KC, Powell TPS (1983) The cortical Proc Natl Acad Sci U S A 98:3561–3566
relationships of certain basal ganglia and the choliner- Sperling RA, Aisen PS, Beckett LA et al (2011) Toward
gic basal forebrain nuclei. Brain Res 261:327–330 defining the preclinical stages of Alzheimer’s disease:
Petersen PC, Buzsáki G (2020) Cooling of medial septum recommendations from the National Institute on
reveals theta phase lag coordination of hippocampal Aging-Alzheimer’s Association workgroups on diag-
cell assemblies. Neuron 107:731–744.e3 nostic guidelines for Alzheimer’s disease. Alzheimers
Puzzo D, Lee L, Palmeri A, Calabrese G, Arancio O Dement 7:280–292
(2014) Behavioral assays with mouse models of Spillantini MG, Crowther RA, Jakes R, Hasegawa M,
Alzheimer’s disease: practical considerations and Goedert M (1998) α-Synuclein in filamentous
guidelines. Biochem Pharmacol 88:450–467 inclusions of Lewy bodies from Parkinson’s disease
Roussarie JP, Yao V, Rodriguez-Rodriguez P, and dementia with Lewy bodies. Proc Natl Acad Sci
Oughtred R, Rust J, Plautz Z, Kasturia S, USA 95:6469–6473
Albornoz C, Wang W, Schmidt EF, Dannenfelser R, Spires-Jones TL, Hyman BT (2014) The intersection of
Tadych A, Brichta L, Barnea-Cramer A, Heintz N, Hof amyloid beta and tau at synapses in Alzheimer’s dis-
PR, Heiman M, Dolinski K, Flajolet M, Troyanskaya ease. Neuron 82(4):756–771
OG, Greengard P (2020) Selective neuronal vulnera- Sposato V, Canu N, Fico E, Fusco S, Bolasco G, Ciotti
bility in Alzheimer’s disease: a network-based analy- MT, Spinelli M, Mercanti D, Grassi C, Triaca V,
sis. Neuron 107:1–15 Calissano P (2019) The medial septum is insulin resis-
Salehi A, Delcroix JD, Belichenko PV, Zhan K, Wu C, tant in the AD presymptomatic phase: rescue by nerve
Valletta JS, Takimoto-Kimura R, Kleschevnikov AM, growth factor-driven IRS1 activation. Mol Neurobiol
Sambamurti K, Chung PP, Xia W, Villar A, Campbell 56:535–552
WA, Kulnane LS, Nixon RA, Lamb BT, Epstein CJ, Stypulkowski PH, Stanslaski SR, Giftakis JE (2017) Mod-
Stokin GB, Goldstein LS, Mobley WC (2006) ulation of hippocampal activity with fornix deep brain
Increased App expression in a mouse model of stimulation. Brain Stimul 10:1125–1132
Down’s syndrome disrupts NGF transport and causes Szutowicz A, Bielarczyk H, Jankowska-Kulawy A,
cholinergic neuron degeneration. Neuron 51:29–42 Pawełczyk T, Ronowska A (2013) Acetyl-CoA the
164 V. Triaca et al.

key factor for survival or death of cholinergic neurons current scenario in Alzheimer’s disease therapy. J
in course of neurodegenerative diseases. Neurochem Pharm Pharmacol 70(8):985–993
Res 38:1523–1542 Vogels OJM, Broere CAJ, ter Laak HJ, ten Donkelaar HJ,
Takahashi M Kametani F, Nonaka T, Akiyama H, Nieuwenhuys R, Schulte BPM (1990) Cell loss and
Hisanaga S, Hasegawa M (2015) Extracellular associ- shrinkage in the nucleus basalis Meynert complex in
ation of APP and tau fibrils induces intracellular aggre- Alzheimer’s disease. Neurobiol Aging 11:3–13
gate formation of tau. Acta Neuropathol 129 Wang B, Yang L, Wang Z, Zheng H (2007) Amyolid
(6):895–907 precursor protein mediates presynaptic localization
Tarr PE, Contursi C, Roncarati R, Noviello C, Ghersi E, and activity of the high-affinity choline transporter.
Scheinfeld MH, Zambrano N, Russo T, D’Adamio L Proc Natl Acad Sci U S A 104:14140–14145
(2002) Evidence for a role of the nerve growth factor Wang B, Pan L, Wei M, Wang Q, Liu WW, Wang N,
receptor TrkA in tyrosine phosphorylation and Jiang XY, Zhang X, Bao L (2015) FMRP-mediated
processing of beta-APP. Biochem Biophys Res axonal delivery of miR-181d regulates axon elongation
Commun 295(2):324–329 by locally targeting Map1b and Calm1. Cell Rep
Teipel SJ, Flatz WH, Heinsen H, Bokde AL, Schoenberg 13:2794–2807
SO, Stockel S, Dietrich O, Reiser MF, Moller HJ, Wang M, Qin L, Tang B (2019a) MicroRNAs in
Hampel H (2005) Measurement of basal forebrain Alzheimer’s disease. Front Genet 10:153
atrophy in Alzheimer’s disease using MRI. Brain 128 Wang W, Tanokashira D, Fukui Y, Maruyama M,
(Part 11):2626–2644 Kuroiwa C, Saito T, Saido TC, Taguchi A (2019b)
Teipel SJ, Meindl T, Grinberg L, Grothe M, Cantero JL, Serine phosphorylation of IRS1 correlates with A-
Reiser MF, Möller H-J, Heinsen H, Hampel H (2011) β-unrelated memory deficits and elevation in Aβ level
The cholinergic system in mild cognitive impairment prior to the onset of memory decline in AD. Nutrients
and Alzheimer’s disease: an in vivo MRI and DTI 11:1942
study. Hum Brain Mapp 32:1349–1362 Whitehouse PJ, Price DL, Clark AW, Coyle JT, Delong
Tenreiro S, Eckermann K, Outeiro TF (2014) Protein MR (1981) Alzheimer disease: evidence for selective
phosphorylation in neurodegeneration: friend or foe? loss of cholinergic neurons in the nucleus basalis. Ann
Front Mol Neurosci 7:42 Neurol 10:122–126
Terasawa K, Ichimura A, Sato F, Shimizu K, Tsujimoto G Whitehouse PJ, PriceDL StrubleRG, Clark AW, Coyle JT,
(2009) Sustained activation of ERK1/2 by NGF De-lon MR (1982) Alzheimer’s disease and senile
induces microRNA-221 and 222 in PC12 cells. FEBS dementia: loss of neurons in the basal forebrain. Sci-
J. 276:3269–3276 ence 215:1237–1239
Thal DR, Rüb U, Orantes M, Braak H (2002) Phases of A Whitmer RA (2007) Type 2 diabetes and risk of cognitive
beta-deposition in the human brain and its relevance impairment and dementia. Curr Neurol Neurosci Rep
for the development of AD. Neurology 58:1791–1800 7:373–380
Tilvis RS, Kahonen-Vare MH, Jolkkonen J, Valvanne J, Willette AA, Bendlin BB, Starks EJ et al (2015) Associa-
Pitkala KH, Strandberg TE (2004) Predictors of cogni- tion of insulin resistance with cerebral glucose uptake
tive decline and mortality of aged people over a in late middle–aged adults at risk for Alzheimer dis-
10-year period. J Gerontol Ser A Biol Sci Med Sci ease. JAMA Neurol 72:1013
59:M268–M274 Wu Q, Ye X, Xiong Y, Zhu H, Miao J, Zhang W, Wan J
Triaca V, Sposato V, Bolasco G, Ciotti MT, Pelicci P, (2016) The protective role of microRNA-200c in
Bruni AC, Cupidi C, Maletta R, Feligioni M, Alzheimer’s disease pathologies is induced by beta
Nisticò R, Canu N, Calissano P (2016) NGF controls amyloid-triggered endoplasmic reticulum stress. Front
APP cleavage by downregulating APP phosphoryla- Mol Neurosci 9:140
tion at Thr668: relevance for Alzheimer’s disease. Wurtman RJ (1992) Choline metabolism as a basis for the
Aging Cell 15:661–672 selective vulnerability of cholinergic neurons. Trends
Triaca V, Coccurello R, Giacovazzo G (2018) The neuro- Neurosci 15:117–122
nal Shc adaptor in Alzheimer’s disease. Aging 10 Xu W, Weissmiller AM, White JA 2nd, Fang F, Wang X,
(1):5–6 Wu Y, Pearn ML, Zhao X, Sawa M, Chen S,
Tsanov M (2017) Speed and oscillations: medial septum Gunawardena S, Ding J, Mobley WC, Wu C (2016)
integration of attention and navigation. Front Syst Amyloid precursor protein-mediated endocytic path-
Neurosci 11:67 way disruption induces axonal dysfunction and
Vaegter CB, Jansen P, Fjorback AW, Glerup S, Skeldal S, neurodegeneration. J Clin Invest 126(5):1815–1833
Kjolby M, Richner M, Erdmann B, Nyengaard JR, Zatorre RJ, Fields RD, Johansen-Berg H (2012) Plasticity
Tessarollo L, Lewin GR, Willnow TE, Chao MV, in gray and white: neuroimaging changes in brain
Nykjaer A (2011) Sortilin associates with Trk receptors structure during learning. Nat Neurosci 15(4):528–536
to enhance anterograde transport and neurotrophin sig- Zhang YW, Thomposn R, Zang H, Xu H (2011) APP
naling. Nat Neurosci 14:54–61 processing in Alzheimer’s disease. Mol Brain 4:3
Verma S, Kumar A, Tripathi T, Kumar A (2018) Musca- Zhang YW, Chen Y, Liu Y, Zhao Y, Liao FF, Xu H (2013)
rinic and nicotinic acetylcholine receptor agonists: APP regulates NGF receptor trafficking and
10 NGF and the Amyloid Precursor Protein in Alzheimer’s Disease: From. . . 165

NGF-mediated neuronal differentiation and survival. Zhu QB, Unmehopa U, Bossers K, Hu YT, Verwer R,
PLoS One 8(11):e80571 Balesar R, Zhao J, Bao AM, Swaab D (2016)
Zhao W-Q, Alkon DL (2001) Role of insulin and insulin MicroRNA-132 and early growth response-1 in
receptor in learning and memory. Mol Cell Endocrinol nucleus basalis of Meynert during the course of
177:125–134 Alzheimer’s disease. Brain 139:908–921
A Review of Techniques for Biodelivery
of Nerve Growth Factor (NGF) 11
to the Brain in Relation to Alzheimer’s
Disease

Sumonto Mitra, Ruchi Gera, Bengt Linderoth, Göran Lind,


Lars Wahlberg, Per Almqvist, Homira Behbahani,
and Maria Eriksdotter

Abstract affected individual and associated family.


Age-dependent progressive neurodegeneration The cognitive dysfunctions are at least par-
and associated cognitive dysfunction represent tially due to the degeneration of a specific set
a serious concern worldwide. Currently, of neurons (cholinergic neurons) whose cell
dementia accounts for the fifth highest cause bodies are situated in the basal forebrain
of death, among which Alzheimer’s disease region (basal forebrain cholinergic neurons,
(AD) represents more than 60% of the cases. BFCNs) but innervate wide areas of the
AD is associated with progressive cognitive brain. It has been explicitly shown that the
dysfunction which affects daily life of the delivery of the neurotrophic protein nerve
growth factor (NGF) can rescue BFCNs and
restore cognitive dysfunction, making NGF
S. Mitra (*) · R. Gera interesting as a potential therapeutic substance
Division of Clinical Geriatrics, NVS Department, for AD. Unfortunately, NGF cannot pass
Karolinska Institutet, Stockholm, Sweden through the blood-brain barrier (BBB) and
e-mail: sumonto.mitra@ki.se; ruchi.gera@ki.se
thus peripheral administration of NGF protein
B. Linderoth · G. Lind · P. Almqvist is not viable therapeutically. NGF must be
Section of Neurosurgery, Department of Clinical
delivered in a way which will allow its brain
Neuroscience, Karolinska Institutet, Stockholm, Sweden
e-mail: bengt.linderoth@ki.se; goran.lind@sll.se; per. penetration and availability to the BFCNs to
almqvist@ki.se modulate BFCN activity and viability. Over
L. Wahlberg the past few decades, various methodologies
Gloriana Therapeutics Inc., Warren, RI, USA have been developed to deliver NGF to the
e-mail: luw@glorianatx.com brain tissue. In this chapter, NGF delivery
H. Behbahani methods are discussed in the context of AD.
Division of Clinical Geriatrics, NVS Department,
Karolinska Institutet, Stockholm, Sweden
Karolinska Universitets laboratoriet (LNP5), Karolinska
University Hospital, Stockholm, Sweden
e-mail: homira.behbahani@ki.se Abbreviations
M. Eriksdotter
Division of Clinical Geriatrics, NVS Department, Aβ Amyloid-beta
Karolinska Institutet, Stockholm, Sweden
AD Alzheimer’s disease
Theme Aging, Karolinska University Hospital, Huddinge, APP Amyloid precursor protein
Sweden
BBB Blood-brain barrier
e-mail: maria.eriksdotter@ki.se

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 167
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_11
168 S. Mitra et al.

BFCN Basal forebrain cholinergic neuron dysfunction and Alzheimer’s disease (AD). The
CSF Cerebrospinal fluid currently available drugs against AD targeting the
ECB Encapsulated cell biodelivery cholinergic system, the cholinesterase inhibitors,
EEG Electroencephalography have a modest albeit persistent effect on cognition
FTD Frontotemporal dementia (Birks and Harvey 2018; Zhu et al. 2013; Secnik
HIBI Hypoxic-ischemic brain injuries et al. 2020; Dou et al. 2018). Among many other
ICD Intracerebral delivery targets, several clinical trials have attempted the
ICVD Intracerebroventricular delivery reduction of amyloid-beta (Aβ) peptides or
MMP3/ Matrix metalloproteinase 3/9 plaques, although a majority of the trials have
9 not produced convincing result so far in improv-
mNGF Mature-NGF ing cognition (Cummings et al. 2019, 2020;
MSCs Mesenchymal stem cells Scheltens et al. 2016; Liu et al. 2019). However,
nbM Nucleus basalis of Meynert in the last year, modest positive results on cogni-
NGF Nerve growth factor tion have been attributed to aducanumab, a mono-
NGFR NGF receptor clonal antibody against Aβ (Schneider 2020), and
NSCs Neural stem cells approval by the American FDA is now pending. It
p75 Low-affinity NGF receptor or is important, however, to widen the search for
LNGFR, or p75 neurotrophin therapies for AD, and therefore, delivery of
receptor, or p75NTR NGF as one therapeutic approach is of great
PLGA Poly(lactic-co-glycolic acid) interest.
proNGF Precursor-NGF Under normal physiological conditions, NGF
rh Recombinant human is secreted from cells in a precursor form
TBI Traumatic brain injury (proNGF) which needs to be converted into
tPA Tissue plasminogen activator mature form (mNGF) (Fahnestock and Shekari
TrkA Tropomyosin receptor kinase A 2019; Cuello et al. 2019). This conversion of
uPA Urokinase plasminogen activation proNGF to mNGF is done by the enzyme plas-
min, which itself is generated from the proteolytic
cleavage of plasminogen by tissue plasminogen
activator (tPA) or urokinase plasminogen activa-
11.1 Introduction and Background
tion (uPA), respectively (Bruno and Cuello
2006). Once formed, mNGF can initiate its down-
NGF was the first neurotrophic factor discovered
stream signaling through its receptor interaction
in the 1950s by Rita Levi-Montalcini and Viktor
with TrkA (tropomyosin receptor kinase A) or
Hamburger, which established the family of
p75 (known as low-affinity NGF receptor,
neurotrophic factors and further led to the discov-
LNGFR; or p75 neurotrophin receptor, or
ery of other related proteins (Cohen and Levi-
p75NTR), both collectively called
Montalcini 1956; Cohen et al. 1954; Bradshaw
NGF-receptors (NGFR). Alternatively, mNGF
et al. 2017). Due to its involvement in various
can be further degraded and cleared from the
physiological processes, NGF is a highly
system by the matrix metalloproteinase 3/9
researched molecule (Aloe et al. 2012, 2015,
(MMP3 and MMP9) enzyme. Depending on this
2016; Lambiase et al. 2011; Bracci-Laudiero
metabolic dynamics-controlled availability of dif-
and Manni 2014; Mitra et al. 2019; Simone
ferent NGF forms and expression of the receptor,
et al. 1999; Rocco et al. 2018). A quick search
the final biological effects are initiated (as shown
in PubMed (https://pubmed.ncbi.nlm.nih.gov/)
in Fig. 11.1). If NGF maturation is hampered,
using the keywords “nerve growth factor”
proNGF levels may get accumulated and affect
retrieved more than 74,190 results (as of 29th
neuronal survival through the cooperative signal-
December, 2020). Interestingly, hampered NGF
ing of p75NTR and sortilin receptor (Capsoni
metabolism has also been linked to cognitive
et al. 2013; Skeldal et al. 2012).
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 169

Plasminogen
tPA/uPA
proNGF NGF NGF degradation
Plasmin MMP3/9

Pro-domain

p75 Sortilin p75 TrkA

P P

JNK PKB/AKT MAPK PKC

NF-κB
Caspases

c-Jun TFs Cell Survival &


NF-κB differentiation
Apoptosis

Cell Survival

Fig. 11.1 NGF signaling in the brain. NGF is initially proNGF (precursor-NGF), mNGF (mature-NGF), TrkA
released as proNGF from cells, which is then metabolized (tropomyosin receptor kinase A), p75 (p75 neurotrophin
to mNGF by plasmin, and mNGF is ultimately degraded receptor), MMP3/9 (matrix metalloproteinase 3 or 9), tPA
by MMP3/9 enzyme. NGF (pro or mature) can induce (tissue plasminogen activator), uPA (urokinase plasmino-
different downstream signaling based on their interaction gen activation), JNK (c-Jun N-terminal kinases), NK-κB
with various NGF receptor combinations (i.e., TrkA-p75 (nuclear factor kappa-light-chain-enhancer of activated B
or p75-sortilin). Based on specific NGF and receptor type cells), PKB/AKT (protein kinase B), MAPK (mitogen-
interaction, cell signaling cascades for apoptosis (death), activated protein kinase (MAPK), PKC (protein kinase
survival, or differentiation take place via specific signaling C), and TFs (transcription factors). ‘P’ denotes phosphor-
pathways. Abbreviations: NGF (nerve growth factor), ylation sites

Under AD condition, the level of mNGF is been reported in AD (Mufson et al. 1995). Recent
reduced while proNGF is elevated (Fahnestock studies have confirmed the hampered activity of
and Shekari 2019), implying hampered NGF tPA and elevated levels of MMP3/9 in AD,
metabolism. This could be due to decreased tPA indicating reduced production and enhanced
levels (reduced mNGF production), upregulated clearance of mNGF from the brain tissue (Hanzel
MMP9 activity (enhanced clearance of mNGF), et al. 2014; Pentz et al. 2020). Reduced mNGF
altered TrkA/p75 mediated signaling, or a combi- levels hinder cholinergic signaling (production of
nation of all of them (Mitra et al. 2019). Simulta- the neurotransmitter—acetylcholine) resulting in
neously, hampered retrograde transport of NGF inflammation due to glial activation. Thus, restor-
from hippocampus and cortical tissue has also ing the mNGF levels in the brain for recovery of
170 S. Mitra et al.

NGF delivery methods

Peripheral Intracerebroventricular Intracerebral

Intranasal and Stem Cell Intracerebral


Parenteral Tissue graft
Intraocular mediated infusion

Encapsulated
Gene therapy
biodelivery
Genetically Focused Small
Carrier
modified cell ultrasound molecule
mediated
mediated guided mediators
Genetically Viral vector Non-
•Nanoparticles modified cell direct Biodegradable
Biodegradable
•Microspheres mediated delivery
•Conjugates
•Conduits

Fig. 11.2 Various methods for NGF delivery. NGF is broadly classified as peripheral, intracerebroventricular,
delivered using various strategies targeting the brain tis- and intracerebral, respectively. Further classification is
sue. Based on the initial administration route, methods are based on the technical method employed to deliver NGF

the cholinergic signaling in various clinical and must diffuse wide enough to reach the target cells
pre-clinical studies has attracted significant atten- for an efficacious outcome, and (3) the drug must
tion (Hampel et al. 2018). be delivered in a stable manner over long term to
Although the incidence of dementia has been enhance the probability of a clinically relevant
reported to be declining in high-income countries outcome. Although various delivery methods
(Satizabal et al. 2016), recent results are have been utilized for several types of
conflicting (Binder et al. 2019). Thus, the effort neurotrophic molecules, few newer options
to combat AD-related neuronal dysfunction showing increased clinical efficacy have emerged
remains a primary concern. In the last four in the last few decades (Olson 1993; Ruozi et al.
decades, various research groups have tried 2012; Yi et al. 2014), as further described.
to deliver mNGF to the brain tissue in order to
revive the cholinergic signaling with an aim to
recover cognition. Although one of the major
11.2 Different Methods of NGF
issues with brain delivery of NGF is the intrinsic
Delivery
complexity of delivering drugs continuously for
extended periods of time to achieve a clinical
Based on the tissue targeting ability, the various
output, one major problem is the biological
methods utilized for NGF delivery can be broadly
off-target impact of mNGF on nociception and
classified into three major classes: (1) peripheral
pain induction (Denk et al. 2017; Chang et al.
delivery (systemic administration), (2) intracere-
2016). To achieve an efficacious therapy, some
bral delivery (ICD) (within brain parenchyma),
of the following conditions needs to be fulfilled—
and (3) intracerebroventricular delivery (ICVD)
(1) the drug has to be delivered precisely to the
(to the cerebrospinal fluid—CSF). These classes
requisite target region of the brain at appropriate
are subdivided into different methods depending
quantity to stimulate intended outcome while
on the technical approach as shown in Fig. 11.2.
reducing off-target effects, (2) the delivered drug
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 171

11.2.1 Intracerebroventricular et al. 2014) but also identified hypophagic effect


Delivery (ICVD) of NGF in rats leading to weight loss (Williams
1991). It is at this juncture in 1990 that possible
Early studies during 1970s–1980s demonstrated NGF therapy for AD condition was proposed,
the correlation between loss of BFCNs and pro- based on the ability of NGF to recover lesioned
gressive memory deficits in AD patients, which cholinergic neurons.
led to the proposal of the cholinergic theory of In the following years, Koliatsos et al.
geriatric memory dysfunction in 1982 (Bartus (1991a, b) prepared the recombinant human
et al. 1982). Before and after the proposal of the NGF (rhNGF) protein and showed its efficacy
cholinergic hypothesis, several reports have on rats to revive lesioned BFCNs. For the first
demonstrated the consequence of declining NGF time, Olson and coworkers from Karolinska
levels and the potential therapeutic effects of Institutet showed that mouse NGF (75 μg/day
exogenous NGF delivery with respect to memory for 3 months) could be therapeutically delivered
function (Cuello et al. 2019; Hampel et al. 2018; to the lateral cerebral ventricle of one AD patient
Chen and Mobley 2019). One of the earliest and reported improved 11C-nicotine binding,
methods for delivery of NGF to the brain tissue blood flow, electroencephalography (EEG), and
was by direct injection of NGF to the CSF using verbal episodic memory (Olson et al. 1992;
the ICVD method (Hefti and Schneider 1989). Seiger et al. 1993). These encouraging results
Starting in 1986, different groups reported the prompted the group to perform another trial on
administration of purified NGF protein to adult 3 AD patients which apart from replicating simi-
rats where ICVD NGF infusion over 4 weeks lar beneficial effects on 11C-nicotine binding and
resulted in enhanced survival of cholinergic improved blood-flow unfortunately demonstrated
neurons post fimbria transection (Williams et al. side effects which included back pain and weight
1986; Hefti 1986). Subsequently, Fischer et al. loss (Eriksdotter Jonhagen et al. 1998). These
demonstrated that NGF infusion in old rats for side effects subsided when the NGF infusion
similar duration recovered spatial memory and was stopped, but the patients continued to show
reduced neuron atrophy (Fischer et al. 1987). improved cerebral blood flow for several months
Further studies showed that NGF not only was after the infusion was terminated (Jonhagen
able to recover damaged neurons, but NGF could 2000). Follow-up experiments in rats receiving
also dramatically restore the cholinergic pheno- 7 or 0.7 μg/day NGF intracerebroventricularly
type from a point when no cholinergic cells were for 2 weeks reproduced the pain-inducing effects
apparent by immunostaining of the brain tissue, of NGF while delivery to the brain parenchyma
indicating that NGF is needed for maintaining the did not elicit pain (Hao et al. 2000). The ICVD
cholinergic phenotype (Hagg et al. 1988). method has been shown to allow minimal diffu-
Subsequent rodent studies demonstrated the pres- sion of delivered neurotrophic molecule into the
ence of NGFRs on the BFCNs, and the impor- brain parenchyma, thereby reducing the
tance of NGF for regulation of its receptors biological effectiveness (Yan et al. 1994). Due
(Montero and Hefti 1988; Montero and Hefti to these off-target effects, NGF delivery by
1989). These findings were replicated in higher ICVD was not recommended at higher concentra-
mammals (i.e. monkeys) using mouse-derived tion, but a later study used a combination of NGF
NGF (Koliatsos et al. 1990; Tuszynski et al. with other neurotrophic factors to enhance cho-
1990) showing the conservation and efficacy of linergic signaling in rats (Tirassa et al. 2003).
NGF cross-species. Several other reports showed Interestingly, years after the NGF infusion studies
the beneficial impact of NGF infusion on the were stopped for AD, NGF ICVD infusion was
cerebral vasculature, cellular regeneration, and shown to be capable of offering neuroprotective
proliferation (Winkler et al. 1997; Saffran et al. effects in two new-born children suffering from
1989; Isaacson et al. 1990, 1992; Schlachetzki hypoxic-ischemic brain injuries (HIBI) with
172 S. Mitra et al.

prolonged comatose state, wherein NGF infusion long-lasting protection of cholinergic cells until
led to normalization of EEG and cerebral perfu- after 6 months post NGF therapy (Tuszynski and
sion (Fantacci et al. 2013; Chiaretti et al. 2005). Gage 1995). These models provide exciting data
These studies do not report the impact of NGF on about the possible use of embryonic grafts to
pain induction in these children, which otherwise revive cholinergic tissue in injury models, but
would be an interesting data to evaluate. This these studies do not allow the possibility to eval-
difference may be explained by a different uate the consequences of grafting in a relevant
response to NGF in young versus old brains, AD setting. Moreover, issues regarding transplant
indicating that the restorative capabilities of rejection by the host immune system could be a
NGF are quite strong, especially when serious setback. Attempts have been made to use
administered in younger individuals. autologous adrenal tissues to revive motor deficits
in Parkinson’s patients wherein NGF infusion for
a month was performed to support the adrenal
11.2.2 Intracerebral Delivery grafts (Sydow et al. 1995), but need for regular
NGF infusion and engraftment of non-nervous
To circumvent the off-target effects of NGF (pain tissue in the brain makes the delivery less
and weight loss), targeted delivery within the feasible.
brain parenchyma is considered a preferred
method of choice. Due to the different dynamics 11.2.2.2 Intracerebral Infusion
of drug distribution and clearance pathways As described earlier, several groups have
within the brain tissue (when compared to CSF), experimented with direct delivery of NGF infu-
NGF delivery has been attempted by using vari- sion into the cerebral tissue, in order to avoid the
ous technical methods described further. side effects of ICVD delivery of NGF. This
method was established by Williams et al.
11.2.2.1 Implantation of Tissue Grafts (1987) when they improved the accuracy of ste-
Due to the dependence of BFCN’s on cortical and reotaxic implantation of a cannula within the
hippocampal regions for retrograde NGF supply brain tissue and combined it with the capability
(Mitra et al. 2019), early studies have of Alzet-pumps for continuous infusion of thera-
demonstrated the possibility of using fetal tissue peutic substances. Further support for intracere-
grafts in the cortical regions of the brain to restore bral infusion came from the work of Venero et al.
the BFCNs (Fine et al. 1985; Springer et al. (1996), who demonstrated that 100 times lower
1988a). Briefly, a cortical lesion is introduced NGF concentration is capable of inducing similar
(by chemical agents, e.g., Ibotenic acid) which biological effects when NGF is administered
has been shown to reduce the cholinergic function intracerebrally as compared to ICVD. This pre-
of the basal forebrain in rats, which is then res- ferred method of NGF delivery was later utilized
cued by implanted fetal ventral forebrain tissue to rescue BFCNs in a model of fimbria-fornix
rich in NGF producing cells. These grafts have transection (Tuszynski 2000). This approach of
been shown to induce reversal of memory deficits NGF delivery was not utilized in AD-related
introduced by the cortical lesion. Due to extensive models, possibly due to the enhanced degradation
production of NGF by the submaxillary glands in of NGF in the AD brain environment which is due
rodents, these tissues have also been used as to increased activity of MMP3/9 enzyme activity
grafts in the lateral ventricle of rats, resulting in (Pentz et al. 2020; Allard et al. 2012; Hanzel et al.
increased cell survival after fimbria-fornix tran- 2014). Interestingly, NGF infusion to the brain
section (Springer et al. 1988b). Subsequently, tissue has been shown to reduce the impact of
grafts of hippocampal tissue isolated from viral infection and protect cells from lysis post
embryos (day 18) transplanted in fimbria-fornix infection (Pakzaban et al. 1994), which is
transection models along with simultaneous regarded as one of the latest discovered etiologies
ICVD infusion for NGF for 9 weeks provided of AD (Sochocka et al. 2017; Cairns et al. 2020;
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 173

Balin and Hudson 2018). A robotically assisted cells themselves, some studies reported the use
placement of a catheter system has been recently of stem cell-derived extracellular vesicles as a
developed to intermittently deliver infusions to therapeutic option when injected intracerebrally
brain parenchyma, although this has not been or delivered through intranasal passage in animal
tested to deliver NGF in an AD setting (https:// models of AD (Elia et al. 2019; Losurdo et al.
www.renishaw.com) (Killick-Cole et al. 2019). 2020).

11.2.2.3 Stem Cell Mediated 11.2.2.4 Gene Therapy


Stem cells have been used for several decades in To enhance delivery, without complication from
the field of regenerative medicine, as an inex- NGF-associated side effects and dysregulation of
haustible source of self-replicative cells which stem cell behavior (aberrant differentiation, sur-
can replace damaged tissue and cells (Polak and vival, immune response), different approaches
Mantalaris 2008; De Luca et al. 2019). Certain have been undertaken by utilizing gene therapy.
type of stem cells, neural stem cells (NSCs) and Either cells are transfected with NGF-producing
mesenchymal stem cells (MSCs), has been shown vectors and then injected, or viral particles are
to produce NGF and other neurotrophic factors directly injected into the brain areas which are
(Kamei et al. 2007; Paradisi et al. 2014). Con- targeted to express and be revived by NGF. One
versely, various neurotrophic molecules have advantage over allogeneic grafting is the use of
been shown to influence stem cell behavior; for autologous cells which are immune-compatible
example, proNGF has been shown to induce pro- with the host, while the viral particles can target
liferation in NSCs while mNGF is needed for its multiple cell types at the injection site, transduce
differentiation, indicating the importance of NGF them with NGF coding vectors and induce NGF
metabolism in maintaining neuron generation secretion locally. But the disadvantage with both
from NSCs (Corvaglia et al. 2019; Pramanik approaches is that once injected, these cells or
et al. 2017). Several studies have reported the viral particles cannot be recovered. These
neuroprotective ability of different stem cell approaches are further elaborated.
populations on cell regeneration,
neuroinflammation, and neurobehavior when Use of Genetically Modified Cells To secrete
implanted within the brain tissue under different enhanced levels of NGF in an AD context, the
pathological conditions including AD (Kim et al. use of genetically modified cells was initiated in
2010, 2020; Gu et al. 2015; Lee et al. 2012a; Ager the late 1980s when a mouse fibroblast cell line
et al. 2015; McGinley et al. 2018; Zhang et al. was transfected with rat-NGF-secreting vectors
2014). Unfortunately, recent reports indicate that and implanted into rat brains (Ernfors et al.
amyloid precursor protein (APP) expression and 1989). Improved survival of fetal cholinergic neu-
Aβ peptides can modulate the stem cell prolifera- ronal grafts and increased sprouting of adult
tion and differentiation, thereby affecting their neurons/interneurons were observed when
therapeutic efficiency and survival (Kwak et al. mixed with genetically modified NGF delivering
2006; Sugaya 2008; Bernabeu-Zornoza et al. fibroblast during implantation to cortical tissue in
2019). Stem cell therapy is also complicated by a model of unilateral fimbria fornix dissection.
the involvement of immune rejection wherein the The same group later showed that continuous
host immune system attacks the implanted stem NGF delivery can be successfully achieved over
cells which reduces the therapeutic efficacy and several months by implanting genetically
aggravates inflammatory responses (Liu et al. modified NSCs to aged rat brains, leading to
2017). Thus, the efficiency of stem cell therapy functional recovery in spatial memory
especially in the case of AD needs to be evaluated (Martinez-Serrano et al. 1996; Martinez-Serrano
carefully (Hayashi et al. 2020; Wang et al. 2019). and Bjorklund 1998). Similar studies were
Significant improvements in this field has been reported by other groups where autologous
made recently, wherein instead of using stem fibroblasts were used to harbor NGF-expressing
174 S. Mitra et al.

vectors and were implanted in the brain of adult genetically modified cell section) and two
primates long term (Tuszynski et al. 1996). patients receiving in-vivo AAV2 injection
Follow-up work in rats demonstrated the capabil- showed enhanced NGF signaling and protein
ity of genetically identical but modified levels in the brain tissue post-mortem (Tuszynski
fibroblasts to provide partial recovery from spinal et al. 2015). This encouraging data using AAV2-
cord injury even when administered several NGF vectors was further translated to a dose-
months post-injury induction (Grill et al. 1997). escalating phase 1 study on ten AD patients
These promising effects of NGF delivery using which showed that the delivery was feasible in
ex-vivo gene manipulation led to the first-in- humans, well tolerated, safe, and resulted in long-
human trial of ex-vivo gene therapy and were term NGF expression (Rafii et al. 2014). Further
reported in 2005 by Tuzynski et al. (2005). A analysis of post-mortem brains from the phase
10-year follow-up (until brain collection from 1 dose escalation study revealed that although
those patients’ post-mortem) showed response to AAV2-NGF vectors were capable of inducing
the therapy by neuronal sprouting toward the NGF signaling for almost 7 years in the injected
implanted fibroblasts (Tuszynski et al. 2015), sites, those sites were not within the cholinergic
indicating long-lasting effect of NGF administra- nuclei (nucleus basalis of Meynert, nbM) which
tion. NGF delivering human NSCs implanted in were intended to be activated by the therapy. Due
the brain tissue has also been shown to improve to the limited spread of the injected AAV2-NGF
cognitive functions in ibotenic acid-induced hip- vectors coupled with incorrect stereotactic
pocampal lesion in rats and aging mice, respec- targeting, the clinical trial was not able to induce
tively (Lee et al. 2012b; Park et al. 2013). the cholinergic pathways which were needed for
the improvement of cognition in AD patients
Viral Vector Direct Delivery With the advent of (Castle et al. 2020). A follow-up multicenter pla-
methodologies which offered greater control on cebo controlled phase 2 clinical trial on 49 AD
gene expression (reducing uncontrolled patients replicated the observation from the phase
non-specific viral infection in tissues), hybrid I study that AAV2-NGF delivery was safe but
viral vectors allowed the revival of virus- failed to show any considerable improvement in
mediated direct gene delivery, which had been cognitive score at 2-year follow-up (Rafii et al.
associated with several technical problems 2018), in part because the vector targeting of the
(Thomas et al. 2003; Goswami et al. 2019). One brain was unsatisfactory.
of the first reports of NGF delivery using viral
vectors was performed using a hybrid viral vector 11.2.2.5 Encapsulated Biodelivery
containing the AAV2 genome with AAV sero- With the prospect of developing new tools for
type 5 (AAV2/5) in a rat model of fimbria fornix targeted delivery of neurotrophins to the brain,
transection (Wu et al. 2005). As compared to several important aspects had to be considered,
AAV2 vector, AAV2/5 vectors consistently including the complexity and format of delivery
showed higher transduction efficiency, while the (stereotactic injection/implantation, ICVD,
duration of gene expression in both cases was peripheral delivery) targeting the correct tissue
similar (17 weeks). Simultaneously, other groups or cell type, potential side effects with uncon-
using AAV2 vectors expressing NGF in various trolled delivery (viral vectors), immune rejection
pre-clinical studies could show stable gene (stem cells), and many others. The necessity of
expression for long term (1 year), paving the delivering NGF to the correct anatomical struc-
way for trials in AD patients (Bishop et al. ture within the brain tissue highlights the need for
2008; Nagahara et al. 2009). a delivery system which can deliver NGF in pre-
A pilot phase 1 clinical trial on ten AD cise location for an extended period of time in
patients, eight patients receiving ex-vivo geneti- order to elicit a relevant biologic response trans-
cally modified fibroblasts (described under lating into a positive clinical outcome (Mahoney
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 175

and Saltzman 1999). One of the delivery methods therapeutic protein over a longer time, and due
that was developed to achieve these goals was the to the possibility to use different types of cells
encapsulated cell biodelivery (ECB, using cells within the device and its implantation to any
for encapsulation) or encapsulated biodelivery desired location, this kind of ECB systems were
(EB, using protein encapsulation). This technique preferred to deliver NGF in-vivo/in-situ (Date
essentially contains two parts: (1) a protein of et al. 1996; Orive et al. 2015; Emerich et al.
interest to be delivered or a genetically modified 2014).
cell which produces a therapeutic protein and Tremendous efforts and significant optimiza-
(2) a semi-permeable membrane for encapsula- tion of cell types, gene expression vectors, and
tion. Similarly, the ECB/EB technique can be ECB engineering led to the first successful open-
considered as a hybrid method which possesses label human phase1 clinical trial using ECB-NGF
the qualities of targeted in-situ delivery, long- in 6 AD patients by our group (Eriksdotter-
term release, with a biologically (immunologi- Jonhagen et al. 2012; Wahlberg et al. 2012).
cally) safe method. Thus, the fate of the ECB This study showed that ECB-NGF therapy in
cells/EB-proteins can be verified. Methods AD patients is feasible, safe, and well tolerated
utilizing encapsulated delivery can be for up to 1 year (experimental endpoint of the
differentiated based on the material used to pre- study). Improvement in cognition, EEG, and nic-
pare the outer membrane, and its contents within otinic receptor binding were reported, along with
(cells or pure proteins). Broadly, they are classi- improved cholinergic markers in CSF and
fied as biodegradable and non-biodegradable as reduced basal forebrain atrophy in three patients
further discussed. (Karami et al. 2015; Machado et al. 2020; Ferreira
et al. 2015). An improved version (second gener-
Non-biodegradable This type of encapsulation ation) of the ECB-NGF devices was developed
devices contains an outer semi-permeable mem- which released ten times more NGF and was
brane which is non-biodegradable but biocompat- utilized in a follow-up dose escalation study in
ible, allowing the influx of nutrients and oxygen four AD patients for 6 months, confirming the
necessary for cell survival with simultaneous observations of beneficial effects of first trial
out-flow of the therapeutic molecule (Fjord-Larsen et al. 2012; Eyjolfsdottir et al.
(s) (applicable for ECB). Initially, polymer- 2016; Machado et al. 2020). The NGF releasing
coated pellets of purified NGF proteins were ability of the ECB-NGF devices was found to be
used to deliver NGF over a limited time period affected post-explantation after 12 months and
of time (Powell et al. 1990; Hoffman et al. 1990; after 6 months, respectively, and further in-vitro
Saltzman et al. 1999). However, remarkable studies identified the impact of inflammatory
innovations allowed the use of live genetically cytokines on the NGF releasing cells (Eriksdotter
modified cells encapsulated within a biocompati- et al. 2018).
ble semi-permeable membrane (Tresco et al. Further identification of “factors” which can
1992). Using encapsulated NGF-transfected affect ECB-NGF cells and by modifying the cells
fibroblast cells, neuroprotective effects were to become resistant against these “factors” may
observed in an ibotenic acid rodent model of further improve our ECB-NGF therapy to attain
striatal degeneration (Emerich et al. 1994a). Sim- long-term (several years) clinical efficacy. One
ilarly, NGF delivery using transfected baby ham- major drawback of using non-biodegradable
ster kidney cells in a primate model of fornix ECBs is the invasive procedure which is needed
transection showed enhanced sprouting of septal for its implantation. On the contrary, the major
cholinergic neurons (Emerich et al. 1994b; advantage is that once ECB is placed in its target
Kordower et al. 1994). With the advantage of region, no further drugs or modifications are
potential long life and regeneration capacity of required for an extended period (up to 1 year
cell lines which can produce the intended currently). The ECB can be removed from the
176 S. Mitra et al.

brain if needed to terminate the therapy giving release NGF over extended time periods and
full control over the procedure and treatment demonstrated enhanced properties to induce
duration. Additionally, a “reloading system” NGF-mediated signaling (Sakiyama-Elbert and
may be applied in the future to enable the replace- Hubbell 2000) and protect from neurotoxicity
ment of old with new ECB device, thereby (Menei et al. 2000). Further optimization of
extending clinical therapy duration without the encapsulation materials produced
need of stereotactic procedures during follow-up thermosensitive and chemosensitive materials
procedures. Similarly, due to the presence of which allow activity-based release of NGF from
active protein-releasing cells, ECBs overcome the encapsulation matrix (Lee et al. 2003; Zhao
the problem of protein storage (like in Alzet et al. 2016; Hu et al. 2020). In a few applications,
pumps) which would otherwise affect protein NGF protein was modified to form polypeptides
quality, aggregation, and biological activity due to control its release and diffusion (Sakiyama-
to storage conditions of pure proteins. Presently, Elbert et al. 2001; Song et al. 2012). Currently,
many other types of cell encapsulation procedures nanostructured carriers are being tested for its
are being evaluated in tandem with different efficiency to deliver NGF in models of spinal
encapsulation materials which may lead to further cord injury and AD (Zhu et al. 2016; Zilony-
advancements in the field (Orive et al. 2019; Oh Hanin et al. 2019), although various optimization
et al. 2020; Son et al. 2017). in the delivery vehicles have been proposed for
the future (Wang et al. 2017).
Biodegradable This class of EBs uses encapsu-
lation materials which are usually biological
molecules which could interact with the
11.2.3 Peripheral Delivery
surrounding tissue upon implantation and would
be degraded over time to allow slow and
Due to the invasiveness of procedures mentioned
sustained release of materials encapsulated within
in previous sections (Sects. 11.2.1 and 11.2.2),
them. Usually these EBs deliver therapeutics over
other less-invasive options have been proposed to
a short period of time (days to weeks), due to their
reduce risk and complications associated with the
dissolution by the host tissue enzymes and limited
more invasive NGF delivery methods described
amount of storage. The advantage is its biocom-
earlier. Peripheral delivery methods have the pos-
patibility and ability to release therapeutics in-situ
sibility to consider different approaches which are
which allows its implantation within the tissue.
not bound to NGF protein or gene delivery (small
However, one of the drawbacks would be the
molecules, coated vesicles, etc.) as mentioned in
inability to control their activity (protein release
the following and has the advantage of continu-
and degradation over time), or explantation
ous multiple administration, which is not possible
(if needed) along with their alteration in shape
with invasive approaches. Conversely, one of the
due to tissue dynamics and sensitivity towards
major disadvantages is the inability to control the
enzymes, pH or temperature. Although the use
tissue-targeted delivery of the delivered molecule
of biodegradable encapsulation strategies was
and associated side effects on other body organs.
utilized to deliver various biological materials
Aberrantly increasing NGF signaling has been
from the early 1980s (Lim and Sun 1980), deliv-
shown to be associated with different forms of
ery of NGF using in-vitro and in-vivo rodent
cancer in vulnerable organs (Aloe et al. 2016;
models was developed from 1990s and onward
Hayakawa et al. 2017; Blondy et al. 2019; Griffin
(Camarata et al. 1992; Xu et al. 2002; de Boer
et al. 2018). Several approaches have been
et al. 2010). Optimization of the materials used in
undertaken to achieve NGF delivery to the brain
the encapsulation of NGF led to the production of
through parenteral administration by overcoming
biodegradable microspheres which can slowly
these limitations, as further described.
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 177

11.2.3.1 Intranasal and Intraocular rescue cognitive deficits in animal models of


Delivery AD (Capsoni et al. 2002,2012; De Rosa et al.
Administration of therapeutics has been tried for 2005). Using a mouse model of AD which intrin-
various drugs through the ocular and nasal sically produces anti-NGF antibodies, thereby
passages owing to their direct connection to the depriving the brain tissue of autologously pro-
olfactory and frontal brain regions (Ganger and duced NGF (Capsoni et al. 2011), a direct com-
Schindowski 2018; Mandal et al. 2018; Pardridge parison between ocular and nasal administration
2019). Large molecules like NGF, which is also of recombinant NGF demonstrated enhanced dis-
very sensitive to enzymatic degradation, need a tribution and efficacy of intranasally delivered
direct access to brain tissue to generate a compre- NGF to rescue the phenotypic changes of these
hensive clinically relevant effect, which is actu- mice (Capsoni et al. 2009; Covaceuszach et al.
ally offered by the ocular and nasal passages since 2009). Moreover, intranasal delivery of therapeu-
NGF delivered through these routes bypasses the tics has also been shown to be more effective than
BBB without reaching systemic circulation. intraperitoneal administration in reaching the
Although NGF has been used to treat various brain tissue, in part by bypassing the BBB
eye disease conditions and NGF expression has (Chauhan and Chauhan 2015). Thus, the utility
been found to be increased in optic-nerve tissue of the intranasal approach to deliver NGF seems
(Lambiase et al. 1998; Bonini et al. 2000; more promising than other non-invasive
Lambiase et al. 2005; Guo et al. 2020), the approaches (considering the delivery of NGF pro-
mechanisms of delivery to the brain tissue tein). The efficiency of intranasal NGF delivery-
through this route are not thoroughly understood. mediated neuroprotection has been demonstrated
Moreover, as mentioned previously, NGF deliv- in two female patients with frontotemporal
ery may induce side effects including pain and dementia (FTD) who received NGF therapy
weight loss and using NGF in its natural full- only (for 12–16 months) and were compared to
length form intranasally or intraocularly may another two female patients who were treated
elicit adverse side effects. Discovery of a mutated with memantine and idebenone therapy (de Bellis
form of NGF, termed painless NGF, owing to its et al. 2018). Another study on a 4-year-old trau-
cholinergic signaling inducing properties without matic brain injury (TBI) patient showed a benefi-
inducing pain (Minde et al. 2004; Cattaneo and cial impact of intranasal NGF therapy, resulting
Capsoni 2019), led to studies on this mutated in improved voluntary responses and clinical con-
form for NGF delivery (mentioned below in this dition (Chiaretti et al. 2017). As discussed previ-
section). Nevertheless, there are a few studies ously, although intranasal NGF delivery shows
indicating the possibility to stimulate brain great promise as a translational candidate for
regions through ocular delivery of NGF without future human use, the complex anatomy of the
generating anti-NGF antibody response from the nasal cavity and its interaction with other physio-
host (Lambiase et al. 2007a, b; Tirassa et al. logical functions needs to be evaluated closely
2018), but whether the effects can enhance cogni- prior to the development of a clinically relevant
tive abilities in the context of AD has not yet been therapy (Erdo et al. 2018; Bourganis et al. 2018;
explored. Gomez et al. 2012).
In comparison to ocular delivery, intranasal
delivery of NGF has already been a topic of
11.2.3.2 Parenteral Delivery
thorough research during the early days of NGF
Apart from the delivery methods discussed Sect.
therapy development in 1990s where it was
11.2.3.1, most of the peripheral delivery methods
shown to efficiently reach the brain tissue to
utilize parenteral delivery, i.e., injections of vari-
induce NGF signaling (Altar and Bakhit 1991;
ous molecules in different forms delivered intra-
Chen et al. 1998). Intranasal administration of
venously, to be further discussed.
NGF was found to show neuroprotection and
178 S. Mitra et al.

Small Molecule Mediators Due to the realiza- proteolytic enzymes when present in the blood,
tion that stimulation of the TrkA receptor is and since NGF cannot cross the BBB, it is neces-
needed for cell survival, or conversely possible sary to conjugate it with various carrier
inhibition of p75 may reduce cell death, and that molecules. Diverse methods of conjugation have
both of these biological effects can be activated been applied to different kinds of carrier
without the need for an actual neurotrophin mol- molecules, as further discussed.
ecule, neurotrophin mimicking compounds
(peptides and chemical analogues) were devel- Protein Conjugated Direct conjugation of NGF
oped from the late 1990s (Spiegel et al. 1995; to other proteins has been tried to achieve
Estenne-Bouhtou et al. 1996; Longo et al. 1997; receptor-mediated uptake through BBB. Initial
LeSauteur et al. 1995). With the advent of time reports in 1990s showed the efficacy of the anti-
and technology, various kinds and classes of transferrin receptor antibody-NGF conjugates
small-molecule agonists and antagonists have injected intravenously in reviving septal choliner-
also been developed toward TrkA and p75 gic and non-cholinergic grafted neurons
receptors (Mitra et al. 2019; Covaceuszach et al. implanted in the anterior chamber of the eye
2009; Cattaneo et al. 2008; Longo and Massa (Friden et al. 1993; Granholm et al. 1994). Intra-
2013). Among the various molecules developed venous NGF-conjugated delivery was found to be
till date, a small-molecule p75 modulator effective in rescuing BFCNs from excitotoxic
(LM11A-31) has been found to reduce basal fore- injury, reversed age-related cognitive dysfunc-
brain cholinergic degeneration and rescue tion, and restored electrophysiological currents
neurons from death in various rodent models of (Charles et al. 1996; Backman et al. 1996; Albeck
AD when administered orally (Yang et al. 2020a; et al. 1999). Follow-up studies showed that by
Xie et al. 2019; Nguyen et al. 2014). The com- using transferrin-avidin conjugation to bind
pound is currently tested in an ongoing clinical NGF-biotin conjugates, this could increase the
trial in mild-to-moderate AD patients NGF binding capacity of the transferrin protein
(ClinicalTrials.gov Identifier: NCT03069014). and delivered higher amounts of NGF to the brain
tissue when compared to NGF-anti-transferrin
Genetically Modified Cell-Mediated receptor antibody conjugates (Liao et al. 2001;
Delivery Another approach for NGF delivery to Li et al. 2000).
the brain was undertaken using ex-vivo autolo-
gous genetically modified monocytes, which Nanoparticle Conjugated Nanomaterial-
showed neuroprotection when injected directly mediated drug delivery to the brain has been an
ICVD (Zassler and Humpel 2006) or peripherally attractive option under various experimental
via the blood (Hohsfield et al. 2014). When conditions due to its flexibility and tissue
injected into the cerebral ventricles, the targeting capabilities (Singh et al. 2019). NGF
monocytes were found to survive for 7 days and protein can be delivered to the brain when
resulted in restored basal forebrain function. Sim- adsorbed on various nanoparticle materials and
ilarly, when genetically modified has been reported to enhance cognition and mem-
NGF-expressing monocytes were injected intra- ory in pharmacological models of injury in-vivo
venously, they were found to reach the brain (Kurakhmaeva et al. 2009). Recently nanocapsule
tissue and elevate cognitive outcome. In both structures have been developed which can deliver
intraventricular and parenteral deliveries, geneti- NGF to the rodent and primate brain when
cally modified monocytes were found to deliver injected intravenously (Xu et al. 2019). Further-
NGF without inducing inflammatory reactions. more, advanced nanocarriers which carry
NGF-secreting plasmids entrapped within a lipo-
Carrier-Mediated Delivery Due to the sensitiv- some vehicle passing the BBB by incorporating a
ity of the NGF protein by rapid degradation by transferrin ligand on the surface of the nanocarrier
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 179

have been developed. These liposome expected for studies in AD-relevant animal
nanocarriers have been shown to reduce Aβ levels models (Qu et al. 2020).
and revive synaptic proteins when injected intra-
venously in an AD mice model (Rodrigues et al. Incorporated Within Nerve Guidance
2020). Several other types of nanocarriers have Conduits Although conduits have yet not been
been tested in-vitro and showed improved reten- used in studies of treatment for dementia or AD,
tion and slower degradation of NGF when conju- this technology has been demonstrated to provide
gated to nanomaterials (Marcus et al. 2015; support and guidance for axonal growth in spinal
Razavi et al. 2019), but they are yet to be tested cord injury and peripheral nerve injury cases
in-vivo. (Piotrowicz and Shoichet 2006). Conduits are
3D structures which are coated internally with
Microspheres Mediated Microspheres are molecules (biological or chemical) which
entrapment procedures (much like nanoparticles) enhance the axonal growth. NGF has been used
but with better biocompatibility, mechanical flex- as the chemical que in several studies and shown
ibility and control of drug release kinetics (Varde to enhance guided axonal growth and improved
and Pack 2004). Initial work in the early 1990s nerve health (Liu et al. 2013; Xia and Lv 2018;
showed the ability of intracerebrally injected Hsu et al. 2019). Injectable hydrogels intended
NGF containing microspheres to constantly for stem cell applications in nerve regeneration
release its contents for more than 4 weeks have also been reported to enhance cell survival
(Camarata et al. 1992). Similar studies in later and proliferation when used in conjunction with
work demonstrated that although microspheres NGF (Li et al. 2020).
can be used for continuous controlled delivery
of NGF, the biological activity may abate due to Ultrasound-Guided Brain Delivery Although
the preparation step or due to protein denaturation this method is yet not used in assisting an
and degradation over time (Hadlock et al. 2003). NGF-carrying device or anything similar, ultra-
Newer versions of biodegradable microspheres sound waves have been used in conjunction with
using fibrin were recently reported to deliver other NGF delivery methods to enhance the brain
NGF when injected to the brain (Samal et al. penetration of NGF, when delivered peripherally.
2015). In-vivo studies in rat showed that Since peripherally injected neurotrophins cannot
peripherally administered liposomes can deliver cross the BBB, guided focused ultrasounds were
NGF to different brain regions including the stri- used to momentarily break the BBB in specific
atum, cortex, and basal forebrain (Xie et al. 2005; brain regions, thus facilitating drug uptake. This
Gu et al. 2007), and can recover spatial learning approach of using ultrasound-guided delivery of
ability when implanted in fimbria-fornix transec- nano-/micro-particles is termed “sonochem-
tion models (Gu et al. 2009). Furthermore, an otherapy” (Lammertink et al. 2015). In a spinal
animal model of sciatic nerve injury demonstrated cord injury model, a NGF plasmid contained in
that poly lactic-co-glycolic acid (PLGA) PLGA nanobubbles was intravenously injected
microspheres containing NGF injected to the and focally disintegrated within the injured tissue
injury site can increase nerve recovery (Zhang to release the plasmid using guided ultrasound
et al. 2017). A recent study using exosomes to which led to neuroprotection (Song et al. 2017).
simultaneously pack NGF protein and NGF Safety and tolerability of focused ultrasound-
expressing vectors together and inject them sys- guided BBB opening and drug delivery have
temically in a stroke model showed beneficial recently been discussed as a promising technol-
effects on neuronal cell survival and inflamma- ogy for delivering peripherally administered
tory reactions (Yang et al. 2020b). With new drugs to specific brain regions in AD patients
technologies at the horizon, the development of (Rezai et al. 2020; Lipsman et al. 2018). Efficacy
targeted viable in-vivo delivery of NGF is eagerly of an intravenously administered TrkA agonist
180 S. Mitra et al.

Delivery to brain tissue


Parenteral
Delivery to CSF

Parenteral
ICVD
Parenteral with targeted
possibilities
Stereotactic injection
Encapsulated
Genetically modified
Viral vectors
Stem cell Intraocular
Intracerebral &
Intranasal

Focused
guided
ultrasound
+
Parenteral
drug

Fig. 11.3 Brain targeting capabilities of NGF delivery administration route of each technique and provides an
methods: Although NGF can be administered by various overall view of invasiveness of the technique and its
techniques into different regions of the body, but the brain brain targeting capability. Different administration routes
availability of NGF, post-administration, defines the effi- are color coded. Abbreviations: ICVD (intracerebroven-
cacy of these methods. This figure highlights the initial tricular), CSF (cerebrospinal fluid)

(D3) was delivered to the basal forebrain using administration, whereas data on long-term safety
focused ultrasound and shown to enhance cholin- and efficacy of non-invasive procedures are still
ergic signaling and neurotransmission in a mice awaiting clinical testing. Painless NGF
model of AD (Xhima et al. 2020). equivalents and novel small molecule modulators
are promising for future therapies but need further
evaluation for off-target effects and safety, along
with the possibility to use multiple neurotrophin
11.3 Conclusion and Future
molecules in close connection or simultaneously.
Developments

Over the last three decades, several different


methods have been applied to deliver NGF to References
the brain, as illustrated in Fig. 11.3. The various
methods have evolved over time to meet the Ager RR, Davis JL, Agazaryan A, Benavente F, Poon
WW, LaFerla FM, Blurton-Jones M (2015) Human
medical needs of clinical efficacy, tolerability, neural stem cells improve cognition and promote syn-
and safety, and to minimize off-target effects. aptic growth in two complementary transgenic models
Nonetheless, every delivery method has its pros of Alzheimer’s disease and neuronal loss. Hippocam-
and cons, which should be taken into consider- pus 25(7):813–826. https://doi.org/10.1002/hipo.
22405
ation when assessing their potential for clinical Albeck DS, Backman C, Veng L, Friden P, Rose GM,
application. Granholm A (1999) Acute application of NGF
Invasive techniques have been shown to have increases the firing rate of aged rat basal forebrain
potential for clinically relevant effects in AD neurons. Eur J Neurosci 11(7):2291–2304. https://doi.
org/10.1046/j.1460-9568.1999.00644.x
patients (ECB-NGF, viral vectors, genetically Allard S, Leon WC, Pakavathkumar P, Bruno MA,
modified cells) but need high expertise for Ribeiro-da-Silva A, Cuello AC (2012) Impact of the
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 181

NGF maturation and degradation pathway on the cor- and their involvement in digestive cancers. Cell Death
tical cholinergic system phenotype. J Neurosci 32 Dis 10(2):123. https://doi.org/10.1038/s41419-019-
(6):2002–2012. https://doi.org/10.1523/JNEUROSCI. 1385-8
1144-11.2012 Bonini S, Lambiase A, Rama P, Caprioglio G, Aloe L
Aloe L, Rocco ML, Bianchi P, Manni L (2012) Nerve (2000) Topical treatment with nerve growth factor for
growth factor: from the early discoveries to the poten- neurotrophic keratitis. Ophthalmology 107
tial clinical use. J Transl Med 10:239. https://doi.org/ (7):1347–1351.; Discussion 1351–1342. https://doi.
10.1186/1479-5876-10-239 org/10.1016/s0161-6420(00)00163-9
Aloe L, Rocco ML, Balzamino BO, Micera A (2015) Bourganis V, Kammona O, Alexopoulos A, Kiparissides
Nerve growth factor: a focus on neuroscience and C (2018) Recent advances in carrier mediated nose-to-
therapy. Curr Neuropharmacol 13(3):294–303. brain delivery of pharmaceutics. Eur J Pharm
h t t p s : / / d o i . o r g / 1 0 . 2 1 7 4 / Biopharm 128:337–362. https://doi.org/10.1016/j.
1570159x13666150403231920 ejpb.2018.05.009
Aloe L, Rocco ML, Balzamino BO, Micera A (2016) Bracci-Laudiero L, Manni L (2014) NGF and immune
Nerve growth factor: role in growth, differentiation regulation. In: Kostrzewa RM (ed) Handbook of neu-
and controlling cancer cell development. J Exp Clin rotoxicity. Springer, New York
Cancer Res 35(1):116. https://doi.org/10.1186/s13046- Bradshaw RA, Mobley W, Rush RA (2017) Nerve growth
016-0395-y factor and related substances: a brief history and an
Altar CA, Bakhit C (1991) Receptor-mediated transport of introduction to the international NGF meeting series.
human recombinant nerve growth factor from olfactory Int J Mol Sci 18(6):1143. https://doi.org/10.3390/
bulb to forebrain cholinergic nuclei. Brain Res 541 ijms18061143
(1):82–88. https://doi.org/10.1016/0006-8993(91) Bruno MA, Cuello AC (2006) Activity-dependent release
91077-e of precursor nerve growth factor, conversion to mature
Backman C, Rose GM, Hoffer BJ, Henry MA, Bartus RT, nerve growth factor, and its degradation by a protease
Friden P, Granholm AC (1996) Systemic administra- cascade. Proc Natl Acad Sci U S A 103
tion of a nerve growth factor conjugate reverses (17):6735–6740. https://doi.org/10.1073/pnas.
age-related cognitive dysfunction and prevents cholin- 0510645103
ergic neuron atrophy. J Neurosci 16(17):5437–5442 Cairns DM, Rouleau N, Parker RN, Walsh KG, Gehrke L,
Balin BJ, Hudson AP (2018) Herpes viruses and Kaplan DL (2020) A 3D human brain-like tissue model
Alzheimer’s disease: new evidence in the debate. Lan- of herpes-induced Alzheimer’s disease. Sci Adv 6(19):
cet Neurol 17(10):839–841. https://doi.org/10.1016/ eaay8828. https://doi.org/10.1126/sciadv.aay8828
S1474-4422(18)30316-8 Camarata PJ, Suryanarayanan R, Turner DA, Parker RG,
Bartus RT, Dean RL 3rd, Beer B, Lippa AS (1982) The Ebner TJ (1992) Sustained release of nerve growth
cholinergic hypothesis of geriatric memory dysfunc- factor from biodegradable polymer microspheres. Neu-
tion. Science 217(4558):408–414 rosurgery 30(3):313–319. https://doi.org/10.1227/
Bernabeu-Zornoza A, Coronel R, Palmer C, 00006123-199203000-00001
Monteagudo M, Zambrano A, Liste I (2019) Physio- Capsoni S, Giannotta S, Cattaneo A (2002) Nerve growth
logical and pathological effects of amyloid-beta spe- factor and galantamine ameliorate early signs of
cies in neural stem cell biology. Neural Regen Res 14 neurodegeneration in anti-nerve growth factor mice.
(12):2035–2042. https://doi.org/10.4103/1673-5374. Proc Natl Acad Sci U S A 99(19):12432–12437.
262571 https://doi.org/10.1073/pnas.192442999
Binder N, Balmford J, Schumacher M (2019) A multi-state Capsoni S, Covaceuszach S, Ugolini G, Spirito F,
model based reanalysis of the Framingham heart study: Vignone D, Stefanini B, Amato G, Cattaneo A (2009)
is dementia incidence really declining? Eur J Delivery of NGF to the brain: intranasal versus ocular
Epidemiol 34(11):1075–1083. https://doi.org/10. administration in anti-NGF transgenic mice. J
1007/s10654-019-00567-6 Alzheimers Dis 16(2):371–388. https://doi.org/10.
Birks JS, Harvey RJ (2018) Donepezil for dementia due to 3233/JAD-2009-0953
Alzheimer’s disease. Cochrane Database Syst Rev 6: Capsoni S, Brandi R, Arisi I, D’Onofrio M, Cattaneo A
CD001190. https://doi.org/10.1002/14651858. (2011) A dual mechanism linking NGF/proNGF
CD001190.pub3 imbalance and early inflammation to Alzheimer’s dis-
Bishop KM, Hofer EK, Mehta A, Ramirez A, Sun L, ease neurodegeneration in the AD11 anti-NGF mouse
Tuszynski M, Bartus RT (2008) Therapeutic potential model. CNS Neurol Disord Drug Targets 10
of CERE-110 (AAV2-NGF): targeted, stable, and (5):635–647
sustained NGF delivery and trophic activity on rodent Capsoni S, Marinelli S, Ceci M, Vignone D, Amato G,
basal forebrain cholinergic neurons. Exp Neurol 211 Malerba F, Paoletti F, Meli G, Viegi A, Pavone F,
(2):574–584. https://doi.org/10.1016/j.expneurol. Cattaneo A (2012) Intranasal “painless” human nerve
2008.03.004 growth factor [corrected] slows amyloid
Blondy S, Christou N, David V, Verdier M, Jauberteau neurodegeneration and prevents memory deficits in
MO, Mathonnet M, Perraud A (2019) Neurotrophins
182 S. Mitra et al.

App X PS1 mice. PLoS One 7(5):e37555. https://doi. (11):1538–1547. https://doi.org/10.1080/02699052.


org/10.1371/journal.pone.0037555 2017.1376760
Capsoni S, Amato G, Vignone D, Criscuolo C, Nykjaer A, Cohen S, Levi-Montalcini R (1956) A nerve growth-
Cattaneo A (2013) Dissecting the role of sortilin recep- stimulating factor isolated from snake venom. Proc
tor signaling in neurodegeneration induced by NGF Natl Acad Sci U S A 42(9):571–574. https://doi.org/
deprivation. Biochem Biophys Res Commun 431 10.1073/pnas.42.9.571
(3):579–585. https://doi.org/10.1016/j.bbrc.2013.01. Cohen S, Levi-Montalcini R, Hamburger V (1954) A
007 nerve growth-stimulating factor isolated from sarcom
Castle MJ, Baltanas FC, Kovacs I, Nagahara AH, Barba D, as 37 and 180. Proc Natl Acad Sci U S A 40
Tuszynski MH (2020) Postmortem analysis in a clini- (10):1014–1018. https://doi.org/10.1073/pnas.40.10.
cal trial of AAV2-NGF gene therapy for Alzheimer’s 1014
disease identifies a need for improved vector delivery. Corvaglia V, Cilli D, Scopa C, Brandi R, Arisi I,
Hum Gene Ther 31(7-8):415–422. https://doi.org/10. Malerba F, La Regina F, Scardigli R, Cattaneo A
1089/hum.2019.367 (2019) ProNGF is a cell-type-specific mitogen for
Cattaneo A, Capsoni S (2019) Painless nerve growth fac- adult hippocampal and for induced neural stem cells.
tor: a TrkA biased agonist mediating a broad Stem Cells 37(9):1223–1237. https://doi.org/10.1002/
neuroprotection via its actions on microglia cells. stem.3037
Pharmacol Res 139:17–25. https://doi.org/10.1016/j. Covaceuszach S, Capsoni S, Ugolini G, Spirito F,
phrs.2018.10.028 Vignone D, Cattaneo A (2009) Development of a non
Cattaneo A, Capsoni S, Paoletti F (2008) Towards non invasive NGF-based therapy for Alzheimer’s disease.
invasive nerve growth factor therapies for Alzheimer’s Curr Alzheimer Res 6(2):158–170. https://doi.org/10.
disease. J Alzheimers Dis 15(2):255–283. https://doi. 2174/156720509787602870
org/10.3233/jad-2008-15210 Cuello AC, Pentz R, Hall H (2019) The brain NGF meta-
Chang DS, Hsu E, Hottinger DG, Cohen SP (2016) Anti- bolic pathway in health and in Alzheimer’s pathology.
nerve growth factor in pain management: current evi- Front Neurosci 13:62. https://doi.org/10.3389/fnins.
dence. J Pain Res 9:373–383. https://doi.org/10.2147/ 2019.00062
JPR.S89061 Cummings J, Feldman HH, Scheltens P (2019) The
Charles V, Mufson EJ, Friden PM, Bartus RT, Kordower “rights” of precision drug development for
JH (1996) Atrophy of cholinergic basal forebrain Alzheimer’s disease. Alzheimers Res Ther 11(1):76.
neurons following excitotoxic cortical lesions is https://doi.org/10.1186/s13195-019-0529-5
reversed by intravenous administration of an NGF Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K
conjugate. Brain Res 728(2):193–203. https://doi.org/ (2020) Alzheimer’s disease drug development pipe-
10.1016/0006-8993(96)00398-8 line: 2020. Alzheimers Dement (N Y) 6(1):e12050.
Chauhan MB, Chauhan NB (2015) Brain uptake of https://doi.org/10.1002/trc2.12050
Neurotherapeutics after intranasal versus intraperito- Date I, Ohmoto T, Imaoka T, Ono T, Hammang JP,
neal delivery in mice. J Neurol Neurosurg 2:1–9 Francis J, Greco C, Emerich DF (1996) Cografting
Chen XQ, Mobley WC (2019) Exploring the pathogenesis with polymer-encapsulated human nerve growth
of Alzheimer disease in basal forebrain cholinergic factor-secreting cells and chromaffin cell survival and
neurons: converging insights from alternative behavioral recovery in hemiparkinsonian rats. J
hypotheses. Front Neurosci 13:446. https://doi.org/10. Neurosurg 84(6):1006–1012. https://doi.org/10.3171/
3389/fnins.2019.00446 jns.1996.84.6.1006
Chen XQ, Fawcett JR, Rahman YE, Ala TA, Frey IW de Bellis A, de Bellis M, Aloe L (2018) Long-term non--
(1998) Delivery of nerve growth factor to the brain invasive treatment via intranasal administration of
via the olfactory pathway. J Alzheimers Dis 1 nerve growth factor protects the human brain in
(1):35–44. https://doi.org/10.3233/jad-1998-1102 frontotemporal dementia associated with corticobasal
Chiaretti A, Genovese O, Riccardi R, Di Rocco C, Di syndrome: a pilot study. J Alzheimers Dis Rep 2
Giuda D, Mariotti P, Pulitano S, Piastra M, (1):67–77. https://doi.org/10.3233/ADR-180055
Polidori G, Colafati GS, Aloe L (2005) Intraventricular de Boer R, Knight AM, Spinner RJ, Malessy MJ,
nerve growth factor infusion: a possible treatment for Yaszemski MJ, Windebank AJ (2010) In vitro and
neurological deficits following hypoxic-ischemic brain in vivo release of nerve growth factor from biodegrad-
injury in infants. Neurol Res 27(7):741–746. https:// able poly-lactic-co-glycolic-acid microspheres. J
doi.org/10.1179/016164105X35611 Biomed Mater Res A 95(4):1067–1073. https://doi.
Chiaretti A, Conti G, Falsini B, Buonsenso D, Crasti M, org/10.1002/jbm.a.32900
Manni L, Soligo M, Fantacci C, Genovese O, Calcagni De Luca M, Aiuti A, Cossu G, Parmar M, Pellegrini G,
ML, Di Giuda D, Mattoli MV, Cocciolillo F, Ferrara P, Robey PG (2019) Advances in stem cell research and
Ruggiero A, Staccioli S, Colafati GS, Riccardi R therapeutic development. Nat Cell Biol 21
(2017) Intranasal nerve growth factor administration (7):801–811. https://doi.org/10.1038/s41556-019-
improves cerebral functions in a child with severe 0344-z
traumatic brain injury: a case report. Brain Inj 31
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 183

Denk F, Bennett DL, McMahon SB (2017) Nerve growth survival in vitro. Exp Cell Res 371(1):175–184.
factor and pain mechanisms. Annu Rev Neurosci https://doi.org/10.1016/j.yexcr.2018.08.007
40:307–325. https://doi.org/10.1146/annurev-neuro- Eriksdotter-Jonhagen M, Linderoth B, Lind G,
072116-031121 Aladellie L, Almkvist O, Andreasen N, Blennow K,
De Rosa R, Garcia AA, Braschi C, Capsoni S, Maffei L, Bogdanovic N, Jelic V, Kadir A, Nordberg A,
Berardi N, Cattaneo A (2005) Intranasal administration Sundstrom E, Wahlund LO, Wall A, Wiberg M,
of nerve growth factor (NGF) rescues recognition Winblad B, Seiger A, Almqvist P, Wahlberg L
memory deficits in AD11 anti-NGF transgenic mice. (2012) Encapsulated cell biodelivery of nerve growth
Proc Natl Acad Sci U S A 102(10):3811–3816. https:// factor to the Basal forebrain in patients with
doi.org/10.1073/pnas.0500195102 Alzheimer’s disease. Dement Geriatr Cogn Disord 33
Dou KX, Tan MS, Tan CC, Cao XP, Hou XH, Guo QH, (1):18–28. https://doi.org/10.1159/000336051
Tan L, Mok V, Yu JT (2018) Comparative safety and Ernfors P, Ebendal T, Olson L, Mouton P, Stromberg I,
effectiveness of cholinesterase inhibitors and Persson H (1989) A cell line producing recombinant
memantine for Alzheimer’s disease: a network meta- nerve growth factor evokes growth responses in intrin-
analysis of 41 randomized controlled trials. Alzheimers sic and grafted central cholinergic neurons. Proc Natl
Res Ther 10(1):126. https://doi.org/10.1186/s13195- Acad Sci U S A 86(12):4756–4760. https://doi.org/10.
018-0457-9 1073/pnas.86.12.4756
Elia CA, Tamborini M, Rasile M, Desiato G, Marchetti S, Estenne-Bouhtou G, Kullander K, Karlsson M, Ebendal T,
Swuec P, Mazzitelli S, Clemente F, Anselmo A, Hacksell U, Luthman K (1996) Design, synthesis, tan-
Matteoli M, Malosio ML, Coco S (2019) Intracerebral dem mass spectrometric sequencing and biological
injection of extracellular vesicles from mesenchymal activity of NGF mimetics. Int J Pept Protein Res 48
stem cells exerts reduced Abeta plaque burden in early (4):337–346. https://doi.org/10.1111/j.1399-3011.
stages of a preclinical model of Alzheimer’s disease. 1996.tb00850.x
Cells 8(9):1059. https://doi.org/10.3390/cells8091059 Eyjolfsdottir H, Eriksdotter M, Linderoth B, Lind G,
Emerich DF, Hammang JP, Baetge EE, Winn SR (1994a) Juliusson B, Kusk P, Almkvist O, Andreasen N,
Implantation of polymer-encapsulated human nerve Blennow K, Ferreira D, Westman E, Nennesmo I,
growth factor-secreting fibroblasts attenuates the Karami A, Darreh-Shori T, Kadir A, Nordberg A,
behavioral and neuropathological consequences of Sundstrom E, Wahlund LO, Wall A, Wiberg M,
quinolinic acid injections into rodent striatum. Exp Winblad B, Seiger A, Wahlberg L, Almqvist P
Neurol 130(1):141–150. https://doi.org/10.1006/exnr. (2016) Targeted delivery of nerve growth factor to
1994.1193 the cholinergic basal forebrain of Alzheimer’s disease
Emerich DF, Winn SR, Harper J, Hammang JP, Baetge patients: application of a second-generation
EE, Kordower JH (1994b) Implants of polymer- encapsulated cell biodelivery device. Alzheimers Res
encapsulated human NGF-secreting cells in the nonhu- Ther 8(1):30. https://doi.org/10.1186/s13195-016-
man primate: rescue and sprouting of degenerating 0195-9
cholinergic basal forebrain neurons. J Comp Neurol Fahnestock M, Shekari A (2019) ProNGF and
349(1):148–164. https://doi.org/10.1002/cne. neurodegeneration in Alzheimer’s disease. Front
903490110 Neurosci 13:129. https://doi.org/10.3389/fnins.2019.
Emerich DF, Orive G, Thanos C, Tornoe J, Wahlberg LU 00129
(2014) Encapsulated cell therapy for neurodegenera- Fantacci C, Capozzi D, Ferrara P, Chiaretti A (2013)
tive diseases: from promise to product. Adv Drug Neuroprotective role of nerve growth factor in
Deliv Rev 67–68:131–141. https://doi.org/10.1016/j. hypoxic-ischemic brain injury. Brain Sci 3
addr.2013.07.008 (3):1013–1022. https://doi.org/10.3390/
Erdo F, Bors LA, Farkas D, Bajza A, Gizurarson S (2018) brainsci3031013
Evaluation of intranasal delivery route of drug admin- Ferreira D, Westman E, Eyjolfsdottir H, Almqvist P,
istration for brain targeting. Brain Res Bull Lind G, Linderoth B, Seiger A, Blennow K,
143:155–170. https://doi.org/10.1016/j.brainresbull. Karami A, Darreh-Shori T, Wiberg M, Simmons A,
2018.10.009 Wahlund LO, Wahlberg L, Eriksdotter M (2015) Brain
Eriksdotter Jonhagen M, Nordberg A, Amberla K, changes in Alzheimer’s disease patients with
Backman L, Ebendal T, Meyerson B, Olson L, Seiger, implanted encapsulated cells releasing nerve growth
Shigeta M, Theodorsson E, Viitanen M, Winblad B, factor. J Alzheimers Dis 43(3):1059–1072. https://doi.
Wahlund LO (1998) Intracerebroventricular infusion org/10.3233/JAD-141068
of nerve growth factor in three patients with Fine A, Dunnett SB, Bjorklund A, Iversen SD (1985)
Alzheimer’s disease. Dement Geriatr Cogn Disord Cholinergic ventral forebrain grafts into the neocortex
9 (5):246–257. https://doi.org/10.1159/000017069 improve passive avoidance memory in a rat model of
Eriksdotter M, Navarro-Oviedo M, Mitra S, Wahlberg L, Alzheimer disease. Proc Natl Acad Sci U S A 82
Linderoth B, Tjernberg LO, Behbahani H (2018) Cere- (15):5227–5230. https://doi.org/10.1073/pnas.82.15.
brospinal fluid from Alzheimer patients affects cell- 5227
mediated nerve growth factor production and cell
184 S. Mitra et al.

Fischer W, Wictorin K, Bjorklund A, Williams LR, Neurosci Lett 453(3):204–209. https://doi.org/10.


Varon S, Gage FH (1987) Amelioration of cholinergic 1016/j.neulet.2009.02.027
neuron atrophy and spatial memory impairment in Gu G, Zhang W, Li M, Ni J, Wang P (2015) Transplanta-
aged rats by nerve growth factor. Nature 329 tion of NSC-derived cholinergic neuron-like cells
(6134):65–68. https://doi.org/10.1038/329065a0 improves cognitive function in APP/PS1 transgenic
Fjord-Larsen L, Kusk P, Emerich DF, Thanos C, Torp M, mice. Neuroscience 291:81–92. https://doi.org/10.
Bintz B, Tornoe J, Johnsen AH, Wahlberg LU (2012) 1016/j.neuroscience.2015.01.073
Increased encapsulated cell biodelivery of nerve Guo L, Davis BM, Ravindran N, Galvao J, Kapoor N,
growth factor in the brain by transposon-mediated Haamedi N, Shamsher E, Luong V, Fico E, Cordeiro
gene transfer. Gene Ther 19(10):1010–1017. https:// MF (2020) Topical recombinant human Nerve growth
doi.org/10.1038/gt.2011.178 factor (rh-NGF) is neuroprotective to retinal ganglion
Friden PM, Walus LR, Watson P, Doctrow SR, Kozarich cells by targeting secondary degeneration. Sci Rep 10
JW, Backman C, Bergman H, Hoffer B, Bloom F, (1):3375. https://doi.org/10.1038/s41598-020-60427-2
Granholm AC (1993) Blood-brain barrier penetration Hadlock TA, Sheahan T, Cheney ML, Vacanti JP,
and in vivo activity of an NGF conjugate. Science 259 Sundback CA (2003) Biologic activity of nerve growth
(5093):373–377. https://doi.org/10.1126/science. factor slowly released from microspheres. J Reconstr
8420006 Microsurg 19(3):179–184.; Discussion 185–176.
Ganger S, Schindowski K (2018) Tailoring formulations https://doi.org/10.1055/s-2003-39831
for intranasal nose-to-brain delivery: a review on archi- Hagg T, Manthorpe M, Vahlsing HL, Varon S (1988)
tecture, physico-chemical characteristics and Delayed treatment with nerve growth factor reverses
mucociliary clearance of the nasal olfactory mucosa. the apparent loss of cholinergic neurons after acute
Pharmaceutics 10(3):116. https://doi.org/10.3390/ brain damage. Exp Neurol 101(2):303–312. https://
pharmaceutics10030116 doi.org/10.1016/0014-4886(88)90013-1
Gomez D, Martinez JA, Hanson LR, Frey WH 2nd, Toth Hampel H, Mesulam MM, Cuello AC, Farlow MR,
CC (2012) Intranasal treatment of neurodegenerative Giacobini E, Grossberg GT, Khachaturian AS,
diseases and stroke. Front Biosci (Schol Ed) 4:74–89. Vergallo A, Cavedo E, Snyder PJ, Khachaturian ZS
https://doi.org/10.2741/252 (2018) The cholinergic system in the pathophysiology
Goswami R, Subramanian G, Silayeva L, Newkirk I, and treatment of Alzheimer’s disease. Brain 141
Doctor D, Chawla K, Chattopadhyay S, Chandra D, (7):1917–1933. https://doi.org/10.1093/brain/awy132
Chilukuri N, Betapudi V (2019) Gene therapy leaves a Hanzel CE, Iulita MF, Eyjolfsdottir H, Hjorth E,
vicious cycle. Front Oncol 9:297. https://doi.org/10. Schultzberg M, Eriksdotter M, Cuello AC (2014)
3389/fonc.2019.00297 Analysis of matrix metallo-proteases and the plasmin-
Granholm AC, Backman C, Bloom F, Ebendal T, Gerhardt ogen system in mild cognitive impairment and
GA, Hoffer B, Mackerlova L, Olson L, Soderstrom S, Alzheimer’s disease cerebrospinal fluid. J Alzheimers
Walus LR et al (1994) NGF and anti-transferrin recep- Dis 40(3):667–678. https://doi.org/10.3233/JAD-
tor antibody conjugate: short and long-term effects on 132282
survival of cholinergic neurons in intraocular septal Hao J, Ebendal T, Xu X, Wiesenfeld-Hallin Z, Eriksdotter
transplants. J Pharmacol Exp Ther 268(1):448–459 Jonhagen M (2000) Intracerebroventricular infusion of
Griffin N, Faulkner S, Jobling P, Hondermarck H (2018) nerve growth factor induces pain-like response in rats.
Targeting neurotrophin signaling in cancer: the renais- Neurosci Lett 286(3):208–212. https://doi.org/10.
sance. Pharmacol Res 135:12–17. https://doi.org/10. 1016/s0304-3940(00)01107-1
1016/j.phrs.2018.07.019 Hayakawa Y, Sakitani K, Konishi M, Asfaha S, Niikura R,
Grill RJ, Blesch A, Tuszynski MH (1997) Robust growth Tomita H, Renz BW, Tailor Y, Macchini M,
of chronically injured spinal cord axons induced by Middelhoff M, Jiang Z, Tanaka T, Dubeykovskaya
grafts of genetically modified NGF-secreting cells. ZA, Kim W, Chen X, Urbanska AM, Nagar K,
Exp Neurol 148(2):444–452. https://doi.org/10.1006/ Westphalen CB, Quante M, Lin CS, Gershon MD,
exnr.1997.6704 Hara A, Zhao CM, Chen D, Worthley DL, Koike K,
Gu H, Song C, Long D, Mei L, Sun H (2007) Controlled Wang TC (2017) Nerve growth factor promotes gastric
release of recombinant human nerve growth factor tumorigenesis through aberrant cholinergic signaling.
(rhNGF) from poly[(lactic acid)-co-(glycolic acid)] Cancer Cell 31(1):21–34. https://doi.org/10.1016/j.
microspheres for the treatment of neurodegenerative ccell.2016.11.005
disorders. Polym Int 56(10):1272–1280. https://doi. Hayashi Y, Lin HT, Lee CC, Tsai KJ (2020) Effects of
org/10.1002/pi.2272 neural stem cell transplantation in Alzheimer’s disease
Gu H, Long D, Song C, Li X (2009) Recombinant human models. J Biomed Sci 27(1):29. https://doi.org/10.
NGF-loaded microspheres promote survival of basal 1186/s12929-020-0622-x
forebrain cholinergic neurons and improve memory Hefti F (1986) Nerve growth factor promotes survival of
impairments of spatial learning in the rat model of septal cholinergic neurons after fimbrial transections. J
Alzheimer’s disease with fimbria-fornix lesion. Neurosci 6(8):2155–2162
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 185

Hefti F, Schneider LS (1989) Rationale for the planned vi279–vi279. https://doi.org/10.1093/neuonc/noz175.


clinical trials with nerve growth factor in Alzheimer’s 1171
disease. Psychiatr Dev 7(4):297–315 Kim HJ, Lee JH, Kim SH (2010) Therapeutic effects of
Hoffman D, Wahlberg L, Aebischer P (1990) NGF human mesenchymal stem cells on traumatic brain
released from a polymer matrix prevents loss of injury in rats: secretion of neurotrophic factors and
ChAT expression in basal forebrain neurons following inhibition of apoptosis. J Neurotrauma 27
a fimbria-fornix lesion. Exp Neurol 110(1):39–44. (1):131–138. https://doi.org/10.1089/neu.2008-0818
https://doi.org/10.1016/0014-4886(90)90049-x Kim KY, Suh YH, Chang KA (2020) Therapeutic effects
Hohsfield LA, Ehrlich D, Humpel C (2014) Intravenous of human amniotic epithelial stem cells in a transgenic
infusion of nerve growth factor-secreting monocytes mouse model of Alzheimer’s disease. Int J Mol Sci 21
supports the survival of cholinergic neurons in the (7). https://doi.org/10.3390/ijms21072658
nucleus basalis of Meynert in hypercholesterolemia Koliatsos VE, Nauta HJ, Clatterbuck RE, Holtzman DM,
Brown-Norway rats. J Neurosci Res 92(3):298–306. Mobley WC, Price DL (1990) Mouse nerve growth
https://doi.org/10.1002/jnr.23309 factor prevents degeneration of axotomized basal fore-
Hsu RS, Chen PY, Fang JH, Chen YY, Chang CW, Lu YJ, brain cholinergic neurons in the monkey. J Neurosci 10
Hu SH (2019) Adaptable microporous hydrogels of (12):3801–3813
propagating NGF-gradient by injectable building Koliatsos VE, Applegate MD, Knusel B, Junard EO,
blocks for accelerated axonal outgrowth. Adv Sci Burton LE, Mobley WC, Hefti FF, Price DL (1991a)
(Weinh) 6(16):1900520. https://doi.org/10.1002/advs. Recombinant human nerve growth factor prevents ret-
201900520 rograde degeneration of axotomized basal forebrain
Hu X, Li R, Wu Y, Li Y, Zhong X, Zhang G, Kang Y, cholinergic neurons in the rat. Exp Neurol 112
Liu S, Xie L, Ye J, Xiao J (2020) Thermosensitive (2):161–173. https://doi.org/10.1016/0014-4886(91)
heparin-poloxamer hydrogel encapsulated bFGF and 90066-l
NGF to treat spinal cord injury. J Cell Mol Med 24 Koliatsos VE, Clatterbuck RE, Nauta HJ, Knusel B,
(14):8166–8178. https://doi.org/10.1111/jcmm.15478 Burton LE, Hefti FF, Mobley WC, Price DL (1991b)
Isaacson LG, Saffran BN, Crutcher KA (1990) Intracere- Human nerve growth factor prevents degeneration of
bral NGF infusion induces hyperinnervation of cere- basal forebrain cholinergic neurons in primates. Ann
bral blood vessels. Neurobiol Aging 11(1):51–55. Neurol 30(6):831–840. https://doi.org/10.1002/ana.
https://doi.org/10.1016/0197-4580(90)90062-5 410300613
Isaacson LG, Saffran BN, Crutcher KA (1992) Nerve Kordower JH, Winn SR, Liu YT, Mufson EJ, Sladek JR Jr,
growth factor-induced sprouting of mature, uninjured Hammang JP, Baetge EE, Emerich DF (1994) The
sympathetic axons. J Comp Neurol 326(3):327–336. aged monkey basal forebrain: rescue and sprouting of
https://doi.org/10.1002/cne.903260302 axotomized basal forebrain neurons after grafts of
Jonhagen ME (2000) Nerve growth factor treatment in encapsulated cells secreting human nerve growth fac-
dementia. Alzheimer Dis Assoc Disord 14(Suppl 1): tor. Proc Natl Acad Sci U S A 91(23):10898–10902.
S31–S38. https://doi.org/10.1097/00002093- https://doi.org/10.1073/pnas.91.23.10898
200000001-00006 Kurakhmaeva KB, Djindjikhashvili IA, Petrov VE,
Kamei N, Tanaka N, Oishi Y, Hamasaki T, Nakanishi K, Balabanyan VU, Voronina TA, Trofimov SS,
Sakai N, Ochi M (2007) BDNF, NT-3, and NGF Kreuter J, Gelperina S, Begley D, Alyautdin RN
released from transplanted neural progenitor cells pro- (2009) Brain targeting of nerve growth factor using
mote corticospinal axon growth in organotypic poly(butyl cyanoacrylate) nanoparticles. J Drug Target
cocultures. Spine (Phila Pa 1976) 32(12):1272–1278. 17(8):564–574. https://doi.org/10.1080/
https://doi.org/10.1097/BRS.0b013e318059afab 10611860903112842
Karami A, Eyjolfsdottir H, Vijayaraghavan S, Lind G, Kwak YD, Brannen CL, Qu T, Kim HM, Dong X, Soba P,
Almqvist P, Kadir A, Linderoth B, Andreasen N, Majumdar A, Kaplan A, Beyreuther K, Sugaya K
Blennow K, Wall A, Westman E, Ferreira D, (2006) Amyloid precursor protein regulates differenti-
Kristoffersen Wiberg M, Wahlund LO, Seiger A, ation of human neural stem cells. Stem Cells Dev 15
Nordberg A, Wahlberg L, Darreh-Shori T, Eriksdotter (3):381–389. https://doi.org/10.1089/scd.2006.15.381
M (2015) Changes in CSF cholinergic biomarkers in Lambiase A, Rama P, Bonini S, Caprioglio G, Aloe L
response to cell therapy with NGF in patients with (1998) Topical treatment with nerve growth factor for
Alzheimer’s disease. Alzheimers Dement 11 corneal neurotrophic ulcers. N Engl J Med 338
(11):1316–1328. https://doi.org/10.1016/j.jalz.2014. (17):1174–1180. https://doi.org/10.1056/
11.008 NEJM199804233381702
Killick-Cole C, Woolley M, Johnson D, Lewis O, Lambiase A, Tirassa P, Micera A, Aloe L, Bonini S (2005)
Moore P, Fletcher J, Skinner P, Bienemann A, Gill S Pharmacokinetics of conjunctivally applied nerve
(2019) SCIDOT-35. A novel model for the optimiza- growth factor in the retina and optic nerve of adult
tion of drug-device combinations for the treatment of rats. Invest Ophthalmol Vis Sci 46(10):3800–3806.
brain tumors. Neuro-Oncology 21(Supplement_6): https://doi.org/10.1167/iovs.05-0301
186 S. Mitra et al.

Lambiase A, Pagani L, Di Fausto V, Sposato V, Lim F, Sun AM (1980) Microencapsulated islets as


Coassin M, Bonini S, Aloe L (2007a) Nerve growth bioartificial endocrine pancreas. Science 210
factor eye drop administrated on the ocular surface of (4472):908–910. https://doi.org/10.1126/science.
rodents affects the nucleus basalis and septum: bio- 6776628
chemical and structural evidence. Brain Res 1127 Lipsman N, Meng Y, Bethune AJ, Huang Y, Lam B,
(1):45–51. https://doi.org/10.1016/j.brainres.2006.09. Masellis M, Herrmann N, Heyn C, Aubert I,
102 Boutet A, Smith GS, Hynynen K, Black SE (2018)
Lambiase A, Coassin M, Sposato V, Micera A, Blood-brain barrier opening in Alzheimer’s disease
Sacchetti M, Bonini S, Aloe L (2007b) NGF topical using MR-guided focused ultrasound. Nat Commun 9
application in patients with corneal ulcer does not (1):2336. https://doi.org/10.1038/s41467-018-04529-6
generate circulating NGF antibodies. Pharmacol Res Liu H, Wen W, Hu M, Bi W, Chen L, Liu S, Chen P, Tan
56(1):65–69. https://doi.org/10.1016/j.phrs.2007.03. X (2013) Chitosan conduits combined with nerve
007 growth factor microspheres repair facial nerve defects.
Lambiase A, Mantelli F, Sacchetti M, Rossi S, Aloe L, Neural Regen Res 8(33):3139–3147. https://doi.org/
Bonini S (2011) Clinical applications of NGF in ocular 10.3969/j.issn.1673-5374.2013.33.008
diseases. Arch Ital Biol 149(2):283–292. https://doi. Liu X, Li W, Fu X, Xu Y (2017) The immunogenicity and
org/10.4449/aib.v149i2.1363 immune tolerance of pluripotent stem cell derivatives.
Lammertink BH, Bos C, Deckers R, Storm G, Moonen Front Immunol 8:645. https://doi.org/10.3389/fimmu.
CT, Escoffre JM (2015) Sonochemotherapy: from 2017.00645
bench to bedside. Front Pharmacol 6:138. https://doi. Liu PP, Xie Y, Meng XY, Kang JS (2019) History and
org/10.3389/fphar.2015.00138 progress of hypotheses and clinical trials for
Lee AC, Yu VM, Lowe JB 3rd, Brenner MJ, Hunter DA, Alzheimer’s disease. Signal Transduct Target Ther
Mackinnon SE, Sakiyama-Elbert SE (2003) Controlled 4:29. https://doi.org/10.1038/s41392-019-0063-8
release of nerve growth factor enhances sciatic nerve Longo FM, Massa SM (2013) Small-molecule modulation
regeneration. Exp Neurol 184(1):295–303. https://doi. of neurotrophin receptors: a strategy for the treatment
org/10.1016/s0014-4886(03)00258-9 of neurological disease. Nat Rev Drug Discov 12
Lee HJ, Lee JK, Lee H, Carter JE, Chang JW, Oh W, Yang (7):507–525. https://doi.org/10.1038/nrd4024
YS, Suh JG, Lee BH, Jin HK, Bae JS (2012a) Human Longo FM, Manthorpe M, Xie YM, Varon S (1997) Syn-
umbilical cord blood-derived mesenchymal stem cells thetic NGF peptide derivatives prevent neuronal death
improve neuropathology and cognitive impairment in via a p75 receptor-dependent mechanism. J Neurosci
an Alzheimer’s disease mouse model through modula- Res 48(1):1–17. https://doi.org/10.1002/(sici)1097-
tion of neuroinflammation. Neurobiol Aging 33 4547(19970401)48:1<1::aid-jnr1>3.0.co;2-k
(3):588–602. https://doi.org/10.1016/j.neurobiolaging. Losurdo M, Pedrazzoli M, D’Agostino C, Elia CA,
2010.03.024 Massenzio F, Lonati E, Mauri M, Rizzi L, Molteni L,
Lee HJ, Lim IJ, Park SW, Kim YB, Ko Y, Kim SU Bresciani E, Dander E, D’Amico G, Bulbarelli A,
(2012b) Human neural stem cells genetically modified Torsello A, Matteoli M, Buffelli M, Coco S (2020)
to express human nerve growth factor (NGF) gene Intranasal delivery of mesenchymal stem cell-derived
restore cognition in the mouse with ibotenic acid- extracellular vesicles exerts immunomodulatory and
induced cognitive dysfunction. Cell Transplant 21 neuroprotective effects in a 3xTg model of
(11):2487–2496. https://doi.org/10.3727/ Alzheimer’s disease. Stem Cells Transl Med 9
096368912X638964 (9):1068–1084. https://doi.org/10.1002/sctm.19-0327
LeSauteur L, Wei L, Gibbs BF, Saragovi HU (1995) Small Machado A, Ferreira D, Grothe MJ, Eyjolfsdottir H,
peptide mimics of nerve growth factor bind TrkA Almqvist PM, Cavallin L, Lind G, Linderoth B,
receptors and affect biological responses. J Biol Seiger A, Teipel S, Wahlberg LU, Wahlund LO,
Chem 270(12):6564–6569. https://doi.org/10.1074/ Westman E, Eriksdotter M, Alzheimer’s Disease Neu-
jbc.270.12.6564 roimaging Initiative (2020) The cholinergic system in
Li XB, Liao GS, Shu YY, Tang SX (2000) Brain delivery subtypes of Alzheimer’s disease: an in vivo longitudi-
of biotinylated NGF bounded to an avidin-transferrin nal MRI study. Alzheimers Res Ther 12(1):51. https://
conjugate. J Nat Toxins 9(1):73–83 doi.org/10.1186/s13195-020-00620-7
Li W, Huang A, Zhong Y, Huang L, Yang J, Zhou C, Mahoney MJ, Saltzman WM (1999) Millimeter-scale
Zhou L, Zhang Y, Fu G (2020) Laminin-modified positioning of a nerve-growth-factor source and
gellan gum hydrogels loaded with the nerve growth biological activity in the brain. Proc Natl Acad Sci U
factor to enhance the proliferation and differentiation S A 96(8):4536–4539. https://doi.org/10.1073/pnas.
of neuronal stem cells. RSC Adv 10(29):17114–17122. 96.8.4536
https://doi.org/10.1039/D0RA01723J Mandal A, Pal D, Agrahari V, Trinh HM, Joseph M, Mitra
Liao GS, Li XB, Zhang CY, Shu YY, Tang SX (2001) AK (2018) Ocular delivery of proteins and peptides:
Pharmacological actions of nerve growth factor- challenges and novel formulation approaches. Adv
transferrin conjugate on the central nervous system. J Drug Deliv Rev 126:67–95. https://doi.org/10.1016/j.
Nat Toxins 10(4):291–297 addr.2018.01.008
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 187

Marcus M, Skaat H, Alon N, Margel S, Shefi O (2015) Nguyen TV, Shen L, Vander Griend L, Quach LN,
NGF-conjugated iron oxide nanoparticles promote dif- Belichenko NP, Saw N, Yang T, Shamloo M, Wyss-
ferentiation and outgrowth of PC12 cells. Nanoscale 7 Coray T, Massa SM, Longo FM (2014) Small mole-
(3):1058–1066. https://doi.org/10.1039/c4nr05193a cule p75NTR ligands reduce pathological phosphory-
Martinez-Serrano A, Bjorklund A (1998) Ex vivo nerve lation and misfolding of tau, inflammatory changes,
growth factor gene transfer to the basal forebrain in cholinergic degeneration, and cognitive deficits in
presymptomatic middle-aged rats prevents the devel- AbetaPP(L/S) transgenic mice. J Alzheimers Dis 42
opment of cholinergic neuron atrophy and cognitive (2):459–483. https://doi.org/10.3233/JAD-140036
impairment during aging. Proc Natl Acad Sci U S A 95 Oh B, Swaminathan V, Malkovskiy A, Santhanam S,
(4):1858–1863. https://doi.org/10.1073/pnas.95.4. McConnell K, George PM (2020) Single-cell encapsu-
1858 lation via click-chemistry alters production of para-
Martinez-Serrano A, Fischer W, Soderstrom S, Ebendal T, crine factors from neural progenitor cells. Adv Sci
Bjorklund A (1996) Long-term functional recovery (Weinh) 7(8):1902573. https://doi.org/10.1002/advs.
from age-induced spatial memory impairments by 201902573
nerve growth factor gene transfer to the rat basal fore- Olson L (1993) NGF and the treatment of Alzheimer’s
brain. Proc Natl Acad Sci U S A 93(13):6355–6360. disease. Exp Neurol 124(1):5–15. https://doi.org/10.
https://doi.org/10.1073/pnas.93.13.6355 1006/exnr.1993.1167
McGinley LM, Kashlan ON, Bruno ES, Chen KS, Hayes Olson L, Nordberg A, von Holst H, Backman L,
JM, Kashlan SR, Raykin J, Johe K, Murphy GG, Ebendal T, Alafuzoff I, Amberla K, Hartvig P,
Feldman EL (2018) Human neural stem cell transplan- Herlitz A, Lilja A et al (1992) Nerve growth factor
tation improves cognition in a murine model of affects 11C-nicotine binding, blood flow, EEG, and
Alzheimer’s disease. Sci Rep 8(1):14776. https://doi. verbal episodic memory in an Alzheimer patient (case
org/10.1038/s41598-018-33017-6 report). J Neural Transm Park Dis Dement Sect 4
Menei P, Pean JM, Nerriere-Daguin V, Jollivet C, (1):79–95. https://doi.org/10.1007/BF02257624
Brachet P, Benoit JP (2000) Intracerebral implantation Orive G, Santos E, Poncelet D, Hernandez RM, Pedraz JL,
of NGF-releasing biodegradable microspheres protects Wahlberg LU, De Vos P, Emerich D (2015) Cell
striatum against excitotoxic damage. Exp Neurol 161 encapsulation: technical and clinical advances. Trends
(1):259–272. https://doi.org/10.1006/exnr.1999.7253 Pharmacol Sci 36(8):537–546. https://doi.org/10.1016/
Minde J, Toolanen G, Andersson T, Nennesmo I, Remahl j.tips.2015.05.003
IN, Svensson O, Solders G (2004) Familial insensitiv- Orive G, Santos-Vizcaino E, Pedraz JL, Hernandez RM,
ity to pain (HSAN V) and a mutation in the NGFB Vela Ramirez JE, Dolatshahi-Pirouz A,
gene. A neurophysiological and pathological study. Khademhosseini A, Peppas NA, Emerich DF (2019)
Muscle Nerve 30(6):752–760. https://doi.org/10. 3D cell-laden polymers to release bioactive products in
1002/mus.20172 the eye. Prog Retin Eye Res 68:67–82. https://doi.org/
Mitra S, Behbahani H, Eriksdotter M (2019) Innovative 10.1016/j.preteyeres.2018.10.002
therapy for Alzheimer’s disease-with focus on Pakzaban P, Geller AI, Isacson O (1994) Effect of exoge-
biodelivery of NGF. Front Neurosci 13:38. https:// nous nerve growth factor on neurotoxicity of and neu-
doi.org/10.3389/fnins.2019.00038 ronal gene delivery by a herpes simplex amplicon
Montero CN, Hefti F (1988) Rescue of lesioned septal vector in the rat brain. Hum Gene Ther 5(8):987–995.
cholinergic neurons by nerve growth factor: specificity https://doi.org/10.1089/hum.1994.5.8-987
and requirement for chronic treatment. J Neurosci 8 Paradisi M, Alviano F, Pirondi S, Lanzoni G,
(8):2986–2999 Fernandez M, Lizzo G, Giardino L, Giuliani A,
Montero CN, Hefti F (1989) Intraventricular nerve growth Costa R, Marchionni C, Bonsi L, Calza L (2014)
factor administration prevents lesion-induced loss of Human mesenchymal stem cells produce bioactive
septal cholinergic neurons in aging rats. Neurobiol neurotrophic factors: source, individual variability
Aging 10(6):739–743. https://doi.org/10.1016/0197- and differentiation issues. Int J Immunopathol
4580(89)90011-0 Pharmacol 27(3):391–402. https://doi.org/10.1177/
Mufson EJ, Conner JM, Kordower JH (1995) Nerve 039463201402700309
growth factor in Alzheimer’s disease: defective retro- Pardridge WM (2019) Blood-brain barrier and delivery of
grade transport to nucleus basalis. Neuroreport 6 protein and gene therapeutics to brain. Front Aging
(7):1063–1066. https://doi.org/10.1097/00001756- Neurosci 11:373. https://doi.org/10.3389/fnagi.2019.
199505090-00028 00373
Nagahara AH, Bernot T, Moseanko R, Brignolo L, Park D, Yang YH, Bae DK, Lee SH, Yang G, Kyung J,
Blesch A, Conner JM, Ramirez A, Gasmi M, Kim D, Choi EK, Lee SW, Kim GH, Hong JT, Choi
Tuszynski MH (2009) Long-term reversal of choliner- KC, Lee HJ, Kim SU, Kim YB (2013) Improvement of
gic neuronal decline in aged non-human primates by cognitive function and physical activity of aging mice
lentiviral NGF gene delivery. Exp Neurol 215 by human neural stem cells over-expressing choline
(1):153–159. https://doi.org/10.1016/j.expneurol. acetyltransferase. Neurobiol Aging 34
2008.10.004
188 S. Mitra et al.

(11):2639–2646. https://doi.org/10.1016/j. Rocco ML, Soligo M, Manni L, Aloe L (2018) Nerve


neurobiolaging.2013.04.026 growth factor: early studies and recent clinical trials.
Pentz R, Iulita MF, Ducatenzeiler A, Bennett DA, Cuello Curr Neuropharmacol 16(10):1455–1465. https://doi.
AC (2020) The human brain NGF metabolic pathway org/10.2174/1570159X16666180412092859
is impaired in the pre-clinical and clinical continuum of Rodrigues BDS, Kanekiyo T, Singh J (2020) Nerve
Alzheimers disease. Mol Psychiatry. https://doi.org/10. growth factor gene delivery across the blood-brain
1038/s41380-020-0797-2 barrier to reduce beta amyloid accumulation in AD
Piotrowicz A, Shoichet MS (2006) Nerve guidance mice. Mol Pharm 17(6):2054–2063. https://doi.org/
channels as drug delivery vehicles. Biomaterials 27 10.1021/acs.molpharmaceut.0c00218
(9):2018–2027. https://doi.org/10.1016/j.biomaterials. Ruozi B, Belletti D, Bondioli L, De Vita A, Forni F,
2005.09.042 Vandelli MA, Tosi G (2012) Neurotrophic factors
Polak JM, Mantalaris S (2008) Stem cells bioprocessing: and neurodegenerative diseases: a delivery issue. Int
an important milestone to move regenerative medicine Rev Neurobiol 102:207–247. https://doi.org/10.1016/
research into the clinical arena. Pediatr Res 63 B978-0-12-386986-9.00009-0
(5):461–466. https://doi.org/10.1203/10.1203/PDR. Saffran BN, Woo JE, Mobley WC, Crutcher KA (1989)
0b013e31816a8c1c Intraventricular NGF infusion in the mature rat brain
Powell EM, Sobarzo MR, Saltzman WM (1990) Con- enhances sympathetic innervation of cerebrovascular
trolled release of nerve growth factor from a polymeric targets but fails to elicit sympathetic ingrowth. Brain
implant. Brain Res 515(1–2):309–311. https://doi.org/ Res 492(1-2):245–254. https://doi.org/10.1016/0006-
10.1016/0006-8993(90)90612-f 8993(89)90907-4
Pramanik S, Sulistio YA, Heese K (2017) Neurotrophin Sakiyama-Elbert SE, Hubbell JA (2000) Controlled
signaling and stem cells-implications for neurodegen- release of nerve growth factor from a heparin-
erative diseases and stem cell therapy. Mol Neurobiol containing fibrin-based cell ingrowth matrix. J Control
54(9):7401–7459. https://doi.org/10.1007/s12035- Release 69(1):149–158. https://doi.org/10.1016/
016-0214-7 s0168-3659(00)00296-0
Qu M, Jiang X, Zhou X, Wang C, Wu Q, Ren L, Zhu J, Sakiyama-Elbert SE, Panitch A, Hubbell JA (2001) Devel-
Zhu S, Tebon P, Sun W, Khademhosseini A (2020) opment of growth factor fusion proteins for cell-
Stimuli-responsive delivery of growth factors for tissue triggered drug delivery. FASEB J 15(7):1300–1302.
engineering. Adv Healthc Mater 9(7):e1901714. https://doi.org/10.1096/fj.00-0564fje
https://doi.org/10.1002/adhm.201901714 Saltzman WM, Mak MW, Mahoney MJ, Duenas ET,
Rafii MS, Baumann TL, Bakay RA, Ostrove JM, Siffert J, Cleland JL (1999) Intracranial delivery of recombinant
Fleisher AS, Herzog CD, Barba D, Pay M, Salmon DP, nerve growth factor: release kinetics and protein distri-
Chu Y, Kordower JH, Bishop K, Keator D, Potkin S, bution for three delivery systems. Pharm Res 16
Bartus RT (2014) A phase1 study of stereotactic gene (2):232–240. https://doi.org/10.1023/
delivery of AAV2-NGF for Alzheimer’s disease. a:1018824324275
Alzheimers Dement 10(5):571–581. https://doi.org/ Samal J, Hoban DB, Naughton C, Concannon R, Dowd E,
10.1016/j.jalz.2013.09.004 Pandit A (2015) Fibrin-based microsphere reservoirs
Rafii MS, Tuszynski MH, Thomas RG, Barba D, Brewer for delivery of neurotrophic factors to the brain.
JB, Rissman RA, Siffert J, Aisen PS, Team ANS Nanomedicine (Lond) 10(5):765–783. https://doi.org/
(2018) Adeno-associated viral vector (serotype 2)- 10.2217/nnm.14.221
nerve growth factor for patients with Alzheimer dis- Satizabal C, Beiser AS, Seshadri S (2016) Incidence of
ease: a randomized clinical trial. JAMA Neurol 75 dementia over three decades in the Framingham heart
(7):834–841. https://doi.org/10.1001/jamaneurol. study. N Engl J Med 375(1):93–94. https://doi.org/10.
2018.0233 1056/NEJMc1604823
Razavi S, Seyedebrahimi R, Jahromi M (2019) Scheltens P, Blennow K, Breteler MM, de Strooper B,
Biodelivery of nerve growth factor and gold Frisoni GB, Salloway S, Van der Flier WM (2016)
nanoparticles encapsulated in chitosan nanoparticles Alzheimer’s disease. Lancet 388(10043):505–517.
for schwann-like cells differentiation of human https://doi.org/10.1016/S0140-6736(15)01124-1
adipose-derived stem cells. Biochem Biophys Res Schlachetzki JC, Pizzo DP, Morrissette DA, Winkler J
Commun 513(3):681–687. https://doi.org/10.1016/j. (2014) Intracerebroventricular administration of nerve
bbrc.2019.03.189 growth factor induces gliogenesis in sensory ganglia,
Rezai AR, Ranjan M, D’Haese PF, Haut MW, Carpenter J, dorsal root, and within the dorsal root entry zone.
Najib U, Mehta RI, Chazen JL, Zibly Z, Yates JR, Biomed Res Int 2014:704259. https://doi.org/10.
Hodder SL, Kaplitt M (2020) Noninvasive hippocam- 1155/2014/704259
pal blood-brain barrier opening in Alzheimer’s disease Schneider L (2020) A resurrection of aducanumab for
with focused ultrasound. Proc Natl Acad Sci U S A 117 Alzheimer’s disease. Lancet Neurol 19(2):111–112.
(17):9180–9182. https://doi.org/10.1073/pnas. https://doi.org/10.1016/S1474-4422(19)30480-6
2002571117 Secnik J, Schwertner E, Alvarsson M, Hammar N,
Fastbom J, Winblad B, Garcia-Ptacek S, Religa D,
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 189

Eriksdotter M (2020) Cholinesterase inhibitors in support for axotomized cholinergic neurons. Prog
patients with diabetes mellitus and dementia: an Brain Res 78:401–407. https://doi.org/10.1016/s0079-
open-cohort study of ~23 000 patients from the Swed- 6123(08)60311-8
ish Dementia registry. BMJ Open Diabetes Res Care 8 Springer JE, Collier TJ, Sladek JR Jr, Loy R (1988b)
(1). https://doi.org/10.1136/bmjdrc-2019-000833 Transplantation of male mouse submaxillary gland
Seiger A, Nordberg A, von Holst H, Backman L, increases survival of axotomized basal forebrain
Ebendal T, Alafuzoff I, Amberla K, Hartvig P, neurons. J Neurosci Res 19(3):291–296. https://doi.
Herlitz A, Lilja A et al (1993) Intracranial infusion of org/10.1002/jnr.490190303
purified nerve growth factor to an Alzheimer patient: Sugaya K (2008) Mechanism of glial differentiation of
the first attempt of a possible future treatment strategy. neural progenitor cells by amyloid precursor protein.
Behav Brain Res 57(2):255–261. https://doi.org/10. Neurodegener Dis 5(3-4):170–172. https://doi.org/10.
1016/0166-4328(93)90141-c 1159/000113693
Simone MD, De Santis S, Vigneti E, Papa G, Amadori S, Sydow O, Hansson P, Young D, Meyerson B, Backlund
Aloe L (1999) Nerve growth factor: a survey of activity EO, Ebendal T, Farnebo LO, Freedman R,
on immune and hematopoietic cells. Hematol Oncol 17 Hamberger B, Hoffer B, Seiger A, Stromberq I,
(1):1–10. https://doi.org/10.1002/(sici)1099-1069 Olson L (1995) Long-term beneficial effects of adrenal
(199903)17:1<1::aid-hon635>3.0.co;2-l medullary autografts supported by nerve growth factor
Singh AP, Biswas A, Shukla A, Maiti P (2019) Targeted in Parkinson’s disease. Eur J Neurol 2(5):445–454.
therapy in chronic diseases using nanomaterial-based https://doi.org/10.1111/j.1468-1331.1995.tb00154.x
drug delivery vehicles. Signal Transduct Target Ther Thomas CE, Ehrhardt A, Kay MA (2003) Progress and
4:33. https://doi.org/10.1038/s41392-019-0068-3 problems with the use of viral vectors for gene therapy.
Skeldal S, Sykes AM, Glerup S, Matusica D, Palstra N, Nat Rev Genet 4(5):346–358. https://doi.org/10.1038/
Autio H, Boskovic Z, Madsen P, Castren E, Nykjaer A, nrg1066
Coulson EJ (2012) Mapping of the interaction site Tirassa P, Triaca V, Amendola T, Fiore M, Aloe L (2003)
between sortilin and the p75 neurotrophin receptor EGF and NGF injected into the brain of old mice
reveals a regulatory role for the sortilin intracellular enhance BDNF and ChAT in proliferating
domain in p75 neurotrophin receptor shedding and subventricular zone. J Neurosci Res 72(5):557–564.
apoptosis. J Biol Chem 287(52):43798–43809. https://doi.org/10.1002/jnr.10614
https://doi.org/10.1074/jbc.M112.374710 Tirassa P, Rosso P, Iannitelli A (2018) Ocular nerve
Sochocka M, Zwolinska K, Leszek J (2017) The infectious growth factor (NGF) and NGF eye drop application
etiology of Alzheimer’s disease. Curr Neuropharmacol as paradigms to investigate NGF neuroprotective and
15(7):996–1009. https://doi.org/10.2174/ reparative actions. Methods Mol Biol 1727:19–38.
1570159X15666170313122937 https://doi.org/10.1007/978-1-4939-7571-6_2
Son AI, Opfermann JD, McCue C, Ziobro J, Abrahams JH Tresco PA, Winn SR, Aebischer P (1992) Polymer
3rd, Jones K, Morton PD, Ishii S, Oluigbo C, encapsulated neurotransmitter secreting cells. Potential
Krieger A, Liu JS, Hashimoto-Torii K, Torii M treatment for Parkinson’s disease. ASAIO J 38
(2017) An implantable micro-caged device for direct (1):17–23
local delivery of agents. Sci Rep 7(1):17624. https:// Tuszynski MH (2000) Intraparenchymal NGF infusions
doi.org/10.1038/s41598-017-17912-y rescue degenerating cholinergic neurons. Cell Trans-
Song B, Song J, Zhang S, Anderson MA, Ao Y, Yang CY, plant 9(5):629–636. https://doi.org/10.1177/
Deming TJ, Sofroniew MV (2012) Sustained local 096368970000900508
delivery of bioactive nerve growth factor in the central Tuszynski MH, Gage FH (1995) Bridging grafts and tran-
nervous system via tunable diblock copolypeptide sient nerve growth factor infusions promote long-term
hydrogel depots. Biomaterials 33(35):9105–9116. central nervous system neuronal rescue and partial
https://doi.org/10.1016/j.biomaterials.2012.08.060 functional recovery. Proc Natl Acad Sci U S A 92
Song Z, Wang Z, Shen J, Xu S, Hu Z (2017) Nerve growth (10):4621–4625. https://doi.org/10.1073/pnas.92.10.
factor delivery by ultrasound-mediated nanobubble 4621
destruction as a treatment for acute spinal cord injury Tuszynski MH, U HS, Amaral DG, Gage FH (1990) Nerve
in rats. Int J Nanomed 12:1717–1729. https://doi.org/ growth factor infusion in the primate brain reduces
10.2147/IJN.S128848 lesion-induced cholinergic neuronal degeneration. J
Spiegel K, Agrafiotis D, Caprathe B, Davis RE, Dickerson Neurosci 10(11):3604–3614
MR, Fergus JH, Hepburn TW, Marks JS, Van Dorf M, Tuszynski MH, Roberts J, Senut MC, U HS, Gage FH
Wieland DM et al (1995) PD 90780, a non peptide (1996) Gene therapy in the adult primate brain:
inhibitor of nerve growth factor’s binding to the P75 intraparenchymal grafts of cells genetically modified
NGF receptor. Biochem Biophys Res Commun 217 to produce nerve growth factor prevent cholinergic
(2):488–494. https://doi.org/10.1006/bbrc.1995.2802 neuronal degeneration. Gene Ther 3(4):305–314
Springer JE, Collier TJ, Notter MF, Loy R, Sladek JR Jr Tuszynski MH, Thal L, Pay M, Salmon DP, U HS,
(1988a) Central nervous system grafts of nerve growth Bakay R, Patel P, Blesch A, Vahlsing HL, Ho G,
factor-rich tissue as an alternative source of trophic Tong G, Potkin SG, Fallon J, Hansen L, Mufson EJ,
190 S. Mitra et al.

Kordower JH, Gall C, Conner J (2005) A phase 1 clini- delivery protects septal cholinergic neurons following
cal trial of nerve growth factor gene therapy for axotomy. Brain Res 1061(2):107–113. https://doi.org/
Alzheimer disease. Nat Med 11(5):551–555. https:// 10.1016/j.brainres.2005.08.056
doi.org/10.1038/nm1239 Xhima K, Markham-Coultes K, Nedev H, Heinen S,
Tuszynski MH, Yang JH, Barba D, U HS, Bakay RA, Pay Saragovi HU, Hynynen K, Aubert I (2020) Focused
MM, Masliah E, Conner JM, Kobalka P, Roy S, ultrasound delivery of a selective TrkA agonist rescues
Nagahara AH (2015) Nerve growth factor gene ther- cholinergic function in a mouse model of Alzheimer’s
apy: activation of neuronal responses in Alzheimer disease. Sci Adv 6(4):eaax6646. https://doi.org/10.
disease. JAMA Neurol 72(10):1139–1147. https://doi. 1126/sciadv.aax6646
org/10.1001/jamaneurol.2015.1807 Xia B, Lv Y (2018) Dual-delivery of VEGF and NGF by
Varde NK, Pack DW (2004) Microspheres for controlled emulsion electrospun nanofibrous scaffold for periph-
release drug delivery. Expert Opin Biol Ther 4 eral nerve regeneration. Mater Sci Eng C Mater Biol
(1):35–51. https://doi.org/10.1517/14712598.4.1.35 Appl 82:253–264. https://doi.org/10.1016/j.msec.
Venero JL, Hefti F, Knusel B (1996) Trophic effect of 2017.08.030
exogenous nerve growth factor on rat striatal choliner- Xie Y, Ye L, Zhang X, Cui W, Lou J, Nagai T, Hou X
gic neurons: comparison between intraparenchymal (2005) Transport of nerve growth factor encapsulated
and intraventricular administration. Mol Pharmacol into liposomes across the blood-brain barrier: in vitro
49(2):303–310 and in vivo studies. J Control Release 105
Wahlberg LU, Lind G, Almqvist PM, Kusk P, Tornoe J, (1–2):106–119. https://doi.org/10.1016/j.jconrel.2005.
Juliusson B, Soderman M, Sellden E, Seiger A, 03.005
Eriksdotter-Jonhagen M, Linderoth B (2012) Targeted Xie Y, Meeker RB, Massa SM, Longo FM (2019) Modu-
delivery of nerve growth factor via encapsulated cell lation of the p75 neurotrophin receptor suppresses
biodelivery in Alzheimer disease: a technology plat- age-related basal forebrain cholinergic neuron degen-
form for restorative neurosurgery. J Neurosurg 117 eration. Sci Rep 9(1):5273. https://doi.org/10.1038/
(2):340–347. https://doi.org/10.3171/2012.2. s41598-019-41654-8
JNS11714 Xu X, Yu H, Gao S, Ma HQ, Leong KW, Wang S (2002)
Wang Z, Wang Z, Lu WW, Zhen W, Yang D, Peng S Polyphosphoester microspheres for sustained release
(2017) Novel biomaterial strategies for controlled of biologically active nerve growth factor.
growth factor delivery for biomedical applications. Biomaterials 23(17):3765–3772. https://doi.org/10.
NPG Asia Mater 9(10):e435–e435. https://doi.org/10. 1016/s0142-9612(02)00116-3
1038/am.2017.171 Xu D, Wu D, Qin M, Nih LR, Liu C, Cao Z, Ren J,
Wang SM, Lee CU, Lim HK (2019) Stem cell therapies for Chen X, He Z, Yu W, Guan J, Duan S, Liu F, Liu X,
Alzheimer’s disease: is it time? Curr Opin Psychiatry Li J, Harley D, Xu B, Hou L, Chen ISY, Wen J,
32(2):105–116. https://doi.org/10.1097/YCO. Chen W, Pourtaheri S, Lu Y (2019) Efficient delivery
0000000000000478 of nerve growth factors to the central nervous system
Williams LR (1991) Hypophagia is induced by intracer- for neural regeneration. Adv Mater 31(33):e1900727.
ebroventricular administration of nerve growth factor. https://doi.org/10.1002/adma.201900727
Exp Neurol 113(1):31–37. https://doi.org/10.1016/ Yan Q, Matheson C, Sun J, Radeke MJ, Feinstein SC,
0014-4886(91)90143-z Miller JA (1994) Distribution of intracerebral
Williams LR, Varon S, Peterson GM, Wictorin K, ventricularly administered neurotrophins in rat brain
Fischer W, Bjorklund A, Gage FH (1986) Continuous and its correlation with trk receptor expression. Exp
infusion of nerve growth factor prevents basal fore- Neurol 127(1):23–36. https://doi.org/10.1006/exnr.
brain neuronal death after fimbria fornix transection. 1994.1076
Proc Natl Acad Sci U S A 83(23):9231–9235. https:// Yang T, Liu H, Tran KC, Leng A, Massa SM, Longo FM
doi.org/10.1073/pnas.83.23.9231 (2020a) Small-molecule modulation of the p75
Williams LR, Vahlsing HL, Lindamood T, Varon S, Gage neurotrophin receptor inhibits a wide range of tau
FH, Manthorpe M (1987) A small-gauge cannula molecular pathologies and their sequelae in P301S
device for continuous infusion of exogenous agents tauopathy mice. Acta Neuropathol Commun 8(1):156.
into the brain. Exp Neurol 95(3):743–754. https://doi. https://doi.org/10.1186/s40478-020-01034-0
org/10.1016/0014-4886(87)90313-x Yang J, Wu S, Hou L, Zhu D, Yin S, Yang G, Wang Y
Winkler J, Ramirez GA, Kuhn HG, Peterson DA, (2020b) Therapeutic effects of simultaneous delivery
Day-Lollini PA, Stewart GR, Tuszynski MH, Gage of nerve growth factor mRNA and protein via
FH, Thal LJ (1997) Reversible Schwann cell hyperpla- exosomes on cerebral ischemia. Mol Ther Nucleic
sia and sprouting of sensory and sympathetic neurites Acids 21:512–522. https://doi.org/10.1016/j.omtn.
after intraventricular administration of nerve growth 2020.06.013
factor. Ann Neurol 41(1):82–93. https://doi.org/10. Yi X, Manickam DS, Brynskikh A, Kabanov AV (2014)
1002/ana.410410114 Agile delivery of protein therapeutics to CNS. J Con-
Wu K, Meyer EM, Bennett JA, Meyers CA, Hughes JA, trol Release 190:637–663. https://doi.org/10.1016/j.
King MA (2005) AAV2/5-mediated NGF gene jconrel.2014.06.017
11 A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the. . . 191

Zassler B, Humpel C (2006) Transplantation of NGF Zhu CW, Livote EE, Scarmeas N, Albert M, Brandt J,
secreting primary monocytes counteracts NMDA- Blacker D, Sano M, Stern Y (2013) Long-term
induced cell death of rat cholinergic neurons in vivo. associations between cholinesterase inhibitors and
Exp Neurol 198(2):391–400. https://doi.org/10.1016/j. memantine use and health outcomes among patients
expneurol.2005.12.009 with Alzheimer’s disease. Alzheimers Dement 9
Zhang W, Wang PJ, Sha HY, Ni J, Li MH, Gu GJ (2014) (6):733–740. https://doi.org/10.1016/j.jalz.2012.09.
Neural stem cell transplants improve cognitive func- 015
tion without altering amyloid pathology in an APP/PS1 Zhu SP, Wang ZG, Zhao YZ, Wu J, Shi HX, Ye LB, Wu
double transgenic model of Alzheimer’s disease. Mol FZ, Cheng Y, Zhang HY, He S, Wei X, Fu XB, Li XK,
Neurobiol 50(2):423–437. https://doi.org/10.1007/ Xu HZ, Xiao J (2016) Gelatin nanostructured lipid
s12035-014-8640-x carriers incorporating nerve growth factor inhibit endo-
Zhang W, Zhou G, Gao Y, Zhou Y, Liu J, Zhang L, plasmic reticulum stress-induced apoptosis and
Long A, Zhang L, Tang P (2017) A sequential delivery improve recovery in spinal cord injury. Mol Neurobiol
system employing the synergism of EPO and NGF 53(7):4375–4386. https://doi.org/10.1007/s12035-
promotes sciatic nerve repair. Colloids Surf B 015-9372-2
Biointerfaces 159:327–336. https://doi.org/10.1016/j. Zilony-Hanin N, Rosenberg M, Richman M, Yehuda R,
colsurfb.2017.07.088 Schori H, Motiei M, Rahimipour S, Groisman A,
Zhao YZ, Jiang X, Xiao J, Lin Q, Yu WZ, Tian FR, Mao Segal E, Shefi O (2019) Neuroprotective effect of
KL, Yang W, Wong HL, Lu CT (2016) Using NGF nerve growth factor loaded in porous silicon
heparin-poloxamer thermosensitive hydrogels to nanostructures in an Alzheimer’s disease model and
enhance the nerve regeneration for spinal cord injury. potential delivery to the brain. Small 15(45):e1904203.
Acta Biomater 29:71–80. https://doi.org/10.1016/j. https://doi.org/10.1002/smll.201904203
actbio.2015.10.014
NGF Released from Blood Cells or
Collagen Hydrogels as a Therapeutic 12
Target in Alzheimer’s Disease?

Christian Humpel

Abstract Platelets are small anuclear cells and become


Alzheimer’s disease (AD) is a severe neurode- rapidly activated during vascular lesions, and
generative disorder of the brain characterized they may migrate to lesion sites and repair
by extracellular beta-amyloid plaques, blood vessels and also eliminate toxic beta-
intraneuronal tau inclusions, vascular amyloid depositions in vessels. In order to
impairment, inflammation, neurodegeneration, guarantee a stable and slow release, the use
and memory loss. Acetylcholine is the most of biomaterials is of interest, especially colla-
important neurotransmitter for memory, and gen hydrogels that may be useful to protect
cholinergic neurons selectively degenerate in these transmigrating blood cells. In this
AD, and a loss of acetylcholine directly review, I summarize advantages and
correlates with cognitive decline. Nerve challenges of using transmigrating cells to
growth factor (NGF) is the most potent growth deliver NGF directly into the brain.
factor to support the survival of these cholin-
ergic neurons. Thus, researchers are interested Keywords
to deliver NGF directly into the brain to the NGF · Brain delivery · Cell migration ·
cholinergic neurons. As the brain is isolated by Biomaterial · Collagen · Therapeutic
the blood-brain barrier, the large protein NGF
cannot easily pass into the brain, and periph-
eral administration of NGF also causes severe 12.1 Introduction
side effects. Blood cells may represent a potent
therapeutic strategy to deliver NGF into the 12.1.1 Pathology of Alzheimer’s
brain. Monocytes can be isolated and loaded Disease
with NGF and may transmigrate into the brain.
As monocytes are precursors of microglia, Alzheimer’s disease (AD) is a severe neurodegen-
they may differentiate and release NGF but erative disorder of the brain, characterized by
also phagocyte and eliminate toxic plaques. extracellular beta-amyloid (Aβ) depositions
(toxic beta-amyloid (42) plaques) and
intraneuronal hyperphosphorylated tau inclusions
C. Humpel (*)
Laboratory for Psychiatry and Experimental Alzheimer’s (neurofibrillary tangles). These two pathologies
Research, Medical University Innsbruck, Innsbruck, are accompanied by inflammatory processes
Austria including reactive astroglia and microglia, break-
e-mail: christian.humpel@i-med.ac.at; http://www2.i- down of the blood-brain barrier (BBB), and
med.ac.at/psychlab/

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 193
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_12
194 C. Humpel

vascular dysfunctions and deposition of beta- (b) selectively to the target neurons, and
amyloid (40) in vessels (called cerebral amyloid (c) during the whole lifespan.
angiopathy, CAA). A major hallmark in AD is
also the cell death of neurons synthesizing and
releasing acetylcholine. The major neuronal 12.1.3 Delivery of Therapeutics into
populations containing cholinergic neurons are the Brain
located in the septum and in the basal nucleus of
Meynert (nBM) and innervate the hippocampus In order to deliver protective proteins directly into
and whole cortex, respectively. In fact, a decline the tissue, several methods have been used: direct
of cholinergic neurons directly correlates with infusion of growth factors; gene transfer of DNA
cognitive decline and loss of memory. The plasmids using, e.g., adeno- or retroviruses; or
reasons for AD progression are not known yet. transplantation of genetically modified cells
Clinically, acetylcholinesterase inhibitors and the including the development and delivery of
NMDA antagonist memantine are so far the only biomaterials. The requirements for effective
therapeutic choice. delivery systems regarding NGF are (a) to effec-
tively stimulate cholinergic neurons, (b) to con-
tinuously deliver NGF over years, (c) to restrict
12.1.2 NGF and the Cholinergic the distribution to sites of degeneration and to
Neurons in Alzheimer’s Disease avoid adverse effects, and (d) to simplify the
mode of delivery due to the growing disease
Growth factors have been shown to support the population.
survival of different subtypes of neurons in the
central and peripheral nervous system. The sur- Neurosurgical-Invasive Strategies Different
vival of cholinergic neurons is directly dependent invasive strategies of NGF delivery to the brain
on the classical nerve growth factor (NGF) (Levi- have been reported to show that indeed NGF
Montalcini et al. 1995). It is well known that in could be a promising therapeutic in AD, either
AD, the NGF system is markedly impaired intracerebroventricular administration (Olson
playing a role in degeneration of neurons and 1993, Eriksdotter-Jönhagen et al. 1998) or
considered as a therapeutic molecule (Levi- ex vivo gene therapy using grafts of
Montalcini et al. 1995). However, so far it is not NGF-secreting fibroblasts in AD forebrain
known if cell death of cholinergic neurons is a (Tuszynski et al. 2015) or intraparenchymal
primary event in AD or is caused by the dramatic viral gene delivery (Nagahara et al. 2009). How-
deposition of Aβ plaques in the cortex and hippo- ever, it must be completely clear that such an
campus. In our lab, we have extensively studied invasive strategy is limited only to a few selected
the cholinergic neurons of the nBM as patients due to extreme costs and invasive sur-
organotypic brain slices (Weis et al. 2001; gery. As we expect a very high number of AD
Humpel and Weis 2002). We and others showed patients within the next 50 years (10 million in
that NGF (>0.1 ng/ml) is required to support the Europe), it is fully clear that we must develop an
survival of the cholinergic neurons and such nBM alternative to surgery and explore novel noninva-
slices can be cultured for several weeks. We fur- sive delivery strategies.
ther showed that the cholinergic neurons are
dependent on NGF during the whole culture Noninvasive Strategies Noninvasive strategies
period and NGF can only be withdrawn for must focus on novel delivery strategies, such as
2 weeks before starting again NGF delivery in a intravenous, intranasal, or intraocular. In fact
continuous cycle (Weis et al. 2001; Humpel and many different strategies have been tested long
Weis 2002). Thus, it is well established that NGF time ago, but did not succeed due to the complex-
may be a useful neuroprotective therapeutic but ity of targeted delivery without any side effects:
must be applied (a) early before the degeneration, peripheral delivery (Lapchak 1993), BBB
12 NGF Released from Blood Cells or Collagen Hydrogels as a Therapeutic. . . 195

transferrin receptor-mediated transcytosis with papers have been published on monocytes as


OX-26 coupled to NGF (Granholm et al. 1994), therapeutic cells in AD (see our review: Hohsfield
intranasal or intraocular application (Capsoni et al. and Humpel 2015; Malm et al. 2010). Monocytes
2009), NGF absorbed on poly(butyl cyanoacry- can (a) be induced to differentiate into microglia
late) nanoparticles (Kurakhmaeva et al. 2009), and macrophages, (b) be stimulated to migrate to
NGF-loaded microspheres (Gu et al. 2009), or lesion sites or plaques, and (c) eliminate and
cell-based strategies (Kramer et al. 1995). phagocytize plaques (Fig. 12.1b).

Differentiation of Monocytes It has been


suggested that monocytes are the precursors of
12.2 Blood Cell-Derived
microglia and macrophages, and there is clear
Therapeutics
evidence that different cytokines, e.g., tumor
necrosis factor alpha or granulocyte-macrophage
The idea of cell-based strategies takes advantage
colony-stimulating factor, can convert the cells
of the fact that blood cells transmigrate into the
into a microglia-like phenotype (Prinz et al.
brain during normal immune patrol and that, in
2014; Hohsfield et al. 2013).
neurodegenerative diseases, the BBB is broken
and more cells can penetrate into the brain
Migration of Monocytes Emerging evidence
(Kramer et al. 1995). Indeed, during an effective
from studies in stroke, brain trauma, and AD
immune response, the vascular permeability is
indicate that these severe brain disorders can
increased along with blood flow to the site of
lead to BBB breakdown and subsequent migra-
inflammation or injury. This is accompanied by
tion of blood-derived monocytes into the brain.
the selective entry of immune cells from the
Monocytes are recruited to sites of Aβ plaque
periphery. It has been well established that many
deposition (triggered by Aβ40 and Aβ42) and can
different cells can transmigrate into the brain (see
eliminate Aβ deposits via phagocytosis (Fiala
our review: Hohsfield and Humpel 2015), e.g.,
et al. 2005; Giri et al. 2000; Humpel 2008). In
activated T lymphocytes, microglia, monocytes,
vivo it was reported that monocyte deposition is
B cells, mast cells, stem cells, but also platelets
only present in the brains of AD transgenic mice,
(Kniewallner et al. 2015). The general aim of
neighboring Aβ plaques, whereas little or no cell
such a strategy is (a) to isolate blood cells from
deposition is present in the brains of control
patients; (b) to manipulate ex vivo, e.g., to load
animals (Lebson et al. 2010; Stalder et al. 2005).
NGF; and (c) to infuse the cells back into the
Monocytes can be triggered to migrate to a lesion
same patient (homotransplant), so that (d) the
site, and indeed monocyte chemoattractant
cells can transmigrate into the brain and release
protein-1 (MCP-1) can induce migration of
the protective/regenerative factor at the target of
monocytes (Fig. 12.1b). Thus, the release of
interest.
MCP-1 from activated microglial cells around
plaques may recruit monocytes to plaques.

12.2.1 Monocytes in the AD Brain Phagocytic Function of Monocyte-Derived


Microglia Extensive in vivo and in vitro evi-
Monocytes play an important role during innate dence indicates that blood-derived monocytes
and adaptive immune responses, and inflamma- can phagocytize Aβ (Hawkes and McLaurin
tion leads to an increased recruitment of 2009; Malm et al. 2010); however, the exact
monocytes to peripheral tissues aiding in host mechanism of how monocytes clear Aβ remains
immune system defense and repair (Fig. 12.1a). unclear (Fig. 12.1b). Some studies have
Monocytes are 5–10μm blood cells and are demonstrated that insoluble fibrillar Aβ can bind
identified by their expression of, e.g., CD14, to receptors on microglia and activate these cells
CD11b, CD115, F4/80, or Ly6C+. In fact, many to produce a number of Aβ-degrading enzymes,
196 C. Humpel

Fig. 12.1 Blood-derived cells may support survival of monocytes can be loaded with nerve growth factor
cholinergic neurons (a), phagocytize and eliminate (NGF) and may penetrate a damaged blood-brain barrier
plaques (b), or repair brain vessels (c). (a) Primary in Alzheimer patients. These NGF-loaded monocytes may
12 NGF Released from Blood Cells or Collagen Hydrogels as a Therapeutic. . . 197

including insulysin, neprilysin, and others NGF-loaded monocytes could partly recover sur-
(Frenkel et al. 2013). We have shown that vival of cholinergic nBM neurons (Hohsfield
monocyte-derived microglia can phagocytize et al. 2014). In vivo, we (Hohsfield et al. 2014)
FITC-labeled Aβ (Hohsfield and Humpel infused monocytes (5  106 cells, 100μl via the
2015a). In a previous paper, we (Hohsfield and dorsal penile vein i.v. 1/week) into mice and
Humpel 2015a) showed that monocytes derived found that a majority was captured in the lung,
from young 2-week-old mice were effective to liver, and spleen 24 h after a single injection.
eliminate plaques in a transgenic APP_SweDI However, after repeated infusion (2 months
mouse model after tail vein infusion. In fact, the weekly), monocytes were found in the cortex/
number of small (5–20μm) and large (>40μm) hippocampus and slightly increased the number
Aβ plaques was slightly reduced. This clearly of cholinergic neurons in the nBM after infusion
points to the migratory and phagocytic capacity of NGF-loaded monocytes (Fig. 12.1A).
of especially young monocytes in the brain. Much
more work is necessary to prove that indeed
monocytes can counteract plaque pathologies in 12.2.2 Platelets in the AD Brain
humans.
Platelets (thrombocytes) are responsible for
Transmigration of NGF-Loaded Monocyte- maintaining the integrity of blood vessels and
s Unfortunately, the transfection of primary promoting hemostasis, and they rapidly migrate
monocytes is difficult. In a previous work, we to a damaged vessel. As in AD the vessels are
(Hohsfield et al. 2013a) used electroporation, the damaged and the BBB is broken, platelets are an
lipids effectene and FuGene, nucleofection, important cell for vessel repair. They are very
BioPORTER delivery, as well as a lentiviral small cells of 3–5μm size and do not have a
transfection in order to load NGF into monocytes. nucleus. Platelets are characterized by different
The most efficient method was lentiviral transfec- integrins, e.g., CD71, CD62P, or CD31
tion giving 16 ng/ml NGF release within 24 hours (Kniewallner et al. 2014). Platelets are of particu-
per one million monocytes. Using nucleofection lar interest in AD since they contain high amounts
or BioPORTER, the efficiency was 20 less of amyloid-precursor protein (APP) and release
(0.6–0.8 ng/ml NGF release), and nucleofection Aβ40 into the bloodstream (Evin et al. 2003),
also damaged monocytes, while in BioPORTER, which may play a role in blood clotting and vessel
they were saved. Anyhow BioPORTER repair. Furthermore, studies have shown that the

ä
Fig. 12.1 (continued) migrate to cholinergic nerve also migrate to cholinergic neurons and support their sur-
terminals, where NGF is released from monocytes and vival. (d) Blood cells may be loaded into colla-
acts as a therapeutic growth factor for cholinergic neurons gen hydrogels or other biomaterials, where they are
to support their survival. This is of particular interest as protected and released within time; alternatively, biomate-
every neuron is located close to a vessel capillary rial may be chemically modified to be blood stable (e.g., as
(20–50μm distance). (b) Monocytes have the capacity to microspheres); finally incorporation of targeting
differentiate into microglia or macrophages and migrate to molecules (e.g., antibodies, magnetic beads, integrins)
lesion sites when activated by, e.g., macrophage into biomaterials may allow selective transmigration
chemoattractant protein-1 (MCP-1). In the Alzheimer through the blood-brain barrier and targeting to specific
brain, such monocyte-derived microglia/macrophages brain areas. Taken together, an approach where blood cells
may phagocytize and eliminate beta-amyloid plaques. (c) are derived from humans, manipulated ex vivo, protected
Platelets have the capacity to migrate to damaged vessels, with biomaterials, and then injected into the same patient
clotting the lesion site. Platelets loaded with the beta- (as a homotransplant) may provide a novel therapeutic
amyloid degrading-enzyme neprilysin may migrate to ves- approach to counteract Alzheimer pathologies (I thank
sel lesions and degrade plaques in vessels (the cerebral Lindsay Hohsfield for help with drawings)
amyloid angiopathy). Platelets co-loaded with NGF may
198 C. Humpel

larger 130 kDa APP isoform is significantly as sponges or hydrogels, and is known for its
reduced in platelets isolated from AD patients, excellent biocompatibility due to its low toxicity
implicating their potential role in altered APP and poor immunogenic reactions. Collagen
metabolism (Padovani et al. 2001). Thus, there hydrogels (CollH) are easily and rapidly
is clear evidence that platelets could become an generated using different chemical cross-linkers
interesting target in AD, either diagnostic with such as glutaraldehyde, formaldehyde, or poly-
APP as a biomarker or therapeutic as a tool for ethylene glycol (PEG), as well as physical treat-
vessel repair. In a previous study, we ment with dry heat and ultraviolet irradiation
(Kniewallner et al. 2015) infused platelets into (Friess 1998; Caliari and Burdick 2016). CollH
the brain (5  108, 100μl i.v. tail vein, 1/ can be efficiently loaded with therapeutically
week) and found that the majority was trapped active substances and release them in a
in the liver and lung 24 h after infusion. Only a degradation-based fashion, in which the sub-
minority entered the brain, but those were seen in stance is released along with the degradation of
the ventricle and partly close to a plaque. We the collagen (Foidl et al. 2018). CollH are
(Kniewallner et al. 2014) further showed that injectable, which provides direct application to
platelets can be loaded with NGF using ultra- the target site of action rather than a systemic
sound giving 6.5 ng/mg NGF content. Indeed administration route. As the bioavailability of
those NGF-loaded platelets can rescue cell death many therapeutics to the brain is greatly restricted
of cholinergic neurons in organotypic brain slices by the BBB, injectable CollH may be a minimally
(Kniewallner et al. 2014). Much more work is invasive method of administration that provides
necessary to evaluate if platelets can have a ther- local, controlled, and prolonged release of the
apeutic function in mouse models or even growth factor with less systemic side effects. In
humans. We favor the idea to load platelets with addition to difficulties in passage through the
the plaque-degrading enzyme neprilysin, which BBB, many small molecules are unstable in the
could be useful to eliminate plaques in the vessels body and degrade rapidly before they reach their
and counteract symptoms of CAA (Fig. 12.1c). target. Application of therapeutics within CollH
will prevent rapid degradation of the therapeutics
before they reach the brain and may also be useful
12.3 Collagen Hydrogels as for systemic applications.
Therapeutics

12.3.1 Collagen Hydrogels in Brain 12.3.2 NGF Released from Collagen


Repair Hydrogels

The use of biomaterials provides new CollH have been widely used to deliver growth
opportunities for protection and regeneration of factors to different tissues. Coating of collagen
the brain, and hydrogels and microspheres from sponges with fibroblast growth factor-2 facilitated
various natural or synthetic biomaterials can be early dermal and epidermal wound healing
efficiently loaded with therapeutically active (Marks et al. 1991). Platelet-derived growth
agents, e.g., growth factors (see our review: factors loaded into collagen sponges enhanced
Ucar and Humpel 2018). Thereby, these capillary formation after 1 week (Royce et al.
hydrogels and/or micro-/nanospheres provide 1995), and bone morphogenetic proteins loaded
application of relevant therapeutics in a localized into collagen sponges promoted osseous regener-
manner, which minimizes unwanted side effects ation (Kenley et al. 1993). Collagen hollow
that are observed with systemic administration spheres fabricated with a template method were
and enhances the effectiveness of the therapy. useful to load large amounts of growth factors,
Collagen has been extensively applied directly e.g., NGF (Kraskiewicz et al. 2013) or vascular
upon different tissues, either as fluid collagen or endothelial growth factor (VEGF) (Nagai et al.
12 NGF Released from Blood Cells or Collagen Hydrogels as a Therapeutic. . . 199

2010), without causing any toxicity in vitro and Thus, the targeting directly into the brain area of
in vivo. It is well known that NGF can be released interest is the most important issue to be solved.
from different types of collagen-associated bio-
material: Jin et al. (2013) reported that NGF Delivery Methods As discussed earlier, noninva-
released from collagen and hyaluronan hydrogels sive delivery of therapeutics is essential when
can stimulate neurite fiber growth for peripheral thinking about an AD strategy. As we expect
nerve repair. Zeng et al. (2014) incorporated NGF millions of AD patients, an invasive surgical
into collagen-chitosan hydrogels and observed strategy will not be useful. In addition, we must
sustained release of NGF within 4 weeks in vitro. also develop strategies to protect the cholinergic
Others reported controlled release of NGF but neurons and/or to eliminate plaques already at the
with a focus on hollow fibrin microspheres beginning of the disease. As AD starts already
(Samal et al. 2015) or highly cross-linked colla- 30 years before plaque development, such a strat-
gen spheres (Kraskiewicz et al. 2013). In our egy must be done in younger individuals without
group, we (Foidl et al. 2018) showed that NGF any marked cognitive impairment (mild cognitive
can be loaded into CollH and release sufficient impairment, MCI). Again, a simple and easy
biological active NGF in organotypic brain slices application strategy is essential. Four modes of
to counteract cholinergic neurodegeneration. application are suggested: intravenous, intrana-
Much more work needs to be done to show the sal, intraocular, and intracerebrospinal. Intra-
usefulness of CollH especially via the systemic venous peripheral application provides a simple
application route. and easy noninvasive strategy, but the most seri-
ous problems are stability in blood, trapping in
liver-spleen-lung or elsewhere, high metabolism,
12.4 Outlook and Hope side effects on peripheral neurons, low delivery
into the brain, low passage through BBB, or high
The topic that “NGF can be used as a therapeutic amounts of factor needed. Intranasal or intraocu-
in AD” is now nearly 30–40 years old but did not lar application may take advantage of the fact that
succeed to become a human therapy. The prob- the BBB is more open at those sites and factors
lem is the complexity of application and the can easier pass. In fact, NGF could be applied as a
targeting to the brain area of interest. In addition, “NGF spray” via the nose and may reach the brain
most studies with NGF have been conducted too via the olfactory bulb. Intraspinal application is
late, at a time where most of the cholinergic an alternative, as the cerebrospinal fluid (CSF) is
neurons are already gone. So what to protect if in direct contact with the brain. In fact, CSF is
there is nothing to protect? But still, the story is routinely used for analysis of different brain-
ongoing, and indeed there is hope that NGF could derived biomarkers and allows to diagnose AD
become a therapeutic factor in AD. with high specificity. Such an application may
target the therapeutic of interest better to the
Safety and Biocompatibility First, it is important brain. But, even if the therapeutic reaches the
to test the biocompatibility and stability of the brain, it must be targeted to the brain area of
therapeutic strategy in ex vivo and in vivo interest, and this is the hippocampus or the cortex,
models. This will give us information if (a) the if NGF is considered to be taken up by nerve
therapeutic has neurotoxic effects, induces reac- terminals as a retrograde-acting growth factor.
tive gliosis, or causes microglial immune
reactions and on (b) how long is the therapeutic Blood-Stable Collagen Hydrogels An impor-
stable and (c) how long can cells survive after tant step in the delivery process could be to stabi-
application. This is extremely important espe- lize the growth factor or cell in the blood or nasal
cially when such a therapy will be transferred to fluid or CSF. Especially long-term repeated appli-
humans. The major concern of applying NGF is cation may make this necessary. Thus,
the side effect at the spinal cord neurons resulting researchers develop methods to encapsulate cells
in uncontrolled nerve fiber growth and pain.
200 C. Humpel

or drugs in biomaterials, e.g., microspheres or References


nanospheres. Such small vehicles may easily
transport a drug of interest into the brain and are Caliari SR, Burdick JA (2016) A practical guide to
also stabilized. On the other hand, such a bioma- hydrogels for cell culture. Nat Methods 13(5):405–414
Capsoni S, Covaceuszach S, Ugolini G, Spirito F,
terial is not degradable and can be concentrated in Vignone D, Stefanini B, Amato G, Cattaneo A (2009)
cells and cause neurotoxic effects or immune Delivery of NGF to the brain: intranasal versus ocular
responses. Thus, the use of biodegradable mate- administration in anti-NGF transgenic mice. J
rial, e.g., collagen, could overcome such potential Alzheimers Dis 16:371–388
Eriksdotter-Jönhagen M, Nordberg A, Amberla K,
problems, because it is degraded over time and Bäckman L, Ebendal T, Meyerson B, Olson L, Seiger
eliminated by the cells. SM, Theodorsson E, Viitanen M, Winblad B, Wahlund
LO (1998) Intracerebroventricular infusion of nerve
Targeting to the Brain As mentioned previ- growth factor in three patients with Alzheimer’s dis-
ease. Dement Geriatr Cogn Disord 9:246–257
ously, targeting of the therapeutic to the brain Evin G, Zhu AHR, Masters CL, Li QX (2003) Proteolytic
and more specifically to the brain area of interest processing of the Alzheimer's disease amyloid precur-
is the most important challenge. This issue must sor protein in brain and platelets. J Neurosci Res
involve a transdisciplinary approach including 74:386–392
Fiala M, Lin J, Ringman J, Kermani-Arab V, Tsao G,
neurobiologists, chemists, and biotechnologists. Patel A, Lossinsky AS, Graves MC, Gustavson A,
The delivery vehicle (be it a blood cell, a micro- Sayre J, Sofroni E, Suarez T, Chiappelli F, Bernard G
sphere, or a collagen) must be chemically (2005) Ineffective phagocytosis of amyloid-beta by
modified to target the BBB. One may think of macrophages of Alzheimer's disease patients. J
Alzheimers Dis 7:221–232
magnetically labeled vehicles which could be Foidl BM, Ucar B, Schwarz A, Rebel AL, Pandit A,
targeted to the brain with strong magnets. In Humpel C (2018) Nerve growth factor released from
fact, magnetic stimulation is a well-established collagen scaffolds protects axotomized cholinergic
approach in clinics, e.g., by using MRI or TMS neurons of the basal nucleus of Meynert in organotypic
brain slices. J Neurosci Methods 295:77–86
(transmagnetic stimulation). It is suggested to Frenkel D, Wilkinson K, Zhao L, Hickman SE, Means TK,
load vehicles with a therapeutic and magnetic Puckett L, Farfara D, Kingery ND, Weiner HL, El
beads and to magnetically stimulate the brain Khoury J (2013) Scara1 deficiency impairs clearance
area of interest. Strong magnets could be applied of soluble amyloid-β by mononuclear phagocytes and
accelerates Alzheimer’s-like disease progression. Nat
outside of the brain and force the magnetically Commun 4:2030
labeled vehicles to migrate into the brain area of Friess W (1998) Collagen – Biomaterial for drug delivery.
interest. Eur J Pharm Biopharm 45(2):113–136. https://doi.org/
10.1016/S0939-6411(98)00017-4
Giri R, Shen Y, Stins M, Du Yan S, Schmidt AM, Stern D,
Kim KS, Zlokovic B, Kalra VK (2000) Beta-amyloid-
12.5 Conclusion induced migration of monocytes across human brain
endothelial cells involves RAGE and PECAM-1. Am J
Physiol Cell Physiol 279:C1772–C1781
An approach where blood cells are derived from Granholm AC, Bäckman C, Bloom F, Ebendal T, Gerhardt
humans, manipulated ex vivo, protected with GA, Hoffer B, Mackerlova L, Olson L, Söderström S,
biomaterials, and then injected into the same Walus LR, Friden P (1994) NGF and anti-transferrin
receptor antibody conjugate: short and long-term
patient (as a homotransplant) may provide a effects on survival of cholinergic neurons in intraocular
novel therapeutic strategy to counteract septal transplants. J Pharmacol Exp Ther 268:448–459
Alzheimer pathologies. Blood cells or growth Gu H, Long D, Song C, Li X (2009) Recombinant human
factors loaded into biomaterials (e.g., collagen NGF-loaded microspheres promote survival of basal
forebrain cholinergic neurons and improve memory
or microspheres) may protect the cells in blood impairments of spatial learning in the rat model of
and may enhance specific transmigration through Alzheimer's disease with fimbria-fornix lesion.
the BBB and selective physical targeting to spe- Neurosci Lett 453:204–209
cific brain areas. Hawkes CA, McLaurin J (2009) Selective targeting of
perivascular macrophages for clearance of beta-
12 NGF Released from Blood Cells or Collagen Hydrogels as a Therapeutic. . . 201

amyloid in cerebral amyloid angiopathy. Proc Natl Brain targeting of nerve growth factor using poly(butyl
Acad Sci U S A 106:1261–1266 cyanoacrylate) nanoparticles. J Drug Target 17
Hohsfield L, Humpel C (2015) Migration of blood cells to (8):564–574
beta-amyloid plaques in Alzheimer's disease. Review. Lapchak PA (1993) Nerve growth factor pharmacology:
Exp Gerontol 65:8–15 application to the treatment of cholinergic
Hohsfield L, Humpel C (2015a) Intravenous infusion of neurodegeneration in Alzheimer’s disease. Exp Neurol
monocytes isolated from 2-week-old mice enhances 124(1):16–20
clearance of beta-amyloid plaques in an Alzheimer Lebson L, Nash K, Kamath S, Herber D, Carty N, Lee DC,
mouse model. PlosONE 10(4):e0121930. https://doi. Li Q, Szekeres K, Jinwal U, Koren J, Dickey CA,
org/10.1371/journal.pone.0121930. eCollection 2015 Gottschall PE, Morgan D, Gordon MN (2010) Traf-
Hohsfield LA, Ammann C, Humpel C (2013) Inflamma- ficking CD11b-positive blood cells deliver therapeutic
tory status of transmigrating primary rat monocytes in genes to the brain of amyloid-depositing transgenic
a novel perfusion model simulating blood flow. J mice. J Neurosci 30:9651–9658
Neuroimmunol 258(1–2):17–26 Levi-Montalcini R, Dal Toso R, della Valle F, Skaper SD,
Hohsfield LA, Geley S, Reindl M, Humpel C (2013a) The Leon A (1995) Update of the NGF saga. J Neurol Sci
generation of NGF-secreting primary rat monocytes: a 130(2):119–127
comparison of different transfer methods. J Immunol Malm T, Koistinaho M, Muona A, Magga J, Koistinaho J
Methods 391(1–2):112–124 (2010) The role and therapeutic potential of monocytic
Hohsfield LA, Ehrlich D, Humpel C (2014) Intravenous cells in Alzheimer's disease. Glia 58:889–900
infusion of nerve growth factor-secreting monocytes Marks MG, Doillon C, Silvert FH (1991) Effects of
supports the survival of cholinergic neurons in the fibroblasts and basic fibroblast growth factor on facili-
nucleus basalis of Meynert in hypercholesterolemia tation of dermal wound healing by type I collagen
Brown-Norway rats. J Neurosci Res 92:298–306 matrices. J Biomed Mater Res 25:683–696
Humpel C (2008) Basolateral aggregated rat amyloidbeta Nagahara AH, Bernot T, Moseanko R, Brignolo L,
(1-42) potentiates transmigration of primary rat Blesch A, Conner JM, Ramirez A, Gasmi M,
monocytes through a rat blood-brain barrier. Curr Tuszynski MH (2009) Long-term reversal of choliner-
Neurovasc Res 5:185–192 gic neuronal decline in aged non-human primates by
Humpel C, Weis C (2002) Nerve growth factor and cho- lentiviral NGF gene delivery. Exp Neurol 215
linergic CNS neurons studied in organotypic brain (1):153–159
slices: implications in Alzheimer’s disease? Journal Nagai N, Kumasaka N, Kawashima T, Kaji H,
Neural Transmission Supplement 62:253–263 Nishizawa M, Abe T (2010) Preparation and charac-
Jin J, Limburg S, Joshi SK, Landman R, Park M, Zhang Q, terization of collagen microspheres for sustained
Kim HT, Kuo AC (2013) Peripheral nerve repair in rats release of VEGF. J Mater Sci Mater Med
using composite hydrogel-filled aligned nanofiber 21:1891–1898
conduits with incorporated nerve growth factor. Tissue Olson L (1993) NGF and the treatment of Alzheimer’s
Eng Part A 19(19–20):2138–2146 disease. Exp Neurol 124:5–15
Kenley RA, Yim K, Abrams J, Ron E, Turek T, Marden Padovani A, Pastorino L, Borroni B, Colciaghi F,
LJ, Hollinger JO (1993) Biotechnology and bone graft Rozzini L, Monastero R, Perez J, Pettenati C,
substitutes. Pharm Res 10:1393–1401 Mussi M, Parrinello G, Cottini E, Lenzi L,
Kniewallner K, Grimm N, Humpel C (2014) Platelet- Trabucchi M, Cattabeni F, Di Luca M (2001) Amyloid
derived nerve growth factor supports the survival of precursor protein in platelets: a peripheral marker for
cholinergic neurons in organotypic rat brain slices. the diagnosis of sporadic AD. Neurology
Neurosci Lett 574:64–69 57:2243–2248
Kniewallner KM, Ehrlich D, Kiefer A, Marksteiner J, Prinz M, Tay TL, Wolf Y, Jung S (2014) Microglia:
Humpel C (2015) Platelets in the Alzheimers disease unique and common features with other tissue
brain: do they play a role in cerebral amyloid macrophages. Acta Neuropathol 128(3):319–331
angiopathy? Curr Neurovasc Res 12:4–14 Royce PM, Kato T, Ohsaki KI, Miura A (1995) The
Kramer R, Zhang Y, Gehrmann J, Gold R, Thoenen H, enhancement of cellular infiltration and vascularisation
Wekerle H (1995) Gene transfer through the blood- of a collagenous dermal implant in the rat by platelet-
nerve barrier: NGF-engineered neuritogenic T derived growth factor BB. J Dermatol Sci 10:42–52
lymphocytes attenuate experimental autoimmune neu- Samal J, Deirdre B, Naughton C, Concannon R, Dowd E,
ritis. Nature Med 1:1162–1166 Pandit A (2015) Fibrin-based microsphere reservoirs
Kraskiewicz H, Breen B, Sargeant T, McMahon S, Pandit for delivery of neurotrophic factors to the brain.
A (2013) Assembly of protein-based hollow spheres Nanomedicine (Lond) 10(5):765–783
encapsulating a therapeutic factor. ACS Chem Stalder AK, Ermini F, Bondolfi L, Krenger W, Burbach
Neurosci 4:1297–1304 GJ, Deller T, Coomaraswamy J, Staufenbiel M,
Kurakhmaeva KB, Djindjikhashvili IA, Petrov VE, Landmann R, Jucker M (2005) Invasion of
Balabanyan VU, Voronina TA, Trofimov SS, hematopoietic cells into the brain of amyloid precursor
Kreuter J, Gelperina S, Begley D, Alyautdin (2009) protein transgenic mice. J Neurosci 25:11125–11132
202 C. Humpel

Tuszynski MH, Yang JH, Barba D, Hoi-Sang U, Bakay Weis C, Marksteiner J, Humpel C (2001) Nerve growth
RA, Pay MM, Masliah E, Conner JM, Kobalka P, factor and glial cell line-derived neurotrophic factor
Roy S, Nagahara AH (2015) Nerve growth factor restore the cholinergic neuronal phenotype in
gene therapy activates neuronal responses in organotypic brain slices of the basal nucleus of
Alzheimer’s disease. JAMA Neurol 72(10):1139–1147 Meynert. Neuroscience 102:129–138
Ucar B, Humpel C (2018) Collagen for brain repair: thera- Zeng W, Rong M, Hu X, Xiao W, Qi F, Huang J, Luo Z
peutic perspectives – a mini-review. Neural Regen Res (2014) Incorporation of chitosan microspheres into
13(4):595–598 collagen-chitosan scaffolds for the controlled release
of nerve growth factor. PLoS One 9:e101300
Part IV
Complex Behaviors
When Nerve Growth Factor Met
Behavior 13
Daniela Santucci, Arianna Racca, and Enrico Alleva

Abstract described since then, and further studies car-


Since its first characterization in the early ried out on humans showed that NGF is
1950s, the role of the polypeptidic nerve released in the bloodstream of parachutists at
growth factor (NGF) in controlling behavior their first skydiving experience and in the case
remained elusive. Since the mid-1980s, we of ranking high on the Passionate Love Scale
undertook a series of experiments aimed at (amour fou). Ethological data from lab rodents
elucidating the biological role(s) played by helped in understanding NGF function in sub-
neurotrophins, particularly NGF, in adult tly controlling social “status” of male mice: the
rodents. At the beginning, we concentrated considerations about the interplay among neu-
on the submandibular salivary gland of the robiological, physiological, and behavioral
male mouse, which was known to store mas- factors in structuring the dominant vs subordi-
sive amount of NGF. We found that under nate phenotypes may well apply to other ver-
specific stress conditions, the salivary NGF is tebrate species, specifically addressing the
released in the bloodstream: intermale fighting underlying role of neurotrophins in relating
between isolated males was the first reported behavior and brain neuroplasticity.
context in which salivary NGF was released,
thus providing a physiological significance for
its presence in the adult, territorial males. We
also found that dominant males release less 13.1 Introduction
NGF than subordinates and provided a loop-
type model which includes intermale social It was July 1983 when, in central Rome, in the
confrontation, adrenal gland size, and func- elegant attic of Viale di Villa Massimo 3–5, fol-
tional status, corticosterone release, a model lowing an almost 10-year-long discussion with
resulting in likelihood to be stabilized in a Rita Levi-Montalcini (RLM) about the mysteri-
“dominant” or a “subordinate” social status. ous, yet likely physiological, role of NGF stored
A variety of social anxiety contexts of in the submaxillary “salivary” glands of adult
mammals, humans included, has been male mice, in collaboration wither her closest
fellow Luigi Aloe, we decided to attempt what
is now regarded as a crucial experiment
D. Santucci (*) · A. Racca · E. Alleva
Reference Center for Behavioural Sciences and Mental (Fig. 13.1).
Health, Istituto Superiore di Sanità, Rome, Italy The origin of NGF release into the circulation
e-mail: Daniela.santucci@iss.it; arianna.racca@guest.iss. was in the early Eighty highly debated, and we
it; enrico.alleva@iss.it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 205
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_13
206 D. Santucci et al.

tested whether, under an ethological evolutionary neurons located in the central nervous system
perspective, intermale aggressive behavior could and, in an explosive, unusual advancement of
represent the “elusive” triggering factor causing new knowledge, a jump which illuminated a vari-
NGF release. We found the mysterious trigger. ety of neuroethological phenomena, on a variety
This was indeed the case: NGF was quite of immune cell types (Alleva and Calamandrei
rapidly released after about 25–30 separate inter- 1990); for the evolution of growth factor proteins,
male “territorial-type” fighting episodes occur- we refer to the enlightening paper by
ring in previously isolated adult male mice Black (1986).
during a single 20-min session (subjects with a Aloe, in close collaboration with our ISS
previous parental experience showed peak levels; group, was the first to demonstrate that NGF is
isolation per se is an enhancer). More interest- released in humans following stress. We
ingly, by exploiting a dyadic 10-repeated exploited Holger Ursin’s model (CAT; Ursin
encounters paradigm, with intermale encounters and Eriksen 2004) of intensive training and the
occurring in a “neutral cage,” devoid of any very first analysis, with a really original perspec-
marking scent odor, subordinated males released tive, of skydiving experience of military
about twice as much NGF as “dominants.” Rather parachutists as a potent, rather unique, stressor.
surprisingly, a series of non-social stressors, such To jump from an airplane in the void is really a
as a very mild level of escapable or inescapable neuromotor, emotional, and even cognitive per-
foot shock, cold water swimming, or forced biting formance violating one of the most basic survival
of a wooden stick, all failed to elicit NGF release. rules.
Only changes in social bonding, including Even neonates, immediately after parturition,
pre-copula arousal and maternal nest-defending show a kind of fixed-action pattern (Lorenz 1935)
(this against a potentially infanticidal male which triggers a compulsive response to avoid
attacker), triggered a high level of NGF release falling down (Fox 1965).
in the bloodstream (Branchi et al. 2004). Therefore, even if “cultural” (inner motivation,
NGF was discovered, biochemically military hard training, sport necessity, war con-
characterized, and searched for its physiological text, intermale attempt to emerge, etc.) produces
role(s) by Levi-Montalcini already at the begin- such an anti-Darwinian performance act, the NGF
ning of the 1950s (Levi-Montalcini 1987a, b). release remains however an adjustment response
Since NGF discovery, which opened a vast ave- to cope with a particularly severe stress situation.
nue in the early and late exploration and exploita- In the subsequent decades, various laboratories
tion of neurotrophic/trophic proteins in a wide found that even much lower-level stressors were
variety of biomedical endpoints, no sound expla- enough to trigger NGF release in the bloodstream.
nation was available during almost three decades Even presently, the most likely possibility to store
to disentangle NGF physiological role in adult and release NGF in the human species remains
mammals. Most neurobiologists explained its some cellular elements belonging to the immune
presence in adult subjects as a kind of long-term system (Torcia et al. 1996).
remains of a molecule having exerted a pivotal At the end of prolonged and standardized
physiological role during early or middle training, however with some individual
ontogeny. variability in its duration (a skillful officer even-
This also provided for a while an “unstable,” tually decided that the single parachutist is ready
fallacious early explanation for the presence of to jump), one evening, a young parachutist of a
NGF receptors even in the adult brain. selected (also by temperamental attitudes) Air-
NGF was originally described by RLM and borne Brigade of the Italian army (Esercito
colleagues as a protein selectively acting on sym- Italiano) was eventually given the possibility by
pathetic peripheral neurons (initially in bird expert military trainers to eventually launch from
embryos), and only decades later, it was found an airplane the following morning.
to be also exerting some physiological role on
13 When Nerve Growth Factor Met Behavior 207

Fig. 13.1 Laurea honoris


causa in Biomedical
Sciences to Luigi Aloe,
“Alma Mater” Bologna
University (Italy) 22/11/
1989

Immediately after being informed that his Those phenomena were only considered in the
training was ended and the jumping event was realm of social sciences for many decades, a
scheduled, blood NGF levels start to raise, in trend that progressively evolved when “cogni-
anticipation of the highly emotional skydiving tive” sciences, mainly cognitive ethology, started
experience that will follow the subsequent morn- to emerge. Some major bias still affects these
ing. Social competition among young men, living developing science areas, since it is well known
in sexually segregated groups, may well exert a that jumping from a different (regularly lower)
major triggering role in such a sudden NGF level of analysis, especially in a typical reduction-
release (Branchi et al. 2004). istic, even paleo-positivistic, perspective, is still
After so many years, it is still surprising to very often a classical mistake, obscuring the deli-
observe in how many cases NGF is released cate relationship between the expression of indi-
mainly following social and stressful events. vidual behavioral phenotypes and their
Racing horses—likely because of equine social physiological, lower-level determinants (molecu-
competition—release NGF in the bloodstream lar, nuclear, cellular, tissue, organ, system, full
upon running in a furious game. individual), just to mention some major descrip-
In humans, Aloe’s group showed that during tive hierarchies and their various levels of analy-
labor and lactation, NGF was fivefold enhanced. sis. We do recommend the enlightening reflection
The fatiguing burden of prolongedly assisting a by Steven Rose (1981).
cognitively impaired close relative raises NGF These were rarely approached in the past by
blood levels up to 200% (Hadjiconstantinou means of standardized methods of behavioral
et al. 2001). Also during the recognized stressful neurobiology, cognitive ethology, experimental
phase of smoking habit cessation, NGF levels psychology, endocrinology, or functional neuro-
grow rapidly, almost doubling in only 3 days anatomy, just to mention some sub-fields where
(Lang et al. 2002). the mistaken explanations violating level-of-anal-
Behavioral and neurobiology scientists of this ysis considerations are rather common. Human
Third Millennium are revisiting several phenom- geneticist Luigi “Luca” Cavalli-Sforza in his orig-
ena, such as social competition, proudness, and inal reconstruction of human evolution (Cavalli-
even love, often considered as unique to humans Sforza 1996; Diamond 1998) well testified of this
and thus dealing with emotions and passions. dualistic, cross-pollinating approach,
208 D. Santucci et al.

encompassing the gap between the Two Cultures, Carnival, in the early 1950s. Her enduring love
Hard Sciences vs Humanities. A third culture, for the biological properties of this protein,
melting social-type phenomena with more almost two decades after her Nobel Prize, lasted
“hard-science” methodologies, may be eventually until the end of her life.
the most useful response for contemporary analy-
sis of behavioral (e.g., coping response to a vari-
ety of stressors administered under different 13.2 Early Phases of Development:
contexts) phenomena. The Mother-Pups Interaction
Psychoneuroendocrinology already devoted
years ago an entire issue to the “neurobiology of The importance of the role of neurotrophins in
love” in 1998 (Vol. 23, Issue 8). This editorial directing brain growth and neuronal functionality
effort included a paper by Emanuele et al., has been increasingly recognized.
exploiting a rather reductionist, yet relevant, sim- In the last three decades, in the scientific
ilar approach, concomitantly assessing the levels Rome-based group, coordinated and inspired by
of the four most characterized neurotrophins Rita, the studies carried out by William T. “Bill”
(NGF, BDNF, NT-3, and NT-4) in the human Mobley, then at Stanford Medical School on the
circulation to disentangle the neurobiological NGF role on developing postnatal cholinergic
bases of what is described as “romantic love.” central nervous system neurons, represented a
(Incidentally, in some psychiatric manuals, such veritable step: a neurotrophin reportedly
a “state” or a syndrome (!) is considered a regulating (selectively) early adrenergic develop-
pre-alerting condition, possibly followed, even ment of sympathetic sensory neurons in birds was
very rarely and with a marked sex bias, by a in fact demonstrated to speed up CNS
suicidal act). cholinergics in rats.
This is in line with several other analyses When Mobley visited Rome at a special inti-
aimed at attempting to understanding the physio- mate dinner in Rita’s apartment, such former
logical, sometimes “key,” regulators underlying selective properties lose their specificity. Her pro-
such a complex behavioral phenomenon, possibly found and durable scientific and personal rela-
not unique to the human species, that mixes tionship with Eric Shooter, then Mobley’s
feelings, passions, emotions, mental states, inner supervisor at Stanford, also played a relevant
desires, and personalized motivations. The most role since he magnified Mobley’s results as well
intriguing finding in the paper by Emanuele et al. as his scientific attitudes.
is that exclusively NGF levels were affected by Much later, it was observed that variants of
such a “falling in love,” further demonstrating the maternal care in mammals can promote hippo-
unique role of this proteic neurotrophin (histori- campal synaptogenesis and spatial learning and
cally, the first prototype of this “family” of tro- memory through systems known to mediate
phic molecules) in regulating social bonding in experience-dependent neural ontogeny. Offspring
mammals, including human beings (Emanuele of highly competent rat mothers showing high
2011). Again, we refer to Black (1986), for the levels of pup licking and grooming and arched-
evolutionary meaning of neurotrophic bonding back nursing (the triadic ethogrammatic score to
and their likely taxonomic progression. evaluate maternal behavior in rodents) had
Rita Levi-Montalcini fell in love with NGF increased expression of mRNA BDNF and
several decades ago, when she astonishingly strengthened hippocampal cholinergic innerva-
observed the magnificent “halo effect” produced tion, spatial learning, and memory capability.
in vitro when even minimal amounts of this “bio- A variety of studies also revealed a role for
logically powerful” protein reached an explanted neurotrophic proteins as “mediators” of CNS and
sensory ganglion of the chick embryo, rather rap- peripheral nervous system (PNS) structural
idly stimulating massive dendritic growth. This changes, not limited to the early developmental
happened in Rio de Janeiro during the yearly phases. In fact, the continuous adult plasticity of
13 When Nerve Growth Factor Met Behavior 209

neuronal nets from synaptogenesis to trophic and than controls, had lowered levels of attack-type
differentiative phenomena is also molded by ethogrammatic elements, and showed a longer
neurotrophic actions. latency to attack counterpart (Bigi et al. 1992).
NGF administration inhibits a reliable indicator of
“ambiguous,” transitional element of the inter-
13.3 Social Milieu and NGF Release male aggressive pattern, i.e., aggressive
grooming, a behavioral item belonging to the
In general, when summarizing a vast variety of mouse aggressive repertoire consisting of
data, collected not exclusively on vertebrates, grooming the back of the opponent and indicative
including the human species, a “lack of controlla- of a marked motivation to attack. In fact, follow-
bility” could act to trigger NGF release or to ing repeated and elongated episode of aggressive
enhance its magnitude. Further evidence of the grooming, some veritable biting episodes tended
association between increased NGF levels and a to occur.
“subordinate-like” profile emerged from a study
performed long time ago in our laboratory. Lac-
tating female mice are known to display intraspe- 13.4 Adrenal Pathway
cific aggression patterns toward male intruders to
actively defend their litter. In fact, in the mouse One probable way by which NGF could exert a
species as in bears and in many feline species, physiological role seemed to be, since our first
male attack pups of lactating females to kill them. studies, via the adrenal gland, known since a long
When the physical and bio-behavioral acts time to be among the main targets of the NGF
involved in lactation (nipple stimulation, peptide released following intermale aggressive behavior.
release, maternal behaviors, etc.) are interrupted, Moreover, repeated (once for ten consecutive
the mother often undergoes estral phase; there- days) exogenous NGF administration results in
fore, the infanticidal adult male may eventually marked adrenal gland hypertrophy (Aloe et al.
fecundate her to spread his gene while eliminating 1986; Bigi et al. 1992). Repeated administrations
the genetic contribution of the former father, the (6–10 consecutive days) cause a dose-dependent
one that made the mother pregnant. increase in both adrenal weight and volume, med-
NGF levels in lactating female mice were pos- ullary and cortical layers undergoing a marked
itively correlated with the degree to which sex- volume and area increase.
and species-specific defensive behavioral patterns Level changes in the production of adrenal
were displayed by the mother: when confronting hormones (in particular, raised corticosterone
a potentially infanticide male intruder, lactating levels) resulting from increased levels of blood
females show the greater amount of defensive NGF could, in turn, affect the expression of
behavioral acts also displaying higher levels of aggressive behavior. Rodent data show that both
circulating plasma NGF (Alleva et al. 1996a). ACTH and glucocorticoid hormones affect ago-
Female aggression, usually of a very high inten- nistic behavior, yet the negative feedback action
sity and targeted at vulnerable (e.g., ventral, head) between these hormones reveals that their effects
parts of the body, is considered a seemingly are complex; also depending on the profile of
“uncontrollable” protective reaction of the mother their plasma release, glucocorticoids directly pro-
toward potentially dangerous predators of her mote submissiveness, hierarchical state well sig-
pups (Alleva 1993). These include adult male naled in rodents because of very evident postures
mice showing highly cannibalistic attacking and/or vocal emission; at the same time, they
behavior toward her litter. increase aggression indirectly through a negative
Moreover, systemic administration of physio- feedback suppression of ACTH synthesis.
logical levels of exogenous NGF causes a “sub- Leshner and colleagues (Leshner et al. 1980)
ordinate-like” profile in adult males. NGF-treated reported that ACTH facilitates submissiveness in
subjects spend proportionally less time fighting mice. A direct action of NGF on the peripheral
210 D. Santucci et al.

neural substrates regulating aggressive behavior It has however been demonstrated that admin-
can then be also hypothesized. istration of exogenous NGF promotes structural
NGF release may in fact exert some inhibitory recovery following specific hypothalamic dam-
feedback effect on aggressive behavior. The age; also, genes encoding NGF and its receptor
higher NGF release scored in subordinate (s) are expressed in the hypothalamus. Various
subjects (Maestripieri et al. 1990) and their hyper- lines of evidence support the hypothesis that
trophic adrenals led to hypothesize a “regulative hypothalamic NGF might be involved in neuro-
loop” involving NGF-promoted adrenal hormone endocrine function, thereby acting to regulate
trigger to stimulate and facilitate a submissive behavioral outcomes. Paracrine and/or autocrine
profile (Alleva et al. 1996a; Leshner et al. 1980). loops have been repeatedly suggested.
Such a proposed functional loop, maintained and The relevance of the observed changes in NGF
regulated by both by a finely tune interplay levels at the hypothalamic level upon intraspecific
between behavioral and neurobiological male “territorial” fighting and the mechanisms
components, was detailed in Alleva et al. 1996b. involved is however still a matter of speculation.
Other mouse studies lend further support to the Alleva and Aloe (1989) hypothesized that the
idea that NGF expression may be regulated by rapid increase in expression levels of brain
activation modulated by social behavior NGF, following a psychosocial stressful event,
(Spillantini et al. 1989). It was in fact reported could allow some subtle phenomena of adult
that intermale mouse fighting leads to a marked brain plasticity to rapidly being activated (see
increase in mRNA NGF production in hypotha- Figs. 13.2 and 13.3).
lamic neurons. Elevated levels of mRNA NGF, NGF could contribute to subtle structural
which followed a single, separated fighting epi- changes, such as emergence of dendritic spines
sode, were already evident at 30 min, peaked or collateral sprouting, eventually sculpturing the
about 3 h later, and declined to control levels structure of neural connections in the mature
between 6 and 12 h. Such a drastic increase in brain. In other words, NGF, as most likely other
mRNA NGF in the hypothalamic area was neurotrophins, represent a major, or at least not
paralleled by an increase in biologically active minor, key regulator, translating behavioral
NGF; notably, NGF antibodies markedly events or states in structural changes of neural
inhibited neurite outgrowth elicited from sensory architecture. It is however rather obvious that
and sympathetic ganglia from hypothalamic NGF expression in the hypothalamus might influ-
tissues extracted from isolated fighting mice. ence levels of other peptides and hormones pres-
Labeled cells were observed in the magnocellular ent in this structure, all cooperating in structural
preoptic and ventrolateral hypothalamic nuclei. changes in neural networking. A series of interac-
Importantly, no specific labeling of glial cells tive effects between NGF and thyroid hormones,
was observed in this or other brain regions ACTH, and peptides have, in fact, been reported
supporting the view that only neuronal cells since a long time.
were involved. The increase in hypothalamic
NGF following intermale aggressive behavior is
not abolished by sialoadenectomy, surgical abla- 13.5 The Most Famous Sibling: The
tion of the salivary glands, suggesting as expected Brain-Derived Neurotrophic
that the NGF scored in the hypothalamic area is (BDNF) Factor
not of salivary origin (Aloe et al. 1986).
Several lines of converging evidence, from BDNF appeared on the scientific scene much later
both intact and injured CNS structures, have than NGF.
shown that NGF is expressed in the hypothalamic It is rarely mentioned that any the
zones, although its definite roles in regulating “primigenial” NGF sources were tumor tissue,
these structures are still elusive (Spillantini et al. bull semen, and most importantly Naja naja
1989). snake “venom.” Reportedly (Rita
13 When Nerve Growth Factor Met Behavior 211

NON-PSYCHOSOCIAL
Unescapable footshock
Escapable footshock NO
Cold water swimming NO*
Restraint stress NO
Forced biting NO

PSYCHOSOCIAL
Male fighting YES (high, 100-300 ng/ml serum)
Female fighting YES (low, 3-13 ng/ml serum)
Pre-copula arousal YES (low, 2-9 ng/ml serum)
Running competition YES (prominent neurite outgrowth)

* In rats published observations indicated a decrease of NGF level and NGF receptor expression in the striatum following cold-water swimming stress while
an increase was shown in the hippocampus.
Aloe L, Tirassa P, Alleva E (1994) Cold water swimming stress alters NGF and low-affinity NGF receptor distribution in developing rat brain. Brain Res
Bull:173-178.
** Matsuda H, Koyama H, Oikawa M et al (1991) Nerve Growth Factor-like Activity detected in Equine Peripheral Blood after Running Exercise. J
Vet Med 38:557-559.

Fig. 13.2 Non-psychosocial and psychosocial contexts in which NGF is released in the bloodstream

Levi-Montalcini, in verbis), it occurred that the and available in laboratories source of NGF. It
biochemist Stanley Cohen, who shared in 1986 was indeed the case, and such a rich,
the Nobel Prize with RLM, had the enlightening concentrated, and anatomically “friendly” source
idea, by exploiting a purely “comparative” of the protein made it quite comfortable to extract,
approach to look at mouse submandibular “sali- to purify, and therefore to biochemically charac-
vary” glands, as the analogous structure of snake terize the protein; this last was Cohen’s job when
venom glands, in search of an easily accessible Rita hired him at Washington University. Many

Mouse/ Rat (nearly absent) submandibolar salivary gland

Snake Naja naja venom*

Bull, Rabbit, Camel, Llama, Alpa seminal plasma **

Horse unknown

*a neurotropic effect to enhance neuromotor impairment; Cohen S, Levi-Montalcini R (1956) A Nerve Growth-stimulating
factor isolated from snake venom. Proc Natl Acad Sci USA 42:572-574.
** Castellini C, Mattioli S, Dal Bosco A, et al. (2020) A, Role of NGF on sperm traits: A review, Theriogenology 150:210-214.

Fig. 13.3 NGF sources in various vertebrate species


212 D. Santucci et al.

years later, in Hans Thoenen laboratory, and by of high-frequency synaptic transmission, the
concentrating a rather large quantity of brain tis- neurotrophin BDNF is nowadays well known to
sue from pigs, the BDNF molecule was isolated stand out among members of the neurotrophin
and relatively rapidly characterized from a bio- family, for its faculty to regulate synaptic plastic-
chemical point of view. ity and, in particular, for generating sustained
Years later, NT3 and NT4 were added to the structural and functional changes at hippocampal
“taxonomical” unit named neurotrophins. synapses underlying learning and memory.
A pivotal study showed that the protein BDNF Recent advances show how BDNF function is
elicits dramatic sprouting of basal dendrites controlled and diversified by molecular and cellu-
associated with a concomitant regression of den- lar mechanisms including trafficking and subcel-
dritic spines. When compared to green fluorescent lular compartmentalization of different BDNF
protein (GFP)-transfected controls, such newly mRNA species, pre- vs postsynaptic release of
originated dendrites and spines were highly BDNF, control of BDNF signaling by tropomyo-
unstable. Experiments utilizing Trk receptor sin receptor kinase B (TrkB) receptor interactors,
antibodies, K252a, along with overexpression of and conversion of pro-BDNF to mature BDNF
NGF molecules demonstrated that such an ele- and BDNF propeptide (De Vincenti et al. 2019;
vated level of instability was due to secreted Yang et al. 2020). Altogether, those complex
BDNF interacting with extracellular TrkB mechanisms and processes provide general
receptors. In other words, BDNF promotes struc- framework of how neurotrophic activities regu-
tural instability in dendrites and spines. When late environmental inputs, coping responses, sen-
those connection bonds are restricted to particular sory motor integration, neuronal plasticity, and
portions of a dendritic arbor, they may well coop- neural tissue continuous reorganization.
erate in translating activity patterns into specific,
yet minute, brain structural changes.
In addition to the governing neuronal survival 13.6 Conclusion
and differentiation, neurotrophins reportedly play
a role in synapse development and plasticity. Despite reported, long-term claims in the biomed-
More in general, their trophic effects, addressing ical literature, few evidences are given for future
neural growth toward target in PNS and CNS therapeutical uses of neurotrophins; NGF, in par-
areas, have been demonstrated, mainly for NGF. ticular, was claimed a remedy for curing
Exposure to BDNF triggers long-term potenti- Alzheimer’s disease, Huntington’s disease,
ation (LTP) in CA1 synapses of the neonatal neuroimmune disturbances, etc. However, the
hippocampal zones, which otherwise display hyperalgesic side effect of systemically
only short-term potentiation. A putative, yet administered NGF, firstly recorded by us in
quite robust, explanation points to an attenuation mice (Levi-Montalcini et al. 1990) and by others
of synaptic fatigue induced by repeated high- in rats, is consistently emerging as a potential
frequency stimulation. Such a selective potentia- avenue for treating diabetic neuropathic pain or,
tion of high-frequency transmission by BDNF more in general, for chronic pain conditions.
appears to play a direct role correcting BDNF Coming to behavioral disturbances, there are
effects on LTP, rather than indirectly inducing series of clinical, preclinical, and animal studies
release of additional diffusible factors. The pref- assuming a role for neurotrophic growth factors
erential BDNF potentiation of highly active (GFs) in the etiology and drug treatment of clini-
synapses may well be implicated in the cal conditions related to human mood disorders.
Hebbian-type mechanisms involved in synaptic Convergent evidence accumulated in the last
plasticity. decades indicates that neuroplastic mechanisms
Following the report by Gottschalk et al. involving GFs (and BDNF in particular) are in
(1999), who initially described the biologically some way altered in depressive disorders and in
transducing events mediating BDNF modulation animal models of stress.
13 When Nerve Growth Factor Met Behavior 213

More in general, all data overall substantiate Acknowledgments The authors would like to acknowl-
the view that GF plays an important role in the edge Stella Falsini and Francesco Ciabattoni for format
and critical reading of the manuscript. We would also
pathophysiology of several psychiatric disorders express our deep gratitude to Dr. Luigi Aloe for sharing
and in the reinterpretable mechanism of action of his passion for science with us.
psychotropic drugs and that, conversely, drugs
that selectively stimulate the production of
neurotrophins might represent a new generation
of, e.g., antidepressants. References
The present contribution, which essentially
Alboni S, van Dijk RM, Poggini S et al (2017) Fluoxetine
summarizes some decades of experimental work effects on molecular, cellular and behavioral
mainly carried out by exploiting animal models endophenotypes of depression are driven by the living
and, among them, the mouse for its species- environment. Mol Psychiatry 22(4):552–561. https://
doi.org/10.1038/mp.2015.142
specific social structure (Bronson 1979), was
Alleva E (1993) Assessment of aggressive behaviour in
prominent when compared with the various rat rodents. In: Conn PM (ed) Paradigms for the study of
strains, this latter species being in fact behavior, vol 14. Academic Press, New York, pp
characterized by a colonial-type social organiza- 111–137
Alleva E, Aloe L (1989) Physiological roles of nerve
tion (Alleva et al. 1995).
growth factor in adult rodents: a biobehavioral perspec-
However, after several years of animal studies, tive. Int J Comp Psychol 2:147–162
a progressive amount of evidence was Alleva E, Calamandrei G (1990) On the functional role of
accumulated for the human species, where NGF polypeptide growth factors in rodent neurobehavioral
development. Acta Neurobiol Exp (Wars) 50
and the other neurotrophins BDNF, NT3, and
(4–5):341–352
NT4 play physiological and pathophysiological Alleva E, Fasolo A, Lipp HP, Nadel L et al (1995)
roles rather similar to that observed in Behavioural brain research in naturalistic and semi-
non-human mammals. naturalistic settings: possibilities and perspectives.
Kluwer Academic Publishers, Dordrecht, p 466
We just mention that Branchi and co-workers
Alleva E, Aloe L, Cirulli F et al (1996a) Serum NGF levels
(Alboni et al. 2017) have much more recently increase during lactation and following maternal
accumulated sound evidences that the renewed aggression in mice. Physiol Behav 59(3):461–466.
brain plasticity and the continuous neural network https://doi.org/10.1016/0031-9384(95)02083-7
Alleva E, Petruzzi S, Cirulli F et al (1996b) NGF regu-
reorganization, and even minute processes such
latory role in stress and coping of rodents and humans.
as synaptogenesis, are involved in common Pharmacol Biochem Behav 54(1):65–72. https://doi.
human psychopathological disorders (mainly, org/10.1016/0091-3057(95)02111-6
depression), providing a framework for an origi- Aloe L, Alleva E, Böhm A et al (1986) Aggressive behav-
ior induces release of nerve growth factor from mouse
nal reinterpretation for these psychiatric
salivary gland into the bloodstream. Proc Natl Acad
conditions. Sci U S A 83(16):6184–6187. https://doi.org/10.1073/
Perhaps more importantly, these lines of evi- pnas.83.16.6184
dence provided new insights on the recognized Bigi S, Maestripieri D, Aloe L et al (1992) NGF decreases
isolation-induced aggressive behavior, while increas-
variable therapeutical efficacy of the most com-
ing adrenal volume, in adult male mice. Physiol Behav
mon antidepressant drugs, presently prescribed 51(2):337–343. https://doi.org/10.1016/0031-9384
very commonly. The very high proportion of (92)90150-z
non-responder patients can hopefully benefit of Black IB (1986) Trophic molecules and evolution of the
nervous system. Proc Natl Acad Sci U S A 83
these new interpretative steps elucidating opaque
(21):8249–8252. https://doi.org/10.1073/pnas.83.21.
aspects of neurotrophin-served mechanisms and 8249
processes linking specific social and non-social Branchi I, Francia N, Alleva E (2004) Epigenetic control
behavioral patterns, synthesis, storage, and of neurobehavioural plasticity: the role of
neurotrophins. Behav Pharmacol 15(5–6):353–362.
release of proteic agents and structural brain
https://doi.org/10.1097/00008877-200409000-00006
(and sometimes PNS) plasticity. Bronson FH (1979) The reproductive ecology of the house
mouse. Q Rev Biol 54(3):265–299. https://doi.org/10.
1086/411295
214 D. Santucci et al.

Cavalli-Sforza LG (1996) Geni, popoli e lingue. Adelphi, Engl 26(8):707–716. https://doi.org/10.1002/anie.


Milano, p 354 198707073
De Vincenti AP, Ríos AS, Paratcha G et al (2019) Levi-Montalcini R (1987b) The nerve growth factor:
Mechanisms that modulate and diversify BDNF thirty-five years later. EMBO J 6(5):1145–1154
functions: implications for hippocampal synaptic plas- Levi-Montalcini R, Aloe L, Alleva E (1990) A role for
ticity. Front Cell Neurosci 13:135. https://doi.org/10. nerve growth factor in nervous, endocrine and immune
3389/fncel.2019.00135 systems. Prog Neuro-Endocrinol Immunol 3:1–10
Diamond J (1998) Armi Acciaio e Malattie, Breve storia Lorenz K (1935) Der Kumpan in der Umwelt des Vogels,
del mondo negli ultimi tredicimila anni. Einaudi, J. Orn. In: Schiller C (ed) Instinctive behavior, vol 83.
Torino, p 366 International University Press, New York, pp 137–289
Emanuele E (2011) NGF and romantic love. Arch Ital Biol Maestripieri D, De Simone R, Aloe L et al (1990) Social
149(2):265–268. https://doi.org/10.4449/aib.v149i2. status and nerve growth factor serum levels after ago-
1367 nistic encounters in mice. Physiol Behav 47
Fox WM (1965) Reflex-ontogeny and behavioural devel- (1):161–164. https://doi.org/10.1016/0031-9384(90)
opment of the mouse. Anim Behav 13(2):234–241. 90056-a
https://doi.org/10.1016/0003-3472(65)90041-2 Rose SPR (1981) From causations to translations: what
Gottschalk WA, Jiang H, Tartaglia N et al (1999) Signal- biochemists can contribute to the study of behavior. In:
ing mechanisms mediating BDNF modulation of syn- Bateson PPG, Klopfer PH (eds) Perspectives in ethol-
aptic plasticity in the hippocampus. Learn Mem 6 ogy. Springer, Boston, MA. https://doi.org/10.1007/
(3):243–256 978-1-4615-7575-7_8
Hadjiconstantinou M, McGuire L, Duchemin AM et al Spillantini MG, Aloe L, Alleva E et al (1989) Nerve
(2001) Changes in plasma nerve growth factor levels growth factor mRNA and protein increase in hypothal-
in older adults associated with chronic stress. J amus in a mouse model of aggression. Proc Natl Acad
Neuroimmunol 116(1):102–106. https://doi.org/10. Sci U S A 86(21):8555–8559. https://doi.org/10.1073/
1016/s0165-5728(01)00278-8 pnas.86.21.8555
Lang UE, Gallinat J, Kuhn S et al (2002) Nerve growth Torcia M, Bracci-Laudiero L, Lucibello M et al (1996)
factor and smoking cessation. Am J Psychiatry 159 Nerve growth factor is an autocrine survival factor for
(4):674–675. https://doi.org/10.1176/appi.ajp.159.4. memory B lymphocytes. Cell 85(3):345–356. https://
674-a doi.org/10.1016/s0092-8674(00)81113-7
Leshner AI, Korn SJ, Mixon JF et al (1980) Effects of Ursin H, Eriksen HR (2004) The cognitive activation
corticosterone on submissiveness in mice: some tem- theory of stress. Psychoneuroendocrinology 29
poral and theoretical considerations. Physiol Behav 24 (5):567–592. https://doi.org/10.1016/S0306-4530(03)
(2):283–288. https://doi.org/10.1016/0031-9384(80) 00091-X
90087-6 Yang T, Nie Z, Shu H et al (2020) The role of BDNF on
Levi-Montalcini R (1987a) The nerve growth factor: neural plasticity in depression. Front Cell Neurosci
35 years later (Nobel lecture). Angew Chem Int Ed 14:82. https://doi.org/10.3389/fncel.2020.00082
Cross Talk of BDNF and GDNF in Spinal
Substantia Gelatinosa (Lamina II): Focus 14
on Circuitry

Adalberto Merighi

Abstract functional interplay in the regulation of


BDNF and GDNF display the notable qualities nociception and pain.
of undergoing a regulated secretion in neurons
and being anterogradely transported to nerve
terminals, where they can modulate fast syn-
aptic transmission. That BDNF positively 14.1 Introduction
modulates nociception and/or pain is today
widely accepted, as the growth factor can Neurotrophic factors are an assorted group of
start and maintain physiological and patholog- molecules supporting the development, mainte-
ical pain. The contribution of GDNF to nance, and differentiation of immature and
nociception is by far most elusive, but evi- mature neurons. Some neurotrophic factors also
dence is accumulating that the molecule affect synaptic activity and plasticity and thus
displays analgesic activity, at least in rodents. fulfil the criteria to be included in the group of
Here I resume the current knowledge on the neuromodulators (Gibon and Barker 2017). Sev-
spinal cord circuits in which these two factors eral neurotrophic factors have received attention
may act as modulators of pain-related synaptic as modulators of nociception and players in the
transmission, focusing on their structural and maladaptive changes leading to chronic pain

A. Merighi (*)
Department of Veterinary Sciences, University of Turin,
Grugliasco, Italy
e-mail: adalberto.merighi@unito.it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 215
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_14
216 A. Merighi

under pathological conditions (Khan and Smith Splicing variants exist for TrkB and TrkC.
2015; Ossipov 2011). The nerve growth factor These truncated receptors are devoid of the tyro-
(NGF) was the first to be studied as a spinal sine kinase domain but still can evoke Ca2+
nociceptive modulator (Merighi et al. 2004), but transients and trigger intracellular signals (Blum
afterward, a much higher interest was raised by and Konnerth 2005). Interestingly, several studies
another neurotrophin, the brain-derived indicate that truncated Trk isoforms typically
neurotrophic factor (BDNF), on the basis of sev- counteract the function of the respective full-
eral histological, functional, and pharmacological length receptors, e.g., by inhibiting dimerization
observations (Merighi et al. 2004; Merighi et al. or disrupting intracellular responses (Gold and
2008b; Bardoni and Merighi 2009). More Gebhart 2010).
recently, evidence has been accumulating also The low-affinity p75NTR also is made of an
for an intervention of the glial cell line-derived extracellular and an intracellular domain. The
neurotrophic factor (GDNF) in the modulation of latter includes a death domain acting on several
spinal nociception (Merighi 2016). Here I recon- cytoplasmic proteins. Expression of p75NTR
sider the spinal cord circuitry in relation to the decreases in parallel with development but
intervention of BDNF and GDNF in the modula- increases after injury (Mogil 2019). p75NTR does
tion of the physiological processing of nocicep- not directly activate the neurotrophins but
tive stimuli and in the maladaptive changes that interacts with Trks giving rise to several different
occur in pathological conditions. downstream effects (Mogil 2019).

14.1.1 BDNF and Its Receptors 14.1.2 GDNF and Its Receptors

BDNF, like the other members of the GDNF is a member of a family of related ligands,
neurotrophin family, i.e., NGF, neurotrophin-3 referred to as GDNF family of ligands (GFLs)
(NT-3), and neurotrophin-4/neurotrophin-5 that also consists of neurturin, artemin, and
(NT-4/NT-5), interacts with two different types persephin. All members of the family have struc-
of membrane receptors: a group of tyrosine kinase tural links with the components of the superfam-
receptors, referred to as tropomyosin receptor ily of the transforming growth factor β
kinases (Trks), and a common receptor, p75NTR (Airaksinen and Saarma 2002). GFLs signal
(Chao and Hempstead 1995). through a multicomponent receptor complex
There are three different Trk isoforms, named consisting of the tyrosine kinase transmembrane
TrkA, TrkB, and TrkC, with different affinities receptor Ret and a high-affinity ligand-binding
for the various neurotrophins: NGF preferentially glycosylphosphatidylinositol-anchored
binds to TrkA, BDNF and NT-4/NT-5 bind to co-receptor referred to as GDNF family receptor
TrkB, and NT-3 is associated with TrkC but α (GFRα). Each molecule of the GFL family ties
also binds to TrkA and TrkB, even though with with a specific GFRα isoform, GFRα1 in the case
a low affinity (Chao and Hempstead 1995). Trk of GDNF, characterized by a distinct structure of
receptors have an extracellular domain that binds the cysteine-rich repeats in the ligand-binding
the neurotrophin(s) and an intracellular domain domains (Cervero and Laird 1999). GFRα-Ret
that possesses tyrosine kinase activity (Reichardt activation mainly signals through Ras/MAPK,
2006). BDNF binding prompts receptor dimeriza- phosphatidylinositol 3-kinase (PI3K)/Akt, and
tion, phosphorylation of the cytoplasmic tyrosine PLCγ, which play a key role in neurite outgrowth
residues, and activation of downstream effectors and cell survival in the nervous system
(Koerber 2009). However, Trk receptors may be (McMahon et al. 1995; Gebhart 2000; Sengupta
also active in the absence of the neurotrophins, 2009).
following trans-activation via several G protein- Consistent with a broader expression of GFRα
coupled receptors (Reichardt 2006). than Ret in neurons, GFLs can also transduce
14 Cross Talk of BDNF and GDNF in Spinal Substantia Gelatinosa (Lamina II): Focus on Circuitry 217

through GFRα independently from Ret (Sengupta et al. 2008; Pan and Pan 2004), and mouse
2009). (Woodbury et al. 2000), lamina II can be further
subdivided into two zones (sublaminae), an outer
and inner lamina II (lamina IIo and IIi). In rodents,
14.2 General Features lamina IIi further consists of a dorsal (IIid) and a
of the Substantia Gelatinosa ventral (IIiv) part that display differences in the
type(s) of interneurons herein contained, their
The substantia gelatinosa of Rolando covers the neurochemistry, and connections with the
apex of the posterior (dorsal) horn of the spinal endings of PAFs (Ribeiro-Da-Silva and De
gray matter and owes its name to its gelatinous, Koninck 2008). The input to the substantia
semitransparent aspect in fresh preparations of the gelatinosa not only derives from the PAFs
spinal cord (see, for review, Merighi 2018). It originating from the DRG neurons but also from
consists of small neurons and glia and a rich the descending fibers deriving from several areas
neuropil but very few myelinated fibers. In of the brain and other spinal cord neurons, among
Rexed’s laminar subdivision of the spinal gray which are the antenna neurons (Schoenen 1982).
matter, the substantia gelatinosa corresponds to These latter are interlaminar neurons with cell
lamina II. Although dissimilar in shape and exten- bodies in laminae III–IV, dendrites that travel
sion, lamina II can be detected at all spinal levels dorsally into lamina II and lamina I, and, at least
and constitutes most of the dorsal part of the head in some cases, axons that go up to the brain along
of the dorsal horn. The gelatinosa receives a direct the STT (Surmeier et al. 1988) or the dorsal
input from the primary afferent fibers (PAFs) columns (Brown and Fyffe 1981). The output of
originating from the primary sensory neurons lamina II chiefly reaches neurons in laminae I, III,
(PSNs) of the dorsal root ganglia (DRGs) and and IV (Bennett et al. 1980; Gobel 1975, 1978a;
reaching the spinal cord through the dorsal roots Light and Kavookjian 1988), but, at least in rat
of the spinal nerves. A substantial fraction of (Giesler et al. 1978) and monkey (Willis et al.
these fibers derives from nociceptors and 1978), there are projections also to the brainstem
thermoreceptors, but some others originate from or the thalamus.
low-threshold mechanoreceptors (LTMs).
Neurons form a fine network inside lamina
II. Their short axons make synapses with each 14.2.1 Neurons of the Substantia
other or with neurons in other laminae of the Gelatinosa
gray matter, including the contralateral lamina
II. Notably, rather than directly contributing pro- In lamina II reside several populations of
jection fibers to the ascending anterolateral sys- interneurons (Merighi 2018). These neurons are
tem or the spinothalamic tracts (STTs) that intralaminar, i.e., they have processes that do not
convey nociceptive information to higher centers, span outside lamina II, or interlaminar, i.e., they
the substantia gelatinosa controls nociception at connect different laminae of the spinal cord
the interface between first-order DRG neurons dorsal horn.
and second-order projection neurons in laminae The islet cells form the best described distinct
I and III–V of the spinal cord. The publication of population of inhibitory neurons, based on their
the Gate Theory of Pain (Melzack and Wall 1965) morphology, neurochemistry, and function. They
was seminal to sustain the pivotal role of lamina II are GABA-, glutamic acid decarboxylase (GAD)-
in the control of pain, but more recent studies , and vesicular GABA transporter (VGAT)-
have implicated the substantia gelatinosa also in immunoreactive and form GABAergic synapses
the integration of pruritic stimuli. (Lu and Perl 2003; Zheng et al. 2010). The islet
In humans (Schoenen 1982), monkey (Light cells are solely inhibitory, as demonstrated by the
and Perl 1979), cat (Light and Perl 1979; Rexed lack of expression of the vesicular glutamate
1952, 1954), rat (Light and Perl 1979; Lorenzo transporter 2 (VGLUT2) gene (Punnakkal et al.
218 A. Merighi

2014). They chiefly display a tonic firing pattern Functionally, stalked/large vertical cells may be
and are presynaptic to transient-firing central cells either nociceptive-specific or wide dynamic range
(Lu and Perl 2003) and postsynaptic to C and Aδ (WDR) neurons (Bennett et al. 1980). They also
PAFs (Grudt and Perl 2002; Hantman et al. 2004; receive excitatory and inhibitory inputs from both
Lu and Perl 2003, 2005; Yasaka et al. 2007) and C (directly) and Aδ (indirectly) PAFs (Grudt and
to the central cells (Zheng et al. 2010). C PAFs Perl 2002; Hantman et al. 2004; Lu and Perl
excite the islet cells directly, whereas Aδ PAFs 2003; Yasaka et al. 2007). Based on earlier ultra-
inhibit them indirectly, via a polysynaptic path structural observations, it was inferred that PAFs
(Yasaka et al. 2007). Remarkably, islet cells contact the dendrites of large vertical cells specif-
establish dendro-axonic synapses onto the PAF ically at type Ia glomeruli (Maxwell et al. 2007),
C boutons at V1 profiles in glomeruli (Ribeiro- but this has, at present, only remained speculative
da-Silva 2015). This is, unfortunately, one of the and does not exclude that contacts occur also at
few information that we have about the origin of other types of glomeruli. The dendritic spines of
glomerular dendrites, as we will discuss in a the large vertical cells form several synapses with
following section of this chapter. Local inputs to VGLUT1-immunoreactive boutons. At least a
the islet cells are both excitatory and inhibitory few of these synapses possibly originate from A
and derive almost exclusively from other LTMs (Yasaka et al. 2014).
substantia gelatinosa neurons (Kato et al. 2007). The small vertical cells are GABAergic,
Central cells look like islet cells but are GAD-immunoreactive inhibitory neurons mainly
smaller. They may be glutamatergic (Lu and residing in lamina IIo (Grudt and Perl 2002;
Perl 2003, 2005) or GABAergic (Hantman et al. Maxwell et al. 2007; Yasaka et al. 2007). They
2004; Hantman and Perl 2005; Maxwell et al. have a smaller dendritic tree than the larger
2007; Todd and McKenzie 1989; Zheng et al. glutamatergic vertical cells. Another difference
2010) and show various firing patterns (tonic, is that the small vertical cells display a tonic firing
transient IA, and transient non-IA). pattern instead of delayed firing. At least some
Glutamatergic tonic and transient non-IA central small vertical cells expressing dynorphin have
cells synapse onto the vertical cells (Lu and Perl been linked to mechanical nociception (Cruz
2003, 2005), whereas GABAergic tonic central and Basbaum 1985; Duan et al. 2014).
cells make synapses onto vertical and islet cells Radial cells are excitatory glutamatergic
(Hantman et al. 2004; Zheng et al. 2010). Central neurons that have a characteristic shape with
cells connect indirectly to PAFs. They receive an compact discoidal dendritic trees and IA firing
excitatory or inhibitory synaptic input from C (Grudt and Perl 2002; Yasaka et al. 2007; Yasaka
PAFs, but a merely inhibitory input from Aδ et al. 2010). Some of them are instead inhibitory,
PAFs (Grudt and Perl 2002; Hantman et al. have a more dispersed dendritic tree, and are
2004; Lu and Perl 2003; Yasaka et al. 2007). immunoreactive for GABA (Maxwell et al.
The stalked cells are a rodent subpopulation of 2007; Todd and McKenzie 1989). C PAFs
large vertical cells. Many of them are (directly) and Aδ PAFs (indirectly) provide excit-
glutamatergic excitatory interneurons with atory and inhibitory inputs to the radial cells
delayed firing (Lu and Perl 2005) that are immu- (Grudt and Perl 2002; Hantman et al. 2004; Lu
noreactive for VGLUT2, but not GABA. At least and Perl 2003; Yasaka et al. 2007).
some large vertical cells contain one or more
neuropeptides (Bennett et al. 1982; Cruz and
Basbaum 1985; Ribeiro-Da-Silva et al. 1991; 14.3 Nociceptive Circuits
Todd et al. 2003). They are interlaminar neurons, in Substantia Gelatinosa
and those cells having their cell body in lamina IIo
are chiefly presynaptic to lamina I spinothalamic Separate routes in mammals process nociceptive
nociceptive neurons (Gobel 1978b) expressing stimuli conveyed from somatic or visceral organs
the NK1 receptor (Polgár et al. 2010). to cortical centers. In particular, somatic sensory
14 Cross Talk of BDNF and GDNF in Spinal Substantia Gelatinosa (Lamina II): Focus on Circuitry 219

fibers from the skin and the locomotor apparatus 14.3.2 Localization of BDNF and Its
follow the trigeminal and spinal nerves, whereas Preferred Receptor TrkB1
sensory nerves in viscera originate from the sym- in DRGs and Dorsal Horn
pathetic (via the white communicating branches of the Spinal Cord
of the thoracic and lumbar nerves) and parasym-
pathetic (via certain cranial nerves and the Initial localization studies demonstrated, in sev-
splanchnic pelvic nerves) autonomic nervous sys- eral species, the presence of the BDNF mRNA
tem. Visceral afferents, for the most, do not reach and protein in a subpopulation of PSNs of the
the substantia gelatinosa, but terminate in deeper DRGs (Ernfors et al. 1992; Josephson et al.
laminae of the spinal cord dorsal horn (Cervero 2001; Widenfalk et al. 2001). In laboratory
and Connell 1984). However, lamina II contains a rodents, BDNF+ DRG neurons are for the most
few terminals of visceral origin, which are sparser of small to medium size (Michael et al. 1997;
and span over a larger number of spinal segments Salio et al. 2005; Salio and Ferrini 2016) and
than the cutaneous afferents (Sugiura et al. 1989). peptidergic, i.e., they also express the calcitonin
Given the primary importance of lamina II in the gene-related peptide (CGRP), which typically
processing of somatic stimuli, I will focus the characterizes the nociceptors encoding thermal
following discussion on this matter. pain (Groenewegen 1988; Bushnell and Duncan
1989), alone or in combination with other
peptides. On the other hand, the non-peptidergic
PSNs, a set of nociceptors chiefly involved in the
14.3.1 Spinal Somatic Nociceptive
transduction of mechanical pain, which can be
Circuits
labelled using the isolectin B4 (IB4), do not
express BDNF (Groenewegen 1988; Bushnell
The PSNs of the DRGs collect nociceptive and
and Duncan 1989). Accordingly, BDNF-positive
other types of sensory stimuli from all parts of the
PAFs in the superficial dorsal horn display the
body except the head and the proximal regions of
same localization of the CGRP-expressing PAFs
the neck, which are innervated by the PSNs of the
in lamina I and lamina IIo (Salio and Ferrini 2016;
trigeminal nerve. The main somatic pain pathway
Conner et al. 1997). BDNF has also been detected
originating from the DRG neurons is the
in PSNs other than those labelled after CGRP or
spinothalamic pathway that is crucial for the sen-
IB4 immunostaining (Salio and Ferrini 2016). In
sory discriminative aspects of pain perception, in
keeping with this observation, by using a BDNF-
other words for the animal to be aware of which
LAcZ reporter transgenic mouse, Basbaum’s
part of the body is suffering pain. It consists of a
group has confirmed a wider expression of
polysynaptic chain of three key neurons, often
BDNF than formerly reported (Borsook and
referred to as the first-, second-, and third-order
Becerra 2011). Specifically, these authors
sensory neurons. First-order neurons are the PSNs
described the occurrence of the neurotrophin in
of the DRGs. Second-order neurons lie in laminae
large myelinated PAFs, suggesting a still poorly
I and V of the dorsal horn of the spinal cord. They
understood role of BDNF also in the processing
give rise to axons that cross the midline and
of non-nociceptive stimuli (Salio and Ferrini
ascend all along the spinal cord and brainstem to
2016).
reach the thalamus. Finally, the ventral postero-
The TrkB protein and mRNA are detectable in
lateral nucleus of the thalamus contains the third-
DRG neurons of variable size (Josephson et al.
order sensory neurons that eventually send their
2001; Widenfalk et al. 2001; Salio et al. 2005;
axons to the cerebral cortex. Modulation of noci-
Thompson and Bushnell 2012). Among these
ceptive signals can occur all along this
trkB+ DRG neurons, we have shown a subset of
polyneuronal chain but takes place mainly at the
level of the substantia gelatinosa (Merighi 2018).
1
Unless otherwise stated, I will refer to full-length func-
tional trkB receptors in this and in the following sections.
220 A. Merighi

CGRP+/BDNF+ nociceptors, suggesting the GFRα1 receptors are expressed by IB4+ PAFs
occurrence of an autocrine activation loop at that, in lamina II, lay ventrally to the GDNF+
their terminals in the substantia gelatinosa (Salio peptidergic terminals (Salio and Ferrini 2016;
et al. 2005). By using a transgenic TrkBtauEGFP Salio et al. 2014).
mouse, the trkB receptor was also demonstrated There is not convincing proof for a
in Aδ LTMs (Li et al. 2011), which, however, propriospinal source of GDNF. On the other
mainly terminate in lamina III of the dorsal horn. hand, GFRα1/Ret are expressed by some dorsal
Sparse propriospinal neurons express BDNF horn neurons, comprising a population of sup-
and/or TrkB. The BDNF mRNA was localized posed projection neurons in lamina I that also
only in the deep dorsal horn of the adult rat express the substance P-preferred receptor NK1
(Widenfalk et al. 2001; Conner et al. 1997). Con- (Jongen et al. 2007).
versely, most spinal cord neurons express the
trkB protein (Widenfalk et al. 2001; Bradbury
et al. 1998). 14.3.4 Role of BDNF and GDNF
in Nociception

14.3.3 Localization of GDNF Although localization studies in PSNs and


and GFRa1/Ret in DRGs substantia gelatinosa powerfully advocate an
and the Dorsal Horn involvement of both BDNF and GDNF in
of the Spinal Cord nociception, the functional relevance of the two
trophic factors in pain physiology remains elu-
Like BDNF, GDNF is chiefly expressed by small- sive. The main reasons for this have up to now
to medium-sized DRG neurons (Salio and Ferrini been the scarcity of knowledge of the spinal cord
2016; Bennett et al. 1998), although the GDNF- circuits made by the two trophic factors and their
expressing neurons form a distinct and smaller receptors, the nonexistence of specific drugs
subpopulation of peptidergic neurons than that targeting the two molecules, as well as the limited
expressing BDNF (Salio and Ferrini 2016). In information that could be obtained from standard
mouse, GDNF+ DRG neurons also express knockout animals. More recently, on the other
CGRP and somatostatin (Salio and Ferrini hand, the progress and readiness of conditional
2016). The central projections of these neurons mutant mice have opened new paths to question
terminate in laminae I–IIo (Salio and Ferrini the influence of BDNF and GDNF in pain
2016; Holstege et al. 1998; Ohta et al. 2001; transmission.
Salio et al. 2014). Studies on the conditional blockade of a
The RET mRNA is detected in more than 50% mutant TrkB receptor in mice concluded that
of the lumbar DRG neurons (Bennett et al. 1998, BDNF did not intervene in the processing of
2000; Kashiba et al. 2003). More than two thirds acute heat, mechanical, or chemical pain sensitiv-
of the RET-expressing lumbar (Kashiba et al. ity. The role of BDNF in nociception was also
2003; Molliver et al. 1997) or thoracic (Josephson studied in a conditional transgenic mouse strain in
et al. 2001) neurons also display the GDNF- which a tamoxifen injection could selectively
specific co-receptor GFRα1 at their membrane. destroy virtually all PSNs expressing BDNF
Remarkably, while the DRG peptidergic neurons (Borsook and Becerra 2011). In these mice, only
mainly express GDNF, it is the non-peptidergic minor effects could be observed, such as an unim-
IB4+ neurons that express the GFRα1/Ret recep- portant increase in the heat thresholds in males
tor complex (Molliver and Snider 1997). but not females, and no effects on mechanical or
Consistent with the expression of GFRα1/Ret cold sensitivity, irrespective of gender (Borsook
in non-peptidergic PSNs, GDNF receptors have and Becerra 2011).
been detected in PAFs of lamina IIi (Salio et al. A different situation follows the ablation of
2014; Jongen et al. 2007). Ultrastructurally, Ret by conditional deletion in nociceptors
14 Cross Talk of BDNF and GDNF in Spinal Substantia Gelatinosa (Lamina II): Focus on Circuitry 221

expressing the Nav1.8 channel (Golden et al. 14.4.1 BDNF and the Excitatory Path
2010): in this case, mice display an augmented
sensitivity to cold and an augmented response to The occurrence or lack of TrkB receptors allows
formalin-induced pain, thus indicating a constitu- for further subdividing BDNF containing PAFs
tive inhibitory role of Ret in spinal nociception. into two subcategories. The first, where TrkB
As I will discuss in the following paragraph, the receptors are absent, is crucial to generate a
findings ex vivo obtained in my laboratory in long-lasting excitation in lamina IIo following
wild-type animals (Salio et al. 2014) are fully two different paths that ultimately converge onto
consistent with these observations in the gelatinosa large vertical/stalked cells. The first
transgenic mice. path (Fig. 14.1—path 1) comprises a series of
three excitatory synapses. The first synapse is
made by a peptidergic BDNF+ C fiber that also
expresses substance P and the transient receptor
14.4 BDNF and GDNF Circuits
potential vanilloid receptor 1 (TRPV1). It initiates
in the Substantia Gelatinosa
the circuit by providing input to an excitatory
central cell in lamina II. The central cell, in turn,
Neuronal circuitries in lamina II and other
is presynaptic to a large vertical cell, which sends
laminae of the superficial dorsal horn remain,
its axon outside lamina II to make the third excit-
for the most, vague. Not only one of the main
atory synapse of the path by contacting a STT
reasons for this substantial lack of information is
projection neuron in lamina I. The other path
our present difficulty to identify functionally dis-
(Fig. 14.1—path 2) also starts with a peptidergic
tinct populations of interneurons, but also the
BDNF+ C fiber but is more complex as it consists
unfeasibility to provide a complete reconstruction
of a chain of four neurons that includes two
of the exact wiring of the substantia gelatinosa
inhibitory islet cells in series that are inserted
neurons has an unquestionable influence. Here I
between the C PAF and the large vertical cell.
try to put together and discuss the data collected
Thus, as the first islet cell drives an inhibition of
in my laboratory with those obtained by other
the second islet cell, the ultimate result is an
colleagues on the circuits made by the PAFs
excitation of the large vertical/stalked cell onto
containing BDNF or GDNF in lamina II and
which the path also converges. As PAFs involved
propose a hypothetical model of parallel
in both circuits contain a blend of messengers
processing of somatosensory information based
with different molecular weight, release can
on the interplay between the two molecules.
occur selectively in dependency of the frequency
According to this model, there are two pathways
and firing pattern of activation (Merighi 2017). In
with opposite functions to process sensory infor-
nociceptive fibers, the firing pattern, rather than
mation in the gelatinosa. One of these is excit-
just the firing rate, is important to encode the
atory and is triggered by a subgroup of PAFs that,
intensity of noxious stimuli. Thus, it was
besides the fast excitatory transmitter glutamate,
demonstrated that, in dependence with the
may release BDNF together with other
strength of nociceptive stimulation of the C fibers,
pronociceptive modulators, chiefly substance
the pronociceptive modulator substance P may be
P. The other is, instead, inhibitory and involves
released together with glutamate above a certain
another subgroup of PAFs that release the fast
threshold of activation (Adelson et al. 2009).
transmitter glutamate but also GDNF together
Notably, substance P mediates the central sensiti-
with the anti-nociceptive modulator somatostatin
zation of nociceptors (Seybold 2009). Yet, NK1
(Merighi 2018).
receptors mainly lay outside the substantia
gelatinosa, and, thus, the peptide’s modulatory
222 A. Merighi

Fig. 14.1 Schematic representation of the circuits transported to terminals. Under basal conditions or in
modulated by BDNF and GDNF in the substantia nociceptive (physiological) pain, the peptidergic C fibers
gelatinosa of the spinal cord. C PAFs containing BDNF only release glutamate, but when a certain threshold of
(yellow) may be subdivided into two groups on the pres- stimulation is reached, they can also release BDNF or
ence or absence of TrkB receptors (autoreceptors). They GDNF with their co-stored neuropeptides according to
also contain the neuropeptides CGRP and substance the pattern and rate of C fiber activation (bottom).
P. The C PAFs containing GDNF (light blue) also contain Interactions chiefly occur at the level of lamina II synaptic
CGRP and somatostatin. Both BDNF and GDNF are glomeruli (colored circles). See text for further
synthesized by subsets of DRG neurons and anterogradely information
14 Cross Talk of BDNF and GDNF in Spinal Substantia Gelatinosa (Lamina II): Focus on Circuitry 223

effects in lamina II are supposed to be inexistent lamina II neurons that occurs after capsaicin-
or primarily extrasynaptic, i.e., taking place stimulated release of BDNF (Merighi et al.
through volume transmission (Liu and 2008a). The same pathway is also compatible
Sandkühler 1995). It is also worth of note that with the observed increase in the frequency of
substance P is co-stored with BDNF (and CGRP) GABAergic/glycinergic mIPSCs and reduction
in large dense core vesicles (LGVs) within of eIPSCs in lamina II neurons after BDNF chal-
peptidergic PAF terminals (Salio et al. 2007). lenge, under pharmacological block of excitatory
Therefore, one can reasonably suppose that the neurotransmission (Bardoni et al. 2007). This cir-
two modulators are co-released from these cuit thus may be the structural and functional
terminals upon LGV exocytosis (Merighi 2017). substrate for a disinhibition (i.e., excitation) of
Different from NK1 receptors, TrkB receptors the large vertical/stalked cells that is mediated
are highly expressed in lamina II, both pre- and synergistically by BDNF and substance P acting
postsynaptically (Salio et al. 2005). We have pre- as co-transmitters after co-release by a subset of
viously demonstrated that the neurotrophin can peptidergic PAFs. Additionally, it may be central
modulate sensory information in the substantia in the long-lasting central sensitization of the
gelatinosa with a combination of excitatory gelatinosa synapses under certain inflammatory
(Merighi et al. 2008a) and inhibitory (Bardoni conditions and/or neuropathic pain (Merighi
et al. 2007) effects. PAFs can trigger the activa- et al. 2008a, b).
tion of the STT projection neurons indirectly,
along a minimally bi-neuronal chain of two
gelatinosa excitatory neurons, a central and a 14.4.2 GDNF and the Inhibitory Path
vertical cell in sequence (Lu and Perl 2005;
Yasaka et al. 2007). It is remarkable that the The peptidergic PAFs containing GDNF
first- to second-order sensory neuron synapses in (together with somatostatin) arise from a subpop-
lamina II do not simultaneously express TrkB ulation of LA4-immunoreactive type C DRG
receptors pre- and postsynaptically (Salio et al. neurons that, before the discovery of GDNF-
2005). This indicates that the activation of den- expressing DRG neurons and the association of
dritic postsynaptic TrkB receptors takes place at GDNF with nociception, were assumed to have
simple axo-dendritic synapses or, more likely, at atypical neurochemical and functional features
the type Ib glomeruli (Fig. 14.1) made by the (Averill et al. 1995). These neurons are roughly
peptidergic PAFs (Salio et al. 2005). Alterna- equivalent in number to those expressing BDNF
tively, or in addition, BDNF can stimulate pre- and substance P (Merighi 2018). Their central
synaptic TrkB autoreceptors (Merighi et al. projections, i.e., the C PAFs formed by these
2008a) at a diverse subset of type Ib glomeruli cells, contain glutamate as the chief excitatory
(Fig. 14.1), as I will discuss later. As certain C neurotransmitter together with GDNF and
PAFs containing BDNF also express TRPV1 somatostatin that negatively modulate
(above), they can release BDNF following capsa- nociception. In keeping with the well-known
icin stimulation, i.e., a condition that mimics simultaneous presence of transmitters with
inflammation (Merighi et al. 2008a). Remarkably contrasting function in these fibers, a study has
and in accord with the electrophysiological data demonstrated that somatostatin-expressing PAFs
obtained after recordings from pairs of gelatinosa are pruritoceptive, rather than nociceptive, and
neurons or stimulating inhibitory neurons with that the peptide, instead, counteracts nociception
the neurotrophin, the tetra-synaptic pathway (Huang et al. 2018). Therefore, as the C fibers
involving the two islet cells (inhibition of inhibi- directly excite the islet, central, radial, and verti-
tion) (Fig. 14.1—path 2) may be the anatomical cal cells (Yasaka et al. 2007), it seems possible
substrate for the long-lasting excitation of several that, under basal conditions, the GDNF/
224 A. Merighi

somatostatin peptidergic C PAFs exert a tonic 14.4.3 Cross Talk Between BDNF
inhibition onto the projection neurons in lamina and GDNF in Nociceptive
I. The underlying circuitry (Fig. 14.1—path 3) Pathways
comprises a type Ib glomerulus made by the C
bouton of the GDNF/somatostatin C PAF and an As discussed in the previous sections, BDNF and
inhibitory central cell that forms an axo-dendritic GDNF are expressed by two distinct populations
synapse onto the vertical cell and, thus, reduces of peptidergic nociceptors (Salio and Ferrini
the outflow of excitatory information from lamina 2016), and, while absolutely pronociceptive or
II. Like it is the case for BDNF/substance P C anti-nociceptive effects cannot be specifically
PAFs, tonic inhibition through this circuit may be linked to one or the other neurotrophic factor,
modulated by the differential release of the fast there is a broad agreement that in neuropathic
transmitter glutamate and the slow-acting pain, BDNF is pronociceptive, while GDNF is
modulators GDNF and somatostatin according anti-nociceptive (Merighi et al. 2008b; Merighi
to the intensity of the C fiber activation. GDNF 2018; Merighi 2016). Many studies in the previ-
could also hyperpolarize the C boutons of IB4+ ous years have discovered how pathological
non-peptidergic PAFs expressing GFRα1 at type situations disturb the expression of BDNF and
Ia glomeruli. The ensuing experimental proofs GDNF in the nervous system, thus proving the
support such a possibility: (1) somatostatin intervention of both molecules in the shift from
promotes central analgesia (Pintér et al. 2006) physiology to pathology. In several
via tonic inhibition of C and Aδ PAFs (Guo circumstances, the levels of the two factors
et al. 2008; Wang et al. 2009) and decrease of change along divergent directions, an observation
the glutamate-dependent stimulation of these that underpins the notion that BDNF and GDNF
fibers (Luo et al. 2010); (2) GDNF, binding to play alternative/complementary roles in the noci-
GFRα1, exerts a sustained inhibition of the ceptive system. Nonetheless, only few studies
glutamatergic excitatory drive of the gelatinosa have focused onto the direct interactions between
neurons (Salio et al. 2014); and (3) the large BDNF and GDNF in the modulation of central
vertical cells are postsynaptic to non-peptidergic neurotransmission. Based on the structural and
PAFs at type Ia glomeruli (Maxwell et al. 2007), functional observations resumed previously, I
thus being the principal targets of IB4+ here propose that the interaction between the
non-peptidergic PAFs. It is also possible that two molecules in lamina II of the spinal cord
synaptic interactions are much more composite, primarily takes place at synaptic glomeruli that
as the vertical cells, in turn, give rise to V1 are particularly abundant in the substantia
profiles at the same type of glomeruli (Merighi gelatinosa (Ribeiro-Da-Silva and Coimbra
2018). Additionally, peptidergic PAFs containing 1982). Glomeruli are crucial to the process of
GDNF and somatostatin may be inhibitory at type presynaptic modulation of sensory information
Ib glomeruli of lamina IIid that have GFRα1- collected from periphery and conveyed to the
immunoreactive dendrites (Salio et al. 2014). superficial dorsal horn (Ribeiro-da-Silva 2015;
These dendrites are likely to stem from lamina Willis and Coggeshall 2004). A central axonal
I–II neurons, perhaps the transient or tonic central bouton (C bouton) made by a PAF, surrounded
cells, which express GFRα1 (Ortega-de San Luis by several dendrites and a few axon terminals,
and Pascual 2016). In keeping with this prospect, forms the glomeruli, which are separated by an
intrathecal GDNF inhibited the thermal hypersen- incomplete glial envelope from the surrounding
sitivity induced by substance P (Malcangio et al. neuropil. There are three types of glomeruli in
2002), and GDNF presynaptically reduced the primates, whereas four types have been described
release of substance P and CGRP from in rodents (Merighi et al. 2008b). Rat (Coimbra
capsaicin-stimulated PAFs (Salio et al. 2014). et al. 1974; Ribeiro-Da-Silva et al. 1985; Ribeiro-
14 Cross Talk of BDNF and GDNF in Spinal Substantia Gelatinosa (Lamina II): Focus on Circuitry 225

Da-Silva and Coimbra 1982) and mouse (Hiura glomeruli with a C bouton expressing GFRα1
et al. 1991; Marker et al. 2006; Salio et al. 2005; (Salio et al. 2014).
Salio et al. 2014) glomeruli only display slight
The peptidergic C PAFs that contain a mix of
ultrastructural differences. In their seminal work,
fast-acting (glutamate) and slow-acting
Ribeiro-da-Silva and Coimbra (1982) divided rat
transmitters are thus crucial in regulating the out-
glomeruli in two types (I and II) with different
flow of nociceptive information from the
ultrastructure (Ribeiro-Da-Silva and Coimbra
substantia gelatinosa in relation to the possibility
1982). Subsequent studies added substantial
that the two types of transmitters are differentially
information onto the differences in the neuro-
released according to the pattern of firing, even-
chemistry of the two categories. Thus, type I
tually leading to the release of glutamate without
glomeruli made by the unmyelinated C PAFs
or with coexisting growth factors or peptides
may be non-peptidergic (type Ia) or peptidergic
(Merighi 2017). It seems possible that under
(type Ib) on the absence or presence of LGVs
basal conditions and in nociceptive pain, only
containing neuropeptides and other high MW
glutamate is released from these fibers, albeit
modulators such as BDNF or GDNF at their C
one can hold that, as discussed previously, the
boutons. Type II glomeruli can also be further
peptidergic C PAFs containing GDNF exert a
subdivided into type IIa and type IIb based on
tonic inhibition onto the peptidergic C fibers
fine ultrastructural features. The C bouton in type
expressing BDNF at the type Ib glomeruli
IIa originates from a poorly myelinated Aδ PAF,
possessing GFRα1 peripheral dendrites
whereas that in type IIb derives from a myelinated
(Fig. 14.1) and thus that GDNF might be released
Aαβ PAF (Coimbra et al. 1984). Remarkably, the
also under basal conditions. When long-lasting
C boutons in type II glomeruli do not contain high
pain ensues, high MW transmitters are released
MW modulators and are, consequently, devoid
together with glutamate. In this case, the
of LGVs.
peptidergic C PAFs containing BDNF release
In relation to their BDNF or GDNF content
the growth factor that exerts its pronociceptive
and expression of the related receptors, type I
effects by enhancing the excitatory drive of lam-
glomeruli form four distinct subpopulations
ina II output large vertical cells, with the interpo-
(Fig. 14.1):
sition of a central cell (Fig. 14.1—path 1) and/or
1. The type Ia glomeruli made by with the intervention of two islet cells in series
non-peptidergic C fibers that are labelled by (Fig. 14.1—path 2). The pronociceptive effect to
IB4 and express GFRα1 (Salio et al. 2014). BDNF can be further increased by stimulation of
2. The type Ib glomeruli made by peptidergic C the TrkB autoreceptors that are present at certain
fibers that are endowed with LGVs containing type Ib glomeruli, as the activation of these pre-
BDNF herein stored with CGRP and sub- synaptic TrkB receptors leads to an additional
stance P (Salio et al. 2007). This second release of glutamate and substance P from C
group of glomeruli also expresses TrkB at PAFs (Merighi et al. 2008a). The effect of
their C boutons (Salio et al. 2005) and BDNF onto nociceptive transmission can be
GFRα1 (Salio et al. 2014) at dendrites. counteracted by GDNF in several different ways
3. The type Ib glomeruli made by peptidergic C (Merighi 2018):
fibers that are provided with LGVs containing
1. The GDNF released at the C boutons of
BDNF, CGRP, and substance P, but devoid of
peptidergic type Ib glomeruli acts onto the
TrkB, which is, instead, expressed at some
GFRα1 glomerular dendrites. These dendrites
glomerular dendrites (Salio et al. 2005).
may belong to a galanin-expressing inhibitory
4. The type Ib glomeruli made by peptidergic C
lamina II interneuron (Merighi 2018). This
fibers that are provided with LGVs containing
results in depressing the excitation of the
GDNF, CGRP, and somatostatin. The overall
large vertical cells (Fig. 14.1—path 3).
picture is completed by a number of type IIa
226 A. Merighi

2. The GDNF released at the C boutons of activation in the rat spinal cord increases with the firing
peptidergic type Ib glomeruli acts onto the frequency of C-fibers. Neuroscience 161:538–553
Airaksinen MS, Saarma M (2002) The GDNF family:
IB4+ non-peptidergic C PAFs expressing signalling, biological functions and therapeutic value.
GFRα1 and engaged in type Ia glomeruli. Nat Rev Neurosci 3:383–394
3. The GDNF released at the C boutons of Averill S, McMahon SB, Clary DO, Reichardt LF,
peptidergic type Ib glomeruli acts onto the Priestley JV (1995) Immunocytochemical localization
of trkA receptors in chemically identified subgroups of
IB4+ non-peptidergic Aδ PAFs expressing adult rat sensory neurons. Eur J Neurosci 7
GFRα1 and engaged in type IIa glomeruli. (7):1484–1494
4. The GDNF released at the C boutons of Bardoni R, Merighi A (2009) BDNF and trkB mediated
peptidergic type Ib glomeruli acts onto the mechanisms in the spinal cord. In: Malcangio M
(ed) Synaptic plasticity in pain. Springer, New York
Aβ PAFs expressing GFRα1 in lamina III. Bardoni R, Ghirri A, Salio C, Prandini M, Merighi A
(2007) BDNF-mediated modulation of GABA and
glycine release in dorsal horn lamina II from postnatal
rats. Dev Neurobiol 67:960–975
14.5 Concluding Remarks Bennett GJ, Abdelmoumene M, Hayashi H, Dubner R
(1980) Physiology and morphology of substantia
gelatinosa neurons intracellularly stained with horse-
I have here tried to provide a comprehensive
radish peroxidase. J Comp Neurol 194:809–827
portrait of the neuronal circuitry in lamina II that Bennett GJ, Ruda MA, Gobel S, Dubner R (1982) Enkeph-
represents the structural substrate for the alin immunoreactive stalked cells and islet cells in
interactions of BDNF and GDNF in the mature lamina IIb of the cat substantia gelatinosa. Brain Res
240:162–166
spinal cord. This portrait is somewhat speculative
Bennett DL, Michael GJ, Ramachandran N, Munson JB,
as our knowledge of the synaptic connections Averill S, Yan Q, McMahon SB, Priestley JV (1998) A
between the PAFs and the lamina II neurons is distinct subgroup of small DRG cells express GDNF
incomplete, and even less information is available receptor components and GDNF is protective for these
neurons after nerve injury. J Neurosci 18:3059–3072
on the connections between neurons inside the
Bennett DL, Boucher TJ, Armanini MP, Poulsen KT,
gelatinosa. This paucity of knowledge depends Michael GJ, Priestley JV, Phillips HS, McMahon SB,
on several reasons: (1) the difficulty in achieving Shelton DL (2000) The glial cell line-derived
a sound morphological and functional classifica- neurotrophic factor family receptor components are
differentially regulated within sensory neurons after
tion of the lamina II neurons, (2) the complexity
nerve injury. J Neurosci 20:427–437
in recording simultaneously from two neurons Blum R, Konnerth A (2005) Neurotrophin-mediated rapid
and to combine electrophysiological results with signaling in the central nervous system: mechanisms
structural information, (3) the still challenging and functions. Physiology 20:70–78
Borsook D, Becerra L (2011) CNS animal fMRI in pain
immunocytochemical approach at the ultrastruc-
and analgesia. Neurosci Biobehav Rev 35:1125–1143
tural level, and (4) the even more challenging Bradbury EJ, Burnstock G, McMahon SB (1998) The
feasibility to perform reliable quantitative studies. expression of P2X3 purinoreceptors in sensory
Yet, we are beginning to understand the impor- neurons: effects of axotomy and glial-derived
neurotrophic factor. Mol Cell Neurosci 12:256–268
tance of high MW neurotransmission in the spinal
Brown AG, Fyffe RE (1981) Form and function of dorsal
somatosensory system and, in particular, the horn neurones with axons ascending the dorsal
importance of the cross talk between different columns in cat. J Physiol 321:31–47
subsets of peptidergic PAFs in the modulation Bushnell MC, Duncan GH (1989) Sensory and affective
aspects of pain perception: is medial thalamus
of physiological pain and in the transition to
restricted to emotional issues? Exp Brain Res 78
long-lasting pathological pain. (2):415–418
Cervero F, Connell LA (1984) Distribution of somatic and
visceral primary afferent fibres within the thoracic spi-
nal cord of the cat. J Comp Neurol 230:88–98
References Cervero F, Laird JMA (1999) Visceral pain. Lancet
353:2145–2148
Adelson D, Lao L, Zhang G, Kim W, Marvizòn JC (2009) Chao MV, Hempstead BL (1995) p75 and Trk: a
Substance P release and neurokinin 1 receptor two-receptor system. Trends Neurosci 18:321–326
14 Cross Talk of BDNF and GDNF in Spinal Substantia Gelatinosa (Lamina II): Focus on Circuitry 227

Coimbra A, Sodré-Borges BP, Magalhaes MM (1974) The Grudt TJ, Perl ER (2002) Correlations between neuronal
substantia gelatinosa Rolandi of the rat. Fine structure, morphology and electrophysiological features in the
cytochemistry (acid phosphatase) and changes after rodent superficial dorsal horn. J Physiol 540:189–207
dorsal root section. J Neurocytol 3:199–217 Guo Y, Yao FR, Cao DY, Pickar JG, Zhang Q, Wang HS,
Coimbra A, Ribeiro-Da-Silva A, Pignatelli D (1984) Zhao Y (2008) Somatostatin inhibits activation of
Effects of dorsal rhizotomy on the several types of dorsal cutaneous primary afferents induced by anti-
primary afferent terminals in laminae I-III of the rat dromic stimulation of primary afferents from an adja-
spinal cord. An electron microscope study. Anat cent thoracic segment in the rat. Brain Res 1229:61–71
Embryol (Berl) 170:279–287 Hantman AW, Perl ER (2005) Molecular and genetic
Conner JM, Lauterborn JC, Yan Q, Gall CM, Varon S features of a labeled class of spinal substantia
(1997) Distribution of brain-derived neurotrophic fac- gelatinosa neurons in a transgenic mouse. J Comp
tor (BDNF) protein and mRNA in the normal adult rat Neurol 492:90–100
CNS: evidence for anterograde axonal transport. J Hantman AW, van den Pol AN, Perl ER (2004) Morpho-
Neurosci 17:2295–2313 logical and physiological features of a set of spinal
Cruz L, Basbaum AI (1985) Multiple opioid peptides and substantia gelatinosa neurons defined by green fluores-
the modulation of pain: immunohistochemical analysis cent protein expression. J Neurosci 24:836–842
of dynorphin and enkephalin in the trigeminal nucleus Hiura A, Ishizuka H, Villalobos EL (1991) GABAergic
caudalis and spinal cord of the cat. J Comp Neurol neurons in the mouse superficial dorsal horn with spe-
240:331–348 cial emphasis on their relation to primary afferent cen-
Duan B, Cheng L, Bourane S, Britz O, Padilla C, Garcia- tral terminals. Arch Histol Cytol 54:195–206
Campmany L, Krashes M, Knowlton W, Velasquez T, Holstege JC, Jongen JL, Kennis JH, Rooyen-Boot AA,
Ren X, S-á R, B-á L, Wang Y, Goulding M, Ma Q Vecht CJ (1998) Immunocytochemical localization of
(2014) Identification of spinal circuits transmitting and GDNF in primary afferents of the lumbar dorsal horn.
gating mechanical pain. Cell 159:1417–1432 Neuroreport 9:2893–2897
Ernfors P, Merlio JP, Persson H (1992) Cells expressing Huang J, Polgár E, HJr S, Mishra SK, Tseng PY,
mRNA for Neurotrophins and their receptors during Iwagaki N, Boyle KA, Dickie AC, Kriegbaum MC,
embryonic rat development. Eur J Neurosci Wildner H, Zeilhofer HU, Watanabe M, Riddell JS,
4:1140–1158 Todd AJ, Hoon MA (2018) Circuit dissection of the
Gebhart GF (2000) J.J. Bonica lecture--2000: physiology, role of somatostatin in itch and pain. Nat Neurosci
pathophysiology, and pharmacology of visceral pain. 21:707–716
Reg Anesth Pain Med 25:632–638 Jongen JL, Jaarsma D, Hossaini M, Natarajan D, Haasdijk
Gibon J, Barker PA (2017) Neurotrophins and proneuro- ED, Holstege JC (2007) Distribution of RET immuno-
trophins: focus on synaptic activity and plasticity in the reactivity in the rodent spinal cord and changes after
brain. Neuroscientist 23:587–604 nerve injury. J Comp Neurol 500:1136–1153
Giesler GJ Jr, Cannon JT, Urca G, Liebeskind JC (1978) Josephson A, Widenfalk J, Trifunovski A, Widmer HR,
Long ascending projections from substantia gelatinosa Olson L, Spenger C (2001) GDNF and NGF family
Rolandi and the subjacent dorsal horn in the rat. Sci- members and receptors in human fetal and adult spinal
ence 202:984–986 cord and dorsal root ganglia. J Comp Neurol
Gobel S (1975) Golgi studies of the substantia gelatinosa 440:204–217
neurons in the spinal trigeminal nucleus. J Comp Kashiba H, Uchida Y, Senba E (2003) Distribution and
Neurol 162:397–416 colocalization of NGF and GDNF family ligand recep-
Gobel S (1978a) Golgi studies of the neurons in layer I of tor mRNAs in dorsal root and nodose ganglion neurons
the dorsal horn of the medulla (trigeminal nucleus of adult rats. Brain Res Mol Brain Res 110:52
caudalis). J Comp Neurol 180:375–394 Kato G, Kawasaki Y, Ji RR, Strassman AM (2007) Differ-
Gobel S (1978b) Golgi studies of the neurons in layer II of ential wiring of local excitatory and inhibitory synaptic
the dorsal horn of the medulla (trigeminal nucleus inputs to islet cells in rat spinal lamina II demonstrated
caudalis). J Comp Neurol 180:395–413 by laser scanning photostimulation. J Physiol
Gold MS, Gebhart GF (2010) Nociceptor sensitization in 580:815–833
pain pathogenesis. Nat Med 16(11):1248–1257 Khan N, Smith MT (2015) Neurotrophins and neuropathic
Golden JP, Hoshi M, Nassar MA, Enomoto H, Wood JN, pain: role in pathobiology. Molecules 20:10657–10688
Milbrandt J, Gereau RW, Johnson EM Jr, Jain S (2010) Koerber HR (2009) Nociceptors: adequate stimulus. In:
RET signaling is required for survival and normal Binder MD, Hirokawa N, Windhorst U (eds) Encyclo-
function of nonpeptidergic nociceptors. J Neurosci pedia of neuroscience. Springer, Berlin, Heidelberg, pp
30:3983–3994 2873–2876
Groenewegen HJ (1988) Organization of the afferent Li L, Rutlin M, Abraira VE, Cassidy C, Kus L, Gong S,
connections of the mediodorsal thalamic nucleus in M-á J, Luo W, Heintz N, Koerber H, Woodbury C,
the rat, related to the mediodorsal-prefrontal topogra- Ginty DD (2011) The functional organization of cuta-
phy. Neuroscience 24:379–431 neous low-threshold mechanosensory neurons. Cell
147:1615–1627
228 A. Merighi

Light AR, Kavookjian AM (1988) Morphology and ultra- spinal cord nociceptive pathways. Prog Brain Res
structure of physiologically identified substantia 146:291–321
gelatinosa (lamina II) neurons with axons that termi- Merighi A, Bardoni R, Salio C, Lossi L, Ferrini F,
nate in deeper dorsal horn laminae (III-V). J Comp Prandini M, Zonta M, Gustincich S, Carmignoto G
Neurol 267:172–189 (2008a) Presynaptic functional trkB receptors mediate
Light AR, Perl ER (1979) Reexamination of the dorsal the release of excitatory neurotransmitters from pri-
root projection to the spinal dorsal horn including mary afferent terminals in lamina II (substantia
observations on the differential termination of coarse gelatinosa) of postnatal rat spinal cord. Dev Neurobiol
and fine fibers. J Comp Neurol 186:117–131 68:457–475
Liu XG, Sandkühler J (1995) The effects of extrasynaptic Merighi A, Salio C, Ghirri A, Lossi L, Ferrini F, Betelli C,
substance P on nociceptive neurons in laminae I and II Bardoni R (2008b) BDNF as a pain modulator. Prog
in rat lumbar spinal dorsal horn. Neuroscience Neurobiol 85:297–317
68:1207–1218 Michael GJ, Averill S, Nitkunan A, Rattray M, Bennett
Lorenzo LE, Ramien M, St.Louis M, De Koninck Y, DLH, Yan Q, Priestley JV (1997) Nerve growth factor
Ribeiro-da-Silva A (2008) Postnatal changes in the treatment increases brain-derived neurotrophic factor
Rexed lamination and markers of nociceptive afferents selectively in trk-a expressing dorsal root ganglion
in the superficial dorsal horn of the rat. J Comp Neurol cells and in their central terminations within the spinal
508:592–604 cord. J Neurosci 17:8476–8490
Lu Y, Perl ER (2003) A specific inhibitory pathway Mogil JS (2019) The translatability of pain across species.
between substantia gelatinosa neurons receiving direct Phil Trans R Soc London B374:20190286
C-fiber input. J Neurosci 23:8752–8758 Molliver DC, Snider WD (1997) Nerve growth factor
Lu Y, Perl ER (2005) Modular organization of excitatory receptor TrkA is down-regulated during postnatal
circuits between neurons of the spinal superficial dorsal development by a subset of dorsal root ganglion
horn (laminae I and II). J Neurosci 25:3900–3907 neurons. J Comp Neurol 381:428–438
Luo R, Guo Y, Cao DY, Pickar JG, Li L, Wang J, Zhao Y Molliver DC, Wright DE, Leitner ML, Parsadanian AS,
(2010) Local effects of octreotide on glutamate-evoked Doster K, Wen D, Yan Q, Snider WD (1997)
activation of Aδ and C afferent fibers in rat hairy skin. IB4-binding DRG neurons switch from NGF to
Brain Res 1322:50–58 GDNF dependence in early postnatal life. Neuron
Malcangio M, Getting SJ, Grist J, Cunningham JR, 19:849–861
Bradbury EJ, Charbel IP, Lever IJ, Pezet S, Perretti Ohta K, Inokuchi T, Gen E, Chang J (2001) Ultrastructural
M (2002) A novel control mechanism based on GDNF study of anterograde transport of glial cell line-derived
modulation of somatostatin release from sensory neurotrophic factor from dorsal root ganglion neurons
neurones. FASEB J 16:730–732 of rats towards the nerve terminal. Cells Tiss Organs
Marker CL, Lujan R, Colon J, Wickman K (2006) Distinct 169:410–421
populations of spinal cord lamina II interneurons Ortega-de San Luis C, Pascual A (2016) Simultaneous
expressing G-protein-gated potassium channels. J detection of both GDNF and GFRα1 expression
Neurosci 26:12251–12259 patterns in the mouse central nervous system. Front
Maxwell DJ, Belle MD, Cheunsuang O, Stewart A, Morris Neuroanat 10:73
R (2007) Morphology of inhibitory and excitatory Ossipov MH (2011) Growth factors and neuropathic pain.
interneurons in superficial laminae of the rat dorsal Curr Pain Headache Rep 15:185–192
horn. J Physiol 584:521–533 Pan YZ, Pan HL (2004) Primary afferent stimulation dif-
McMahon SB, Dmitrieva N, Koltzenburg M (1995) Vis- ferentially potentiates excitatory and inhibitory inputs
ceral pain. Br J Anaesth 75:132–144 to spinal lamina II outer and inner neurons. J
Melzack R, Wall PD (1965) Pain mechanisms: a new Neurophysiol 91:2413–2421
theory. Science 150:971–980 Pintér E, Helyes Z, Szolcsanyi J (2006) Inhibitory effect of
Merighi A (2016) Targeting the glial-derived neurotrophic somatostatin on inflammation and nociception.
factor and related molecules for controlling normal and Pharmacol Ther 112:440–456
pathologic pain. Exp Opin Ther Targets 20:193–208 Polgár E, Al Ghamdi KS, Todd AJ (2010) Two
Merighi A (2017) Neuropeptides and Coexistence. In: populations of neurokinin 1 receptor-expressing pro-
Reference module in neuroscience and biobehavioral jection neurons in lamina I of the rat spinal cord that
psychology. Elsevier. isbn:9780128093245, differ in AMPA receptor subunit composition and den-
Amsterdam. https://doi.org/10.1016/B978-0-12- sity of excitatory synaptic input. Neuroscience
809324-5.02310-5 167:1192–1204
Merighi A (2018) The histology, physiology, neurochem- Punnakkal P, von Schoultz C, Haenraets K, Wildner H,
istry and circuitry of the substantia gelatinosa Rolandi Zeilhöfer HU (2014) Morphological, biophysical and
(lamina II) in mammalian spinal cord. Prog Neurobiol synaptic properties of glutamatergic neurons of the
169:91–134 mouse spinal dorsal horn. J Physiol 592:759–776
Merighi A, Carmignoto G, Gobbo S, Lossi L, Salio C, Reichardt LF (2006) Neurotrophin-regulated signalling
Vergnano AM, Zonta M (2004) Neurotrophins in pathways. Philos Trans R Soc Lond B 361:1545–1564
14 Cross Talk of BDNF and GDNF in Spinal Substantia Gelatinosa (Lamina II): Focus on Circuitry 229

Rexed B (1952) The cytoarchitectonic organisation of the cutaneous stimulation. Physiological and anatomical
spinal cord of the cat. J Comp Neurol 96:415–496 features. J Neurophysiol 59:833–860
Rexed B (1954) A cytoarchitectonic atlas of the spinal Thompson SJ, Bushnell MC (2012) Rodent functional and
cord in the cat. J Comp Neurol 100:297–379 anatomical imaging of pain. Neurosci Lett
Ribeiro-da-Silva A (2015) Chapter 7 – Substantia 520:131–139
gelatinosa of the spinal cord – Paxinos, George. In: Todd AJ, McKenzie J (1989) GABA-immunoreactive
The rat nervous system, 4th edn. Academic Press, San neurons in the dorsal horn of the rat spinal cord. Neu-
Diego, pp 97–114 roscience 31:799–806
Ribeiro-Da-Silva A, Coimbra A (1982) Two types of Todd AJ, Hughes DI, Polgàr E, Nagy GG, Mackie M,
synaptic glomeruli and their distribution in laminae Ottersen OP, Maxwell DJ (2003) The expression of
I-III of the rat spinal cord. J Comp Neurol vesicular glutamate transporters VGLUT1 and
209:176–186 VGLUT2 in neurochemically defined axonal
Ribeiro-Da-Silva A, De Koninck Y (2008) Morphological populations in the rat spinal cord with emphasis on
and neurochemical organization of the spinal dorsal the dorsal horn. Eur J Neurosci 17:13–27
horn. In: Albright TD, Albright TD, Masland RH et Wang J, Guo Y, Cao DY, Luo R, Ma SJ, Wang HS, Pickar
al (eds) The senses: a comprehensive reference. Aca- JG, Zhao Y (2009) Tonic inhibition of somatostatin on
demic Press, New York, pp 279–310 C and Aδ afferent fibers in rat dorsal skin in vivo. Brain
Ribeiro-Da-Silva A, Pignatelli D, Coimbra A (1985) Syn- Res 1288:50–59
aptic architecture of glomeruli in superficial dorsal Widenfalk J, Lundstromer K, Jubran M, Brene S, Olson L
horn of rat spinal cord, as shown in serial (2001) Neurotrophic factors and receptors in the imma-
reconstructions. J Neurocytol 14:203–220 ture and adult spinal cord after mechanical injury or
Ribeiro-Da-Silva A, Pioro EP, Cuello AC (1991) Sub- kainic acid. J Neurosci 21:3457–3475
stance P- and enkephalin-like immunoreactivities are Willis WD Jr, Coggeshall RE (2004) Structure of the
colocalized in certain neurons of the substantia dorsal horn. In: Sensory mechanisms of the spinal
gelatinosa of the rat spinal cord: an ultrastructural cord – primary afferent neurons and the spinal dorsal
double-labeling study. J Neurosci 11:1068–1080 horn, 3rd edn. Kluver Academic/Plenum Publisher,
Salio C, Ferrini F (2016) BDNF and GDNF expression in New York, pp 155–186
discrete populations of nociceptors. Ann Anat Willis WD, Leonard RB, Kenshalo DRJ (1978)
207:55–61 Spinothalamic tract neurons in the substantia
Salio C, Lossi L, Ferrini F, Merighi A (2005) Ultrastruc- gelatinosa. Science 202:986–988
tural evidence for a pre- and post-synaptic localization Woodbury CJ, Ritter AM, Koerber HR (2000) On the
of full length trkB receptors in substantia gelatinosa problem of lamination in the superficial dorsal horn
(lamina II) of rat and mouse spinal cord. Eur J Neurosci of mammals: a reappraisal of the substantia gelatinosa
22:1951–1966 in postnatal life. J Comp Neurol 417:88–102
Salio C, Averill S, Priestley JV, Merighi A (2007) Yasaka T, Kato G, Furue H, Rashid MH, Sonohata M,
Costorage of BDNF and neuropeptides within individ- Tamae A, Murata Y, Masuko S, Yoshimura M (2007)
ual dense-core vesicles in central and peripheral Cell-type-specific excitatory and inhibitory circuits
neurons. Dev Neurobiol 67:326–338 involving primary afferents in the substantia gelatinosa
Salio C, Ferrini F, Muthuraju S, Merighi A (2014) Presyn- of the rat spinal dorsal horn in vitro. J Physiol
aptic modulation of spinal nociceptive transmission by 581:603–618
glial cell line-derived neurotrophic factor (GDNF). J Yasaka T, Tiong SY, Hughes DI, Riddell JS, Todd AJ
Neurosci 34:13819–13833 (2010) Populations of inhibitory and excitatory
Schoenen J (1982) Dendritic organization of the human interneurons in lamina II of the adult rat spinal dorsal
spinal cord: the dorsal horn. Neuroscience horn revealed by a combined electrophysiological and
7:2057–2087 anatomical approach. Pain 151:475–488
Sengupta JN (2009) Visceral pain: the neurophysiological Yasaka T, Tiong SY, Polgár E, Watanabe M, Kumamoto
mechanism. Handbook Exp Pharmacol 194:31–74 E, Riddell JS, Todd AJ (2014) A putative relay circuit
Seybold VS (2009) The role of peptides in central sensiti- providing low-threshold mechanoreceptive input to
zation. In: Canning BJ, Spina D (eds) Sensory nerves. lamina I projection neurons via vertical cells in lamina
Springer, Berlin, Heidelberg, pp 451–491 II of the rat dorsal horn. Mol Pain 10:1744–8069
Sugiura Y, Terui N, Hosoya Y (1989) Difference in distri- Zheng J, Lu Y, Perl ER (2010) Inhibitory neurones of the
bution of central terminals between visceral and spinal substantia gelatinosa mediate interaction of
somatic unmyelinated (C) primary afferent fibers. J signals from primary afferents. J Physiol
Neurophysiol 62:834–840 588:2065–2075
Surmeier DJ, Honda CN, Willis WD Jr (1988) Natural
groupings of primate spinothalamic neurons based on
Part V
Clinical Medicine
BDNF Gene as a Precision Skill of Obesity
Management 15
Helena Marcos-Pasero, Elena Aguilar-Aguilar,
Maria P. Ikonomopoulou, and Viviana Loria-Kohen

Abstract various GWASs. Finally, we assess gene-diet


The scarcity of the results obtained for the interaction as a novel tool to prevent obesity
treatment of obesity leads us to consider new and formulate solid and personalized
strategies, contemplating all the factors nutritional management. Our research group
involved in the development of the disease. has performed the first study on the association
One of the key molecules for controlling of BDNF-AS rs925946 polymorphism and cal-
body weight and energy homeostasis is the cium intake as potential modulators of the
brain-derived neurotrophic factor (BDNF). nutritional status. Although these results
This work summarizes the mechanisms in should be confirmed in future studies, they
which BDNF gene regulates this multifactorial open the path for new prevention
disease. In addition, we discuss the role of opportunities.
other BDNF polymorphisms as genetic
determinants of obesity. In this context, a
total of 14 SNPs near or inside BDNF/
BDNF-AS related to BMI were identified in 15.1 Introduction

Obesity is a complex, multifactorial, and


H. Marcos-Pasero · E. Aguilar-Aguilar
Nutrition and Clinical Trials Unit, GENYAL Platform, preventable disease whose high prevalence
IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain makes it an important public health problem.
e-mail: helena.marcos@imdea.org; elena.aguilar@imdea. According to the World Health Organization
org
(WHO), more than 1.9 billion individuals over
M. P. Ikonomopoulou 18 years had overweight, and more than 650 mil-
Translational Venomics Group, IMDEA-Food, CEI UAM
lion were obese in 2016. In addition, the preva-
+CSIC, Madrid, Spain
lence of obesity has been tripled since 1975
Institute for Molecular Bioscience, The University of
(Obesity and Overweight 2019). Obesity
Queensland, St Lucia, Australia
e-mail: maria.ikonomopoulou@imdea.org increases the risk of a large number of cardiovas-
cular, metabolic, and pulmonary diseases and
V. Loria-Kohen (*)
Nutrition and Clinical Trials Unit, GENYAL Platform, deaths (Hruby and Hu 2015).
IMDEA-Food Institute, CEI UAM + CSIC, Madrid, Spain At early ages these values are also alarming. It
Department of Nutrition and Food Science, Faculty of is estimated that more than 340 million children
Pharmacy, Complutense University of Madrid, Madrid, between 5 and 19 years old are overweight or
Spain obese and that prevalence has been increased
e-mail: viviana.loria@imdea.org; vloria@ucm.es

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 233
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_15
234 H. Marcos-Pasero et al.

from 4% in 1975 to 18% in 2016 (Obesity and studies have suggested that BDNF may regulate
Overweight 2019). obesity via different mechanisms.
Obesity and its many medical complications
emerge from the dysregulation of energy homeo-
stasis. Excessive weight gain might also arise 15.2.1 Role of BDNF in Energy
from alterations in reward systems of the brain Homeostasis and Food Intake
that drive consumption of calorie-dense and pal-
atable foods in the absence of an energy require- Different studies using mouse models argue in
ment (Rios 2014). favor of an endocrine role of BDNF in energy
Energy balance leads us to hypothesize that homeostasis to modulate food intake and
obesity is caused by a mere increase of energy glycemia. In db/db mice (nonfunctional leptin
intake. However, the evidence does not seem to receptor mice provide a model for obesity and
support this argument. On the contrary, there are non-insulin-dependent diabetes mellitus), contin-
indicators that the origin of this disease is uous treatment with BDNF over several days has
underlined by numerous interrelated factors been shown to decrease food intake and lower
(genetic, social, economic, etc.) that make obesity blood glucose to normoglycemic levels. BDNF
a highly complex condition (Hruby and Hu treatment sustained body temperature and oxygen
2015). consumption during food restriction, probably
The scarcity of the results obtained from obe- due to an increase in energy expenditure
sity treatment leads us to consider new strategies, (Nakagawa et al. 2000). In addition, BDNF-
contemplating all the factors involved in the treated db/db mice and vehicle-treated pair-fed
development of this disease in order to find controls display a similar pattern of weight loss.
novel therapeutic avenues for its treatment and Similar observation on energy expenditure has
prevention as well as its associated medical been made on the acute effects of BDNF in db/
afflictions. db mice, where a single injection largely
prevented fasting-induced hypothermia and cold
exposure-induced hypothermia whereas the body
15.2 Implication of the BDNF weight and blood glucose were equally reduced in
in Obesity and Eating treated and vehicle-treated groups. BDNF treat-
Disorders ment has also been shown to increase whole-body
glucose turnover, and to improve serum lipid
Complex interactions between the brain and parameters in db/db mice. All these results high-
peripheral tissues mediate the effective control light that BDNF treatment improves the erratic
of energy balance and body weight. One of the energy balance, which characterizes db/db mice
key molecules for body weight control and (Lebrun et al. 2006).
energy homeostasis is the brain-derived All together show that BDNF mediates the
neurotrophic factor (BDNF). This neurotrophin central control of food intake, which acts as an
(NT) has been extensively studied to elucidate its anorexigenic factor. Most studies on the role of
function as a potential target against obesity BDNF in food intake regulation are focused on
(Lebrun et al. 2006; Rosas-Vargas et al. 2011). the hypothalamus as the probable food intake and
The first study which suggested a role for energy balance target. Until recently, two sites in
BDNF in the regulation of food intake was carried the adult brain have been highlighted for the
out in 1992 by Lapchak and Hefti, who described anorectic role of BDNF: (1) the ventromedial
that the central infusion of BDNF reduces weight nucleus in the hypothalamus, where its mRNA
gain in rats (Lapchak and Hefti 1992). Later on, it levels are modulated by the nutritional state and
was shown that infusion of BDNF in the lateral melanocortin signaling, and (2) the dorsal vagal
ventricle of adult rats caused dose-dependent, complex in the hindbrain, where exogenous pro-
severe appetite suppression and weight loss tein infusion induces anorexia and endogenous
(Pelleymounter et al. 1995). In general, different protein levels are modulated by nutritional status,
15 BDNF Gene as a Precision Skill of Obesity Management 235

peripheral leptin, and cholecystokinin treatments 15.2.3 Genetic Alterations


(Lebrun et al. 2006). in the Genes Encoding BDNF or
Its High-Affinity Receptor TrkB
(Tropomyosin-Related Kinase
15.2.2 Participation of BDNF B)
on Metabolism
Mutations in the BDNF gene and its receptor
TrkB lead to severe obesity in humans. The
Contradicting studies concerning the action of
WAGR (Wilms’ tumor, aniridia, genitourinary
BDNF on metabolism exist in the literature. In
anomalies, and mental retardation) disorder is
this respect, clinical data indicate that decreased
characterized by contiguous gene syndrome and
BDNF plasma levels, which are associated with
displays obesity when deletions extend into the
birth weight and body mass index, may influence
BDNF locus (Han 2016). Deletions of chromo-
the development and pathophysiology of child-
some 11p ranged from 1.0 to 26.5 Mb, and 58%
hood obesity. Similarly, diminished BDNF serum
of the patients had heterozygous BDNF deletions.
levels impair glucose metabolism and correlate
These patients had significantly higher BMI
with obesity and diabetic complications in adults.
z-scores throughout childhood than did patients
Contrary to these findings, fasting serum BDNF
with intact BDNF length (Han et al. 2008).
concentrations have been found to be high in
Different variants of the BDNF-encoding gene
obese women. Furthermore, recent results do not
have been described in human patients with AN
link BDNF and insulin resistance or cardiovascu-
or BN. The most frequently encountered variant
lar risk factors in obese children but demonstrate
(196G/A) leads to a single amino acid substitu-
a positive correlation between the gene alterations
tion (Val66Met) in the pro-region of BDNF. It has
and changes of leptin, probably due to fat mass
been shown that this mutation affects the sorting
(Briana and Malamitsi-Puchner 2018). Neverthe-
of BDNF into the nerve terminals and markedly
less, a recent meta-analysis developed by
reduces its activity-dependent secretion (Chen
Sandrini and colleagues concluded that obese
et al. 2004). Mutations in the gene encoding
patients have normal levels of BDNF and
TrkB have also been linked to eating disorders.
emphasized that data evaluation may be
A de novo mutation leading to a single amino acid
influenced by the procedure of sampling,
substitution (Y722C) on the gene encoding TrkB
handling, and storage and these could lead to a
has been described in a young boy exhibiting
difficulty in the results’ interpretation. Hence,
severe obesity. This mutation has been shown to
standardization of the procedure is urgently
considerably alter the signaling capabilities of the
needed for strong, affordable, and reliable
receptor TrkB (Lebrun et al. 2006). In addition, a
conclusions (Sandrini et al. 2018).
genome-wide association study revealed a link
Correlation studies between BDNF serum
between polymorphisms of BDNF and obesity
levels and eating disorders in humans have also
(Thorleifsson et al. 2009). Likewise, a recent
been undertaken. It has been demonstrated that
meta-analysis developed by Akbarian and
BDNF serum levels are significantly reduced in
colleagues suggests that some polymorphisms in
female patients with anorexia nervosa (AN) or
BDNF including rs925946, rs10501087, rs6265,
bulimia nervosa (BN) (Nakazato et al. 2003;
and rs988712 can be considered as genetic
Monteleone et al. 2005). It has been further
determinants of obesity (Akbarian et al. 2018).
suggested that since BDNF exerts a satiating
The presence of polymorphisms in the BDNF
effect, this may be an adaptation to the decreased
associated with obesity will be discussed deeply
caloric ingestion in these patients (Lebrun et al.
in the following section.
2006).
236 H. Marcos-Pasero et al.

15.2.4 Epigenetic Programming on the dopamine receptor D1 could prevent dopa-


of BDNF mine release in the nucleus accumbens and the
dorsal striatum as a compensatory effect. As the
The interactions between environmental and VTA has been identified as an important source of
genetic factors are reflected by epigenetic BDNF contributing to hedonic eating regulation,
changes, including DNA methylation and histone it would be of interest to evaluate whether BDNF
modifications. These variations are particularly acts on TrkB-expressing neurons in the VTA or
important at early life stages due to their potential the nucleus accumbens to regulate the consump-
impact on individuals’ physiological develop- tion of palatable food. Thus, it is necessary to
ment (Rosas-Vargas et al. 2011). Recent evidence understand the changes in the reward circuit’s
suggests that BDNF may have essential functions structure and function when BDNF signaling is
during pregnancy by modulating fetal growth and defective and promotes hyperphagia on high-fat
participating in the development and maturation diets and leads to obesity (Xu and Xie 2016).
of central and peripheral fetal organs, including
the placenta. This gene probably acts as a nutrient
functional sensor for the placenta and may consti- 15.2.6 BDNF as a Mediator
tute a link between maternal nutrition and fetal in the Effects of Exercise
nutrients’ demand. Furthermore, the diminished and Food Intake
BDNF signaling is linked with energy balance on Neuroplasticity
dysregulation. Indeed, a close association has and the Vulnerability
been described between BDNF and adult of the Brain and Peripheral
nutritional status in the central nervous system. Organs to Metabolic Morbidity
Both the functional loss of one copy of BDNF and and Obesity
a de novo mutation of the full-length TrkB recep-
tor have been associated with severe obesity in Recent evidence suggests that running and other
humans. BDNF is downregulated in preterm types of aerobic exercise can enhance cognitive
infants, with tendency for obesity and develop- performance and increase serum BDNF levels in
ment of metabolic syndrome later in life (Briana human subjects. Running also lessens anxiety and
and Malamitsi-Puchner 2018). depression in animal models by BDNF-mediated
mechanisms. This is consistent with the role of
BDNF in alleviating anxiety and depression in
humans. Dietary energy restriction can exert anxi-
15.2.5 BDNF and Hedonic Eating
olytic effects via the involvement of BDNF and
ketone bodies. All these aspects might have a
In addition to its role in the homeostatic regula-
crucial effect on the obesity control (Marosi and
tion of energy balance, BDNF may suppress
Mattson 2014).
hedonic feeding of palatable food through the
reward system. This system begins in the ventral
tegmental area (VTA) and joins to the nucleus
15.3 SNPs of the BDNF Associated
accumbens. Mice with deficient BDNF-to-TrkB
with the Obesity
signaling exhibit hyperphagia while on high-fat
diets because of the larger meal size. Mice lacking
During the last few decades, significant changes
BDNF synthesis in CaMKIIα-expressing neurons
have been taken place in high-throughput geno-
show an extreme hypersensitivity of the dopa-
mics and thus advanced on the whole-genome
mine receptor D1 as it is shown that peripheral
sequencing data through new molecular
injection of D1 receptor agonist normalizes the
technologies and new analytical methods. The
hyperphagia observed in high-fat diets. We
facilitation of recruiting unrelated individuals
hypothesize that the effect of BDNF deficiency
has contributed to the novel genomics research
15 BDNF Gene as a Precision Skill of Obesity Management 237

through the performance of a large population- rs10501087, rs6265, and rs988712. This study
based biobanks-genetic database for public analyzed the results from 35 cross-sectional
availability. observational and case-control studies where all
These achievements have been possible thanks kinds of BDNF/BDNF-AS SNPs were studied in
to genome-wide association studies (GWASs). relation to BMI. The overall results by means of β
The primary goal of these studies is to better and OR showed a statistically significant associa-
understand the biology of the disease, under the tion between the SNPs and the BMI (Akbarian
study of associations between common traits and et al. 2018).
common variants. In the context of polygenic or However, inconsistencies in the researches’
common diseases, many loci affecting the trait results impose the need for further study. Thus,
segregate in the population should be examined. a summary of the published studies available in
GWAS, up until now, has identified more than the literature from all the BMI-related SNPs of
10,000 robust associations among SNPs and BDNF/BDNF-AS is presented below:
diseases or traits, proving highly replicable, both Note that we found no entries for rs10767654,
within and between populations (Hebbring 2019; rs11030100, rs11030102, rs2049045, rs492346,
Visscher et al. 2017). rs7103411, or rs7481311.
At least, 500 human adiposity-related loci
were identified by GWAS (Loos 2018). In the
majority of these genes, mutations result in mono- 15.3.1 rs10501087
genic obesity and common obesity, including
MC4R, BDNF, PCSK1, POMC, SH2B1, LEPR, This genetic variant is significantly linked to BMI
and NTRK2 (Goodarzi 2018). The latest meta- in the latest meta-analysis and review of
analysis of GWAS for BMI in European BMI-related SNPs of BDNF/BDNF-AS
populations ( p < 1  108) revealed (Akbarian et al. 2018). Results are coincident
941 BMI-related SNPs. In that study, there were with other studies involving both adulthood and
over 250,000 European participants and have childhood populations of European descents (Jiao
been identified more than 100 new BMI-related et al. 2011; Zhao et al. 2009), and with extreme
SNPs, a fact that highlights the necessity for a obesity specifically among French and German
large sample size to increase prediction accuracy children (Scherag et al. 2010).
and to provide insights into complex diseases Interestingly, the Black Women’s Health
(Yengo et al. 2018). Study in African-American women has found no
The GWAS results have been reported in vari- correlation between rs10501087 and BMI (Ruiz-
ous databases. This paper has at length discussed Narváez et al. 2016), maybe due to differences
the researches published in Ensembl (www. caused by race, ethnicity, or country of origin.
ensembl.org/index.html), GWAS Catalog (www. This genetic variant has been highlighted in a
ebi.ac.uk/gwas/), and GWAS Central (www. genetic risk score (GRS) for the study of Spanish
gwascentral.org). We identified 14 SNPs near or obesity in two prospective representatives to the
inside BDNF/BDNF-AS related to BMI: central population cohort studies, Pizarra in the
rs10501087, rs10767654, rs10767664, South of Spain and Zona Urbana de Valladolid
rs11030100, rs11030102, rs11030104, (Martínez-García et al. 2013).
rs16917237, rs2030323, rs2049045, rs4923460,
rs6265, rs7103411, rs7481311, and rs925946
(Fig. 15.1). 15.3.2 rs10767664
These genetic variants and their GWAS-effect
size are shown in Table 15.1. The rs10767664 variation has been associated
Conversely, there are only four BMI-related with BMI in the GOYA study composed of two
SNPs in BDNF/BDNF-AS as a result of the latest large Danish cohorts (Paternoster et al. 2011) as
meta-analysis and review on the topic: rs925946, well as in another European population-based
238 H. Marcos-Pasero et al.

Fig. 15.1 Plotted


ideogram of all BMI-related
SNPs in or near BDNF/
BDNF-AS
(11-chromosome).
Footnote: Lines were
plotted on the chromosome
corresponding to the base-
pair location of each
BDNF/BDNF-AS SNP,
and the line connects to
colored circles representing
the same phenotype
associated with that SNP
(BMI) [PhenoGram (Wolfe
et al. 2013)]

work (Vimaleswaran et al. 2012) that included a Young Finns Study of a European cohort
gene-centric meta-analysis study (Guo et al. (Juonala et al. 2011), or in Chinese children
2013) and a population of Saudi Arabians (Wang et al. 2016). In the case of elderly, this
(Alharbi et al. 2014). However, in other studies polymorphism has been strongly associated with
such an association has not been found (Wang BMI in the AGES-Reykjavik comprising 4,846
et al. 2011; McCaffery et al. 2013). individuals of European ancestry (Murphy et al.
In fact, this association has reported 2013).
conflicting data in children and in elderly This variant has also been studied in the con-
populations. For example, a longitudinal study text of other obesity-related traits, such as the age
shows a distinct effect on BMI during childhood of onset of menarche (Fernández-Rhodes et al.
in the community of Bogalusa in Louisiana (Mei 2013) and type 2 diabetes mellitus in Caucasian
et al. 2012). These results are in line with the data women (De et al. 2017a). Common homozygous
reported in other pediatric cohorts composed of AA showed a daily increase of 100 kcal in their
South Brazilian children (Zandoná et al. 2017). energy intake in the Look AHEAD (Action for
Despite these data, this association proved not to Health in Diabetes) clinical trial (McCaffery et al.
be significant for Cardiovascular Risk in the 2012), and a better improvement in glucose,
15

Table 15.1 Summary of results from the SNPs of the GWAS of BMI inside or near BDNF/BDNF-AS
Risk
Variant allele Mapped gene Location Consequence MAF Beta CI p Study accession
rs10501087 T BDNF-AS 11:27648561 Intron variant 0.230 0.072 unit increase [0.043–0.101] 7  107 Ng et al. (2017)
rs10767654 T LINC00678, BDNF- 11:27618676 Intron variant 0.327 0.054 unit decrease [0.040–0.070] 2  1012 Tachmazidou et al.
AS (2017)
rs10767664 A BDNF, AC103796.1 11:27704439 Intron variant 0.239 0.190 kg/m2 [0.130–0.250] 5  1026 Speliotes et al. (2010)
increase
rs11030100 T BDNF-AS, BDNF 11:27656039 3 prime UTR variant 0.228 0.038 kg/m2 [0.032–0.044] 1  1028 Akiyama et al. (2017)
decrease
rs11030102 C BDNF-AS, BDNF 11:27660049 Noncoding transcript exon 0.114 0.036 unit decrease [0.022–0.051] 1  106 Tachmazidou et al.
variant (2017)
rs11030104 A BDNF, BDNF-AS 11:27662970 Intron variant 0.222 0.047 unit increase [0.038–0.058] 2  1020 Wen et al. (2014)
0.042 kg/m2 [0.034–0.049] 7  1030 Locke et al. (2015)
increase
rs16917237 T BDNF-AS, BDNF 11:27680836 Intron variant 0.221 0.040 unit decrease UN 1  1034 Winkler et al. (2015)
rs2030323 C AC103796.1, BDNF 11:27706992 Intron variant 0.239 0.044 kg/m2 [0.034–0.055] 9  1016 Graff et al. (2017)
increase
BDNF Gene as a Precision Skill of Obesity Management

rs2049045 G BDNF-AS, BDNF 11:27672694 Intron variant 0.062 0.042 kg/m2 [0.029–0.057] 9  1010 Graff et al. (2017)
increase
rs4923460 G BDNF-AS 11:27635242 Intron variant 0.254 0.041 kg/m2 [0.029–0.053] 5  1011 Graff et al. (2017)
increase
rs6265 C BDNF-AS, BDNF 11:27658369 Missense variant 0.201 0.040 unit increase [0.036–0.045] 3  1068 Turcot et al. (2018)
G 4.58% SD increase [3.070–6.090] 5  1010 Thorleifsson et al.
(2009)
T 0.04 kg/m2 [0.034–0.046] 2  1051 Akiyama et al. (2017)
decrease
rs7103411 T BDNF, BDNF-AS 11:27678578 Intron variant 0.247 0.041 kg/m2 [0.033–0.05] 2  1022 Graff et al. (2017)
increase
rs7481311 T BDNF-AS 11:27561582 Intron variant 0.247 3.15% SD increase [1.780–4.520] 8  106 Thorleifsson et al.
(2009)
rs925946 T BDNF-AS 11:27645655 Intron variant 0.252 3.85% SD increase [2.620–5.080] 9  1010 Thorleifsson et al.
(2009)
BDNF-AS, BDNF antisense RNA; LINC00678, long intergenic nonprotein coding RNA 678; AC103796.1, novel transcript, sense overlapping BDNF
MAF Minor allele frequency in 1000 genomes combined population, UN Undefined
239
240 H. Marcos-Pasero et al.

insulin, and HOMA-IR levels after adolescents, a borderline statistical significance


biliopancreatic surgery (De et al. 2016) in com- with leptin levels has been reported (Fu et al.
parison with minority allele carriers. The associa- 2017a).
tion between insulin levels and rs10767664 has This genetic variant has also been investigated
also been found in a clinical trial using European as a genetic risk score for the study of BMI and
obese patients after weight loss caused by a sugar-sweetened beverage consumption in two
hypocaloric diet enriched with monounsaturated Swedish cohorts (Brunkwall et al. 2016).
fatty acids (De et al. 2017b) and with standard
hypocaloric diet (De et al. 2018).
This genetic variant has also been included as 15.3.6 rs6265 (Val66Met)
a genetic risk score for the study of BMI, greater
weight gain, and satiety (Llewellyn et al. 2014; The rs6265 (Val66Met) variant is significantly
Badsi et al. 2014; Rukh et al. 2016). associated with BMI in the only meta-analysis
on genetic variants from BDNF/BDNF-AS
(Akbarian et al. 2018) and in Korean adults
15.3.3 rs11030104 (Hong et al. 2012). Intriguingly, there are reports
on differences in sex stratifications. For example,
The association between rs11030104 and BMI Met66 allele is associated with obesity only in
has been reported in the Pakistanis PROMIS Belgian and British women (Beckers et al. 2008;
cohort (Ahmad et al. 2015) and in Korean people Shugart et al. 2009), while in the Boston Puerto
(Lee et al. 2017). Other obesity-related traits were Rican Health Study, men with GG genotype had
investigated in relation with this polymorphism, higher BMI, waist and hip circumferences, and
including the levels of high-density lipoproteins weight than other studied genotypes. An interac-
in adults (Ramos-Lopez et al. 2018), and the tion has been observed between rs6265 and BMI
satiety response in European preschoolers due to the consumption of polyunsaturated fatty
(Monnereau et al. 2017) showing significant asso- acids and with waist circumference (n-3:n-6
ciation for the former but nominal for the later. ratio). On the contrary, in the case of Puerto
Rican women, GG genotypes are associated
with lower BMI, hip circumference, and weight
15.3.4 rs16917237 (Ma et al. 2012). Similar observations have been
documented in European (Zhao et al. 2009), Chi-
This genetic variant has been documented in rela- nese (Wu et al. 2010), American (Mei et al.
tion with eating disorders. Specifically, patients 2012), Mexican (Martínez-Ezquerro et al. 2017),
with bulimia nervosa TT homozygous exhibited and Brazilian (Zandoná et al. 2017) pediatric
higher BMI and minimum registered weight than cohorts.
GT and GG genotypes (Gamero-Villarroel et al. This genetic variant has also been studied in
2014). the context of other obesity-related traits. Adults
that carry this variant allele are associated with
both an increased risk of gaining weight (border-
15.3.5 rs2030323 line statistical significance p ¼ 0.051)
(McCaffery et al. 2013) and an increased intrinsic
Without a direct reference to BMI association, motivation during exercise (Caldwell et al. 2014).
this variant has been studied in the context of On the other hand, this polymorphism is
another obesity-related trait. An association has associated with abdominal obesity in Chinese
been found between energy intake and rs2030323 children and adolescences (Xi et al. 2013; Fu
in the Diabetes Prevention Program, where an et al. 2017b). Met allele has also been associated
increase of 100–150 kcal per risk allele is noted with AN, restrictive AN, binge eating/purging
(McCaffery et al. 2017). In Chinese children and AN as well as BN (Ribasés et al. 2004; Dardennes
15 BDNF Gene as a Precision Skill of Obesity Management 241

et al. 2007; Ribasés et al. 2003), and rumination GENYAL cohort by our research group. We also
(Beevers et al. 2009). However, these results are observed an interaction between rs925946 and
not significant in adolescents and preadolescents BMI as well as calcium and dairy product intake
(Arija et al. 2010). In reference to the diet, there (Marcos-Pasero et al. 2019).
are evidence to support that GG genotypes are Table 15.2 shows SNPs with other than BMI
associated with higher caloric intake added to a associations according to GWAS described
diet containing higher ratio of meat, eggs, nuts, above.
beans, and dairy products (McCaffery et al.
2012), although no documented evidence is
recorded in relation to the intake of any macronu- 15.4 Precision Nutrition
trient and rs6265 (Park et al. 2013). for Personalizing Nutritional
This genetic variant has also been included in Recommendations
two genetic risk scores for the study of childhood
BMI (Klimentidis et al. 2011; Mitchell et al. The Precision Nutrition (PN) is a modern and
2013). multidimensional concept, which contains indi-
vidual characteristics that define the phenotype
(e.g., sex, age, current diseases, biochemical
15.3.7 rs925946 parameters, etc.), behavioral lifestyle factors
(e.g., dietary habits and patterns, food behavior,
This genetic variant has been associated with physical activity, drugs, stress, etc.), preferences
obesity, weight, and BMI in European, (e.g., food, culinary techniques, flavors, etc.), and
European-American, and African-American genetics. Other conditions, such as resources,
cohorts (Thorleifsson et al. 2009; Sandholt et al. environmental, and sustainability, are also con-
2010) as well as with the only BMI-related meta- sidered in PN (Corella and Ordovás 2018). Thus,
analysis on genetic variants from BDNF/BDNF- the PN emerges due to the integration of many
AS (Akbarian et al. 2018) and with a greater fields of investigation and with a broad definition.
impact on adults than children (den Hoed et al. The origin of PN emerged as a part of Preci-
2010), which could explain the contradictory sion Medicine focused on discoursing the hetero-
results in pediatric populations. No significant geneity in nutrient metabolism between
association has been found with BMI in a subgroups and the inter-individual variability in
European-American descent pediatric cohort responses to diet interventions (Ramos-Lopez
(Zhao et al. 2009), nor in the European Inter99 et al. 2017).
cohort (Andersson et al. 2010). However, in The pillars of PN have been categorized by the
French and German populations, there is an asso- International Society of Nutrigenetics/
ciation with the early onset of extreme obesity Nutrigenomics (ISNN) in stratified,
(Scherag et al. 2010), and weight as well as with individualized, and genotype-directed nutrition
height and BMI in the European ALSPAC cohort in order to show the role of genetic variations
(Elks et al. 2010). An association has also been and individual dietary responses, as well as the
found for babies being born small for gestational role of nutrients in gene expression (de Toro-
age (Morgan et al. 2010). Martín et al. 2017).
In adolescents, rs925946 is associated with the The main goal of the PN is to prevent diseases
metabolic syndrome in Europeans (Dušátková and formulate solid and personalized nutritional
et al. 2013), and with BMI in Australians (border- prescriptions, in other words to establish tailored
line significance p ¼ 0,053) (Liu et al. 2010). dietary instructions regarding multiple variables
Also, adolescents’ carriers of the minor allele to satisfactorily anticipate individual responses to
appeared to absorb less dietetic calcium diet. Nevertheless, clinicians show uncertainty in
(Dušátková et al. 2015). Similar results have the use of PN recommendations, so it is necessary
recently been reported in children of the Spanish further interventional trials to be conducted in
242 H. Marcos-Pasero et al.

Table 15.2 Other GWAS-related associations between BDNF/BDNF-AS SNPs


rs10501087 Morning person (Kichaev et al. 2019)
rs10767654 Waist circumference (Tachmazidou et al. 2017)
rs11030102 Weight (Tachmazidou et al. 2017)
Educational attainment (Lee et al. 2018)
rs11030104 BMI in smokers (Justice et al. 2017)
BMI in nonsmokers (Justice et al. 2017)
Grip strength (Tikkanen et al. 2018)
rs16917237 Coronary artery disease (van der Harst and Verweij 2018)
Age of onset of menopause (Horikoshi et al. 2018)
Age of onset of menarche (Kichaev et al. 2019)
rs2030323 Obesity (Berndt et al. 2013)
rs2049045 Snoring (Jansen et al. 2019)
Tobacco cessation (Liu et al. 2019)
Risk tolerance (Karlsson Linnér et al. 2019)
rs6265 Hip circumference (Shungin et al. 2015)
Smoker status (Liu et al. 2019)
Smoking behavior (Tobacco and Genetics Consortium 2010)
Age of tobacco use initiation (Liu et al. 2019)
Weight (Thorleifsson et al. 2009)
Risky behaviors (Karlsson Linnér et al. 2019)
Cigarettes smoked per day (Liu et al. 2019)
Visceral adipose tissue (Karlsson et al. 2019)
rs7103411 Age of onset of menarche (Perry et al. 2014)
rs7481311 Weight (Thorleifsson et al. 2009)
rs925946 Diastolic blood pressure (Surendran et al. 2016)
Weight (Thorleifsson et al. 2009)

order to increase the level of evidence, strength, part of the GENYAL study to childhood obesity
and reproducibility of the findings (Laddu and prevention (www.clinicaltrials.gov
Hauser 2019). The Food4Me project is focused NCT03419520). A total of 221 schoolchildren
on this area. It tries to establish the relevant from 6 different public schools among the Com-
modus operandi of PN in preventing and manag- munity of Madrid, Spain, were included. 52.50%
ing chronic diseases such as obesity (de Toro- were girls (n ¼ 116) and 47.50% boys (n ¼ 105)
Martín et al. 2017). between 6 and 9 years old. Anthropometric and
On the basis of genomic background, studying dietary data were collected, and children were
gene-diet interactions in obesity requires more genotyped according to the rs925946 polymor-
exhaustive analyses including different kinds of phism obtained from saliva samples. An inverse
individual phenotypes, genotypes, and dietary significant association was observed between the
factors (Heianza and Qi 2017). schoolchildren’s BMI and the mg of calcium
In this context, our research group performed intake/day (β ¼ 0.002 (0.003, 0.0003)
the first study on the association between the p ¼ 0.019) (data adjusted for sex and age). The
BDNF-AS rs925946 polymorphism and calcium distribution caused by genotypes according to the
intake as potential modulators of the nutritional genetic variant rs925946 showed that 55.91% of
status. The objective of this study was to evaluate the participants were GG common homozygous
the association between calcium/dairy intake, the (n ¼ 123), 36.82% heterozygous (n ¼ 81), and
BDNF-AS rs925946 polymorphism, and 7.27% TT homozygous (n ¼ 16). No significant
nutritional status in a group of schoolchildren as differences were observed regarding the BMI
15 BDNF Gene as a Precision Skill of Obesity Management 243

between both genotypes (BMI References


(GG) ¼ 16.97  0.47 kg/m2 and
(GT + TT) ¼ 17.00  0.540 kg/m2). Ahmad S, Zhao W, Renström F, Rasheed A, Samuel M,
A significant interaction was observed Zaidi M et al (2015) Physical activity, smoking, and
genetic predisposition to obesity in people from
between the genetic variant and the BMI. Indeed, Pakistan: the PROMIS study. BMC Med Genet [Inter-
GG common homozygous reduced on average net] 16:114–114. Available from: https://europepmc.
0.34 kg/m2 the BMI for each 100 mg of calcium org/articles/PMC4683724/ [cited 2019 Oct 31]
(β ¼ 0.003 (0.006, 0.001), p ¼ 0.004). Akbarian S-A, Salehi-Abargouei A, Pourmasoumi M,
Kelishadi R, Nikpour P, Heidari-Beni M (2018) Asso-
However, this effect was not observed in T allele ciation of Brain-derived neurotrophic factor gene
carriers, where the consumption of 100 mg of polymorphisms with body mass index: A systematic
calcium was not associated with the BMI review and meta-analysis. Adv Med Sci [Internet] 63
(β ¼ 1.3e-4 (0.0022, 0.0024), p ¼ 0.93). (1):43–56. Available from: http://www.sciencedirect.
com/science/article/pii/S1896112617300536 [cited
To our knowledge, this is the first time that the 2019 Oct 30]
association between BDNF-AS rs925946 and cal- Akiyama M, Okada Y, Kanai M, Takahashi A,
cium intake as potential modulators of the Momozawa Y, Ikeda M et al (2017) Genome-wide
nutritional status has been described, which association study identifies 112 new loci for body
mass index in the Japanese population. Nat Genet
opens up the avenue for future investigations in [Internet] 49:1458. Available from: https://doi.org/10.
this area. We hypothesize that the presence of the 1038/ng.3951
SNP may cause failures in the control of the Alharbi KK, Richardson TG, Khan IA, Syed R,
energy balance and conditioning the effect of Mohammed AK, Boustred CR et al (2014) Influence
of adiposity-related genetic markers in a population of
calcium on satiety or on other mechanisms Saudi Arabians where other variables influencing obe-
involved in body weight control which may pro- sity may be reduced. Dis Markers [Internet]
mote obesity development. The determination of 2014:758232. Available from: https://europepmc.org/
differential effects of calcium/dairy consumption articles/PMC4251424/ [cited 2019 Oct 31]
Andersson EA, Pilgaard K, Pisinger C, Harder MN,
on the nutritional status in accordance with genet- Grarup N, Færch K et al (2010) Do gene variants
ics may contribute to the personalization of future influencing adult adiposity affect birth weight? A
nutritional advice. Nevertheless, these results population-based study of 24 loci in 4,744 Danish
should be confirmed in future prospective studies individuals. PloS One [Internet] 5(12):e14190–
e14190. Available from: https://europepmc.org/
with a larger sample size (Marcos-Pasero et al. articles/PMC2995733/ [cited 2019 Nov 5]
2019). Arija V, Ferrer-Barcala M, Aranda N, Canals J (2010)
BDNF Val66Met polymorphism, energy intake and
BMI: a follow-up study in schoolchildren at risk of
eating disorders. BMC Public Health [Internet]
15.5 Conclusion and Future 10:363–363. Available from: http://europepmc.org/
Perspectives abstract/MED/20573217 [cited 2019 Nov 4]
Badsi MN, Mediene-Benchekor S, Ouhaibi-Djellouli H,
Lardjam-Hetraf SA, Boulenouar H, Meroufel DN et al
The identification of diverse individual nutritional (2014) Combined effect of established BMI loci on
needs and responses to diet is changing standard obesity-related traits in an Algerian population sample.
of nutritional care. The integration of all the BMC Genet [Internet] 15:128–128. Available from:
https://europepmc.org/articles/PMC4247883/ [cited
factors involved (phenotype, lifestyle factors, 2019 Oct 31]
preferences, and, indisputably, genetic) will cre- Beckers S, Peeters A, Zegers D, Mertens I, Gaal LV, Van
ate new treatment options for obesity and other Hul W (2008) Association of the BDNF Val66Met
complex diseases. variation with obesity in women. Mol Genet Metab
[Internet] 95(1):110–112. Available from: http://
www.sciencedirect.com/science/article/pii/
Acknowledgments MPI is supported by the TALENTO S1096719208001741 [cited 2019 Nov 4]
Program from the Regional Madrid Government (2018- Beevers CG, Wells TT, JE MG (2009) The BDNF
T1/BIO-11262). Val66Met polymorphism is associated with rumination
in healthy adults. Emot Wash DC [Internet] 9
244 H. Marcos-Pasero et al.

(4):579–584. Available from: http://europepmc.org/ europepmc.org/abstract/MED/29339649 [cited 2019


abstract/MED/19653783 [cited 2019 Nov 4] Oct 31]
Berndt SI, Gustafsson S, Mägi R, Ganna A, Wheeler E, De DL, Aller R, Izaola O, Primo D, Romero E (2017b)
Feitosa MF et al (2013 May) Genome-wide meta-anal- rs10767664 gene variant in brain-derived neurotrophic
ysis identifies 11 new loci for anthropometric traits and factor is associated with diabetes mellitus type 2 in
provides insights into genetic architecture. Nat Genet Caucasian females with obesity. Ann Nutr Metab
45(5):501–512 [Internet] 70(4):286–292. Available from: http://
Briana DD, Malamitsi-Puchner A (2018) Developmental europepmc.org/abstract/MED/28595187 [cited 2019
origins of adult health and disease: the metabolic role Oct 31]
of BDNF from early life to adulthood. Metabolism De DL, Fernández HO, Izaola O, Primo D, Aller R (2018)
81:45–51 RS 10767664 gene variant in Brain Derived
Brunkwall L, Chen Y, Hindy G, Rukh G, Ericson U, Neurotrophic Factor (BDNF) affect metabolic changes
Barroso I et al (2016) Sugar-sweetened beverage con- and insulin resistance after a standard hypocaloric diet.
sumption and genetic predisposition to obesity in J Diabetes Complications [Internet] 32(2):216–220.
2 Swedish cohorts. Am J Clin Nutr [Internet] 104 Available from: http://europepmc.org/abstract/MED/
(3):809–815. Available from: https://europepmc.org/ 29174117 [cited 2019 Oct 31]
articles/PMC4997292/ [cited 2019 Nov 4] den Hoed M, Ekelund U, Brage S, Grontved A, Zhao JH,
Caldwell AH, Bryan AD, Hagger MS (2014) What keeps a Sharp SJ et al (2010 Nov) Genetic susceptibility to
body moving? The brain-derived neurotrophic factor obesity and related traits in childhood and adolescence:
val66met polymorphism and intrinsic motivation to influence of loci identified by genome-wide association
exercise in humans. J Behav Med [Internet] 37 studies. Diabetes 59(11):2980–2988
(6):1180–1192. Available from: http://europepmc.org/ Dušátková L, Zamrazilová H, Sedláčková B, Včelák J,
abstract/MED/24805993 [cited 2019 Nov 4] Hlavatý P, Aldhoon Hainerová I et al (2013) Associa-
Chen Z-Y, Patel PD, Sant G, Meng C-X, Teng KK, tion of obesity susceptibility gene variants with meta-
Hempstead BL et al (2004) Variant brain-derived bolic syndrome and related traits in 1,443 Czech
neurotrophic factor (BDNF) (Met66) alters the intra- adolescents. Folia Biol (Praha) 59(3):123–133
cellular trafficking and activity-dependent secretion of Dušátková L, Zamrazilová H, Aldhoon-Hainerová I,
wild-type BDNF in neurosecretory cells and cortical Sedláčková B, Včelák J, Hlavatý P et al (2015) A
neurons. J Neurosci 24(18):4401–4411 common variant near BDNF is associated with dietary
Corella D, Ordovás JM (2018) The role of omics in preci- calcium intake in adolescents. Nutr Res N Y N 35
sion nutrition: strengths and weaknesses. Nutr Hosp. (9):766–773
35(Spec No 4):10–18 Elks CE, Loos RJF, Sharp SJ, Langenberg C, Ring SM,
Dardennes RM, Zizzari P, Tolle V, Foulon C, Kipman A, Timpson NJ et al (2010) Genetic markers of adult
Romo L et al (2007) Family trios analysis of common obesity risk are associated with greater early infancy
polymorphisms in the obestatin/ghrelin, BDNF and weight gain and growth. PLoS Med 7(5):e1000284
AGRP genes in patients with Anorexia nervosa: asso- Fernández-Rhodes L, Demerath EW, Cousminer DL,
ciation with subtype, body-mass index, severity and Tao R, Dreyfus JG, Esko T et al (2013) Association
age of onset. Psychoneuroendocrinology [Internet] 32 of adiposity genetic variants with menarche timing in
(2):106–113. Available from: http://europepmc.org/ 92,105 women of European descent. Am J Epidemiol
abstract/MED/17197106 [cited 2019 Nov 4] [Internet] 178(3):451–460. Available from: https://
de Toro-Martín J, Arsenault BJ, Després J-P, Vohl M-C europepmc.org/articles/PMC3816344/ [cited 2019 Oct
(2017 Aug) Precision nutrition: a review of 31]
personalized nutritional approaches for the prevention Fu J, Li G, Li L, Yin J, Cheng H, Han L et al (2017a) The
and Management of Metabolic Syndrome. Nutrients role of established East Asian obesity-related loci on
22:9(8) pediatric leptin levels highlights a neuronal influence
De DL, Izaola O, Primo D, Pacheco D (2016) Effect of the on body weight regulation in Chinese children and
rs10767664 variant of the brain-derived neurotrophic adolescents: the BCAMS study. Oncotarget [Internet]
factor gene on weight change and cardiovascular risk 8(55):93593–93607. Available from: https://
factors in morbidly obese patients after biliopancreatic europepmc.org/articles/PMC5706821/ [cited 2019
diversion surgery. J Nutr Nutr [Internet] 9 Nov 4]
(2–4):116–122. Available from: http://europepmc.org/ Fu LW, Zhang MX, Wu LJ, Gao LW, Mi J (2017b) Gene-
abstract/MED/27544754 [cited 2019 Oct 31] gene interaction on central obesity in school-aged chil-
De DL, Romero E, Izaola O, Primo D, Aller R (2017a) dren in China. Zhonghua Liu Xing Bing Xue Za Zhi
Cardiovascular risk factors and insulin resistance after Zhonghua Liuxingbingxue Zazhi [Internet] 38
two hypocaloric diets with different fat distribution in (7):883–888. Available from: http://europepmc.org/
obese subjects: effect of the rs10767664 gene variant in abstract/MED/28738459 [cited 2019 Nov 4]
brain-derived neurotrophic factor. J Nutr Nutr [Inter- Gamero-Villarroel C, Gordillo I, Carrillo JA, García-
net] 10(5–6):163–171. Available from: http:// Herráiz A, Flores I, Jiménez M et al (2014) BDNF
genetic variability modulates psychopathological
15 BDNF Gene as a Precision Skill of Obesity Management 245

symptoms in patients with eating disorders. Eur Child Juonala M, Juhola J, Magnussen CG, Würtz P, Viikari JS,
Adolesc Psychiatry [Internet] 23(8):669–679. Avail- Thomson R et al (2011) Childhood environmental and
able from: https://doi.org/10.1007/s00787-013-0495-6 genetic predictors of adulthood obesity: the cardiovas-
[cited 2019 Oct 31] cular risk in young Finns study. J Clin Endocrinol
Goodarzi MO (2018) Genetics of obesity: what genetic Metab [Internet] 96(9):E1542–E1549. Available
association studies have taught us about the biology of from: https://europepmc.org/articles/PMC3167668/
obesity and its complications. Lancet Diabetes [cited 2019 Oct 30]
Endocrinol 6(3):223–236 Justice AE, Winkler TW, Feitosa MF, Graff M, Fisher VA,
Graff M, Scott RA, Justice AE, Young KL, Feitosa MF, Young K et al (2017) Genome-wide meta-analysis of
Barata L et al (2017) Genome-wide physical activity 241,258 adults accounting for smoking behaviour
interactions in adiposity ― A meta-analysis of 200,452 identifies novel loci for obesity traits. Nat Commun
adults. PLOS Genet [Internet] 13(4):e1006528. Avail- 8:14977
able from: https://doi.org/10.1371/journal.pgen. Karlsson Linnér R, Biroli P, Kong E, Meddens SFW,
1006528 Wedow R, Fontana MA et al (2019) Genome-wide
Guo Y, Lanktree MB, Taylor KC, Hakonarson H, Lange association analyses of risk tolerance and risky
LA, Keating BJ (2013) Gene-centric meta-analyses of behaviors in over 1 million individuals identify
108 912 individuals confirm known body mass index hundreds of loci and shared genetic influences. Nat
loci and reveal three novel signals. Hum Mol Genet Genet 51(2):245–257
[Internet] 22(1):184–201. Available from: https:// Karlsson T, Rask-Andersen M, Pan G, Höglund J,
europepmc.org/articles/PMC3522401/ [cited 2019 Wadelius C, Ek WE et al (2019) Contribution of genet-
Oct 31] ics to visceral adiposity and its relation to cardiovascu-
Han JC (2016) Rare syndromes and common variants of lar and metabolic disease. Nat Med 25(9):1390–1395
the brain-derived Neurotrophic factor gene in human Kichaev G, Bhatia G, Loh P-R, Gazal S, Burch K, Freund
obesity. Prog Mol Biol Transl Sci 140:75–95 MK et al (2019) Leveraging polygenic functional
Han JC, Liu Q-R, Jones M, Levinn RL, Menzie CM, enrichment to improve GWAS power. Am J Hum
Jefferson-George KS et al (2008) Brain-derived Genet 104(1):65–75
neurotrophic factor and obesity in the WAGR syn- Klimentidis YC, Chen GB, López-Alarcón M, Harris JJ,
drome. N Engl J Med 359(9):918–927 Duarte CW, Fernández JR (2011) Associations of obe-
Hebbring S (2019) Genomic and Phenomic research in the sity genes with obesity-related outcomes in multiethnic
21st century. Trends Genet [Internet] 35(1):29–41. children. Arch Med Res [Internet] 42(6):509–514.
Available from: http://www.sciencedirect.com/sci Available from: https://europepmc.org/articles/
ence/article/pii/S0168952518301689 [cited 2019 Oct PMC3541020/ [cited 2019 Nov 4]
28] Laddu D, Hauser M (2019) Addressing the nutritional
Heianza Y, Qi L (2017) Gene-diet interaction and preci- phenotype through personalized nutrition for chronic
sion nutrition in obesity. Int J Mol Sci 18(4):787 disease prevention and management. Prog Cardiovasc
Hong K-W, Lim J-E, Go MJ, Shin YS, Ahn Y, Han B-G Dis 62(1):9–14
et al (2012) Recapitulation of the association of the Lapchak PA, Hefti F (1992) BDNF and NGF treatment in
Val66Met polymorphism of BDNF gene with BMI in lesioned rats: effects on cholinergic function and
Koreans. Obesity [Internet] 20(9):1871–1875. Avail- weight gain. Neuroreport 3(5):405–408
able from: https://www.onlinelibrary.wiley.com/doi/ Lebrun B, Bariohay B, Moyse E, Jean A (2006) Brain-
abs/10.1038/oby.2011.352 [cited 2019 Nov 4] derived neurotrophic factor (BDNF) and food intake
Horikoshi M, Day FR, Akiyama M, Hirata M, regulation: a minireview. Auton Neurosci Basic Clin
Kamatani Y, Matsuda K et al (2018) Elucidating the 126–127:30–38
genetic architecture of reproductive ageing in the Japa- Lee JS, Cheong HS, Shin HD (2017) BMI prediction
nese population. Nat Commun 9(1):1977 within a Korean population. PeerJ [Internet] 5:e3510–
Hruby A, Hu FB (2015) The epidemiology of obesity: a e3510. Available from: https://europepmc.org/articles/
big picture. PharmacoEconomics 33(7):673–689 PMC5493974/ [cited 2019 Oct 31]
Jansen PR, Watanabe K, Stringer S, Skene N, Bryois J, Lee JJ, Wedow R, Okbay A, Kong E, Maghzian O, Zacher
Hammerschlag AR et al (2019) Genome-wide analysis M et al (2018) Gene discovery and polygenic predic-
of insomnia in 1,331,010 individuals identifies new tion from a genome-wide association study of educa-
risk loci and functional pathways. Nat Genet 51 tional attainment in 1.1 million individuals. Nat Genet
(3):394–403 50(8):1112–1121
Jiao H, Arner P, Hoffstedt J, Brodin D, Dubern B, Liu JZ, Medland SE, Wright MJ, Henders AK, Heath AC,
Czernichow S et al (2011) Genome wide association Madden PA et al (2010) Genome-wide association
study identifies KCNMA1 contributing to human obe- study of height and body mass index in Australian
sity. BMC Med Genomics [Internet] 4:51. Available twin families. Twin Res Hum Genet Off J Int Soc
from: https://europepmc.org/articles/PMC3148553/ Twin Stud [Internet] 13(2):179–193. Available from:
[cited 2019 Oct 30] https://europepmc.org/articles/PMC3232006/ [cited
2019 Nov 5]
246 H. Marcos-Pasero et al.

Liu M, Jiang Y, Wedow R, Li Y, Brazel DM, Chen F et al McCaffery JM, Jablonski KA, Franks PW, Delahanty LM,
(2019) Association studies of up to 1.2 million Aroda V, Marrero D et al (2017) Replication of the
individuals yield new insights into the genetic etiology association of BDNF and MC4R variants with dietary
of tobacco and alcohol use. Nat Genet 51(2):237–244 intake in the diabetes prevention program. Psychosom
Llewellyn CH, Trzaskowski M, Van CJ, Plomin R, Wardle Med [Internet] 79(2):224–233. Available from: https://
J (2014) Satiety mechanisms in genetic risk of obesity. europepmc.org/articles/PMC5285480/ [cited 2019
JAMA Pediatr [Internet] 168(4):338–344. Available Nov 4]
from: https://europepmc.org/articles/PMC3981891/ Mei H, Chen W, Jiang F, He J, Srinivasan S, Smith EN
[cited 2019 Oct 31] et al (2012) Longitudinal replication studies of GWAS
Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day risk SNPs influencing body mass index over the course
FR et al (2015) Genetic studies of body mass index of childhood and adulthood. PLoS One 7(2):e31470
yield new insights for obesity biology. Nature 518 Mitchell JA, Hakonarson H, Rebbeck TR, Grant SFA
(7538):197–206 (2013 Jun) Obesity-susceptibility loci and the tails of
Loos RJ (2018) The genetics of adiposity. Mol Genet the pediatric BMI distribution. Obes Silver Spring Md
Basis Metab Dis [Internet] 50:86–95. Available from: 21(6):1256–1260
http://www.sciencedirect.com/science/article/pii/ Monnereau C, Jansen PW, Tiemeier H, Jaddoe VW, Felix
S0959437X17301259 JF (2017) Influence of genetic variants associated with
Ma XY, Qiu WQ, Smith CE, Parnell LD, Jiang ZY, body mass index on eating behavior in childhood.
Ordovas JM et al (2012) Association between BDNF Obes Silver Spring Md [Internet] 25(4):765–772.
rs6265 and obesity in the Boston Puerto Rican health Available from: https://europepmc.org/articles/
study. J Obes [Internet] 2012:102942. Available from: PMC5496668/ [cited 2019 Oct 31]
http://europepmc.org/abstract/MED/23326649 [cited Monteleone P, Fabrazzo M, Martiadis V, Serritella C,
2019 Nov 4] Pannuto M, Maj M (2005 Jun) Circulating brain-
Marcos-Pasero H, Aguilar-Aguilar E, de la Iglesia R, derived neurotrophic factor is decreased in women
Espinosa-Salinas I, Gómez-Patiño M, Colmenarejo G with anorexia and bulimia nervosa but not in women
et al (2019) Association of calcium and dairy product with binge-eating disorder: relationships to co-morbid
consumption with childhood obesity and the presence depression, psychopathology and hormonal variables.
of a brain derived neurotropic factor-antisense (BDNF- Psychol Med 35(6):897–905
AS) polymorphism. Clin Nutr 38(6):2616–2622 Morgan AR, Thompson JM, Murphy R, Black PN, Lam
Marosi K, Mattson MP (2014) BDNF mediates adaptive WJ, Ferguson LR et al (2010) Obesity and diabetes
brain and body responses to energetic challenges. genes are associated with being born small for gesta-
Trends Endocrinol Metab TEM 25(2):89–98 tional age: results from the Auckland Birthweight Col-
Martínez-Ezquerro JD, Rendón-Macías ME, Zamora- laborative study. BMC Med Genet [Internet] 11:125.
Mendoza G, Serrano-Meneses J, Rosales- Available from: https://europepmc.org/articles/
Rodríguez B, Escalante-Bautista D et al (2017) Asso- PMC2928774/ [cited 2019 Nov 5]
ciation between the brain-derived neurotrophic factor Murphy RA, Nalls MA, Keller M, Garcia M, Kritchevsky
Val66Met polymorphism and overweight/obesity in SB, Tylavsky FA et al (2013) Candidate gene associa-
pediatric population. Arch Med Res [Internet] 48 tion study of BMI-related loci, weight, and adiposity in
(7):599–608. Available from: http://europepmc.org/ old age. J Gerontol A Biol Sci Med Sci [Internet] 68
abstract/MED/29506764 [cited 2019 Nov 4] (6):661–666. Available from: https://europepmc.org/
Martínez-García F, Mansego ML, Rojo-Martínez G, De articles/PMC3660116/ [cited 2019 Oct 30]
GM-S, Morcillo S, Soriguer F et al (2013) Impact of Nakagawa T, Tsuchida A, Itakura Y, Nonomura T,
obesity-related genes in Spanish population. BMC Ono M, Hirota F et al (2000) Brain-derived
Genet [Internet] 14:111. Available from: https:// neurotrophic factor regulates glucose metabolism by
europepmc.org/articles/PMC4222487/ [cited 2019 modulating energy balance in diabetic mice. Diabetes
Oct 30] 49(3):436–444
McCaffery JM, Papandonatos GD, Peter I, Huggins GS, Nakazato M, Hashimoto K, Shimizu E, Kumakiri C,
Raynor HA, Delahanty LM et al (2012) Obesity sus- Koizumi H, Okamura N et al (2003) Decreased levels
ceptibility loci and dietary intake in the look ahead of serum brain-derived neurotrophic factor in female
trial. Am J Clin Nutr [Internet] 95(6):1477–1486. patients with eating disorders. Biol Psychiatry 54
Available from: https://europepmc.org/articles/ (4):485–490
PMC3349457/ [cited 2019 Oct 30] Ng MCY, Graff M, Lu Y, Justice AE, Mudgal P, Liu C-T
McCaffery JM, Papandonatos GD, Huggins GS, Peter I, et al (2017) Discovery and fine-mapping of adiposity
Kahn SE, Knowler WC et al (2013) FTO predicts loci using high density imputation of genome-wide
weight regain in the look AHEAD clinical trial. Int J association studies in individuals of African ancestry:
Obes 2005 [Internet] 37(12):1545–1552. Available African Ancestry Anthropometry Genetics Consor-
from: https://europepmc.org/articles/PMC3750057/ tium. PLOS Genet [Internet] 13(4):e1006719. Avail-
[cited 2019 Oct 31] able from: https://doi.org/10.1371/journal.pgen.
1006719
15 BDNF Gene as a Precision Skill of Obesity Management 247

Park SL, Cheng I, Pendergrass SA, Kucharska-Newton Available from: https://europepmc.org/articles/


AM, Lim U, Ambite JL et al (2013) Association of PMC4808432/ [cited 2019 Oct 30]
the FTO obesity risk variant rs8050136 with percent- Rukh G, Ahmad S, Ericson U, Hindy G, Stocks T,
age of energy intake from fat in multiple racial/ethnic Renström F et al (2016) Inverse relationship between
populations: the PAGE study. Am J Epidemiol [Inter- a genetic risk score of 31 BMI loci and weight change
net] 178(5):780–790. Available from: https:// before and after reaching middle age. Int J Obes 2005
europepmc.org/articles/PMC3755639/ [cited 2019 [Internet] 40(2):252–259. Available from: https://
Nov 4] europepmc.org/articles/PMC4753358/ [cited 2019
Paternoster L, Evans DM, Nohr EA, Holst C, Oct 31]
Gaborieau V, Brennan P et al (2011) Genome-wide Sandholt CH, Sparsø T, Grarup N, Albrechtsen A,
population-based association study of extremely over- Almind K, Hansen L et al (2010) Combined analyses
weight young adults--the GOYA study. PloS One of 20 common obesity susceptibility variants. Diabetes
[Internet] 6(9):e24303. Available from: https:// [Internet] 59(7):1667–1673. Available from: https://
europepmc.org/articles/PMC3174168/ [cited 2019 europepmc.org/articles/PMC2889766/ [cited 2019
Oct 30] Nov 5]
Pelleymounter MA, Cullen MJ, Wellman CL (1995) Sandrini L, Di Minno A, Amadio P, Ieraci A, Tremoli E,
Characteristics of BDNF-induced weight loss. Exp Barbieri SS (2018) Association between obesity and
Neurol 131(2):229–238 circulating brain-derived Neurotrophic factor (BDNF)
Perry JR, Day F, Elks CE, Sulem P, Thompson DJ, levels: systematic review of literature and meta-
Ferreira T et al (2014) Parent-of-origin-specific allelic analysis. Int J Mol Sci 19(8):2281
associations among 106 genomic loci for age at men- Scherag A, Dina C, Hinney A, Vatin V, Scherag S, Vogel
arche. Nature 514(7520):92–97 CI et al (2010) Two new Loci for body-weight regula-
Ramos-Lopez O, Milagro FI, Allayee H, Chmurzynska A, tion identified in a joint analysis of genome-wide asso-
Choi MS, Curi R et al (2017) Guide for current ciation studies for early-onset extreme obesity in
Nutrigenetic, Nutrigenomic, and Nutriepigenetic French and German study groups. PLoS Genet [Inter-
approaches for precision nutrition involving the pre- net] 6(4):e1000916. Available from: https://
vention and Management of Chronic Diseases europepmc.org/articles/PMC2858696/ [cited 2019
Associated with obesity. J Nutr Nutr 10(1–2):43–62 Oct 30]
Ramos-Lopez O, Riezu-Boj JI, Milagro FI, Cuervo M, Shugart YY, Chen L, Day IN, Lewis SJ, Timpson NJ,
Goni L, Martinez JA (2018) Prediction of blood lipid Yuan W et al (2009) Two British women studies
phenotypes using obesity-related genetic replicated the association between the Val66Met poly-
polymorphisms and lifestyle data in subjects with morphism in the brain-derived neurotrophic factor
excessive body weight. Int J Genomics [Internet] (BDNF) and BMI. Eur J Hum Genet EJHG [Internet]
2018:4283078. Available from: https://europepmc. 17(8):1050–1055. Available from: http://europepmc.
org/articles/PMC6276413/ [cited 2019 Oct 31] org/abstract/MED/19209189 [cited 2019 Nov 4]
Ribasés M, Gratacòs M, Armengol L, De RC, Badía A, Shungin D, Winkler TW, Croteau-Chonka DC, Ferreira T,
Jiménez L et al (2003) Met66 in the brain-derived Locke AE, Mägi R et al (2015) New genetic loci link
neurotrophic factor (BDNF) precursor is associated adipose and insulin biology to body fat distribution.
with anorexia nervosa restrictive type. Mol Psychiatry Nature 518(7538):187–196
[Internet] 8(8):745–751. Available from: http:// Speliotes EK, Willer CJ, Berndt SI, Monda KL,
europepmc.org/abstract/MED/12888803 [cited 2019 Thorleifsson G, Jackson AU et al (2010) Association
Nov 4] analyses of 249,796 individuals reveal 18 new loci
Ribasés M, Gratacòs M, Fernández-Aranda F, Bellodi L, associated with body mass index. Nat Genet 42
Boni C, Anderluh M et al (2004) Association of BDNF (11):937–948
with anorexia, bulimia and age of onset of weight loss Surendran P, Drenos F, Young R, Warren H, Cook JP,
in six European populations. Hum Mol Genet [Inter- Manning AK et al (2016) Trans-ancestry meta-
net] 13(12):1205–1212. Available from: http:// analyses identify rare and common variants associated
europepmc.org/abstract/MED/15115760 [cited 2019 with blood pressure and hypertension. Nat Genet 48
Nov 4] (10):1151–1161
Rios M (2014) Neurotrophins and the regulation of energy Tachmazidou I, Süveges D, Min JL, Ritchie GRS,
balance and body weight. Handb Exp Pharmacol Steinberg J, Walter K et al (2017) Whole-genome
220:283–307 sequencing coupled to imputation discovers genetic
Rosas-Vargas H, Martínez-Ezquerro JD, Bienvenu T signals for anthropometric traits. Am J Hum Genet
(2011) Brain-derived neurotrophic factor, food intake [Internet] 100(6):865–884. Available from: http://
regulation, and obesity. Arch Med Res 42(6):482–494 www.sciencedirect.com/science/article/pii/
Ruiz-Narváez EA, Haddad SA, Rosenberg L, Palmer JR S0002929717301593
(2016) Birth weight modifies the association between Thorleifsson G, Walters GB, Gudbjartsson DF,
central nervous system gene variation and adult body Steinthorsdottir V, Sulem P, Helgadottir A et al
mass index. J Hum Genet [Internet] 61(3):193–198. (2009) Genome-wide association yields new sequence
248 H. Marcos-Pasero et al.

variants at seven loci that associate with measures of index. Hum Mol Genet [Internet] 23(20):5492–5504.
obesity. Nat Genet 41(1):18–24 Available from: https://doi.org/10.1093/hmg/ddu248
Tikkanen E, Gustafsson S, Amar D, Shcherbina A, [cited 2019 Oct 29]
Waggott D, Ashley EA et al (2018) Biological insights Winkler TW, Justice AE, Graff M, Barata L, Feitosa MF,
into muscular strength: genetic findings in the UK Chu S et al (2015) The influence of age and sex on
Biobank. Sci Rep 8(1):6451 genetic associations with adult body size and shape: a
Tobacco and Genetics Consortium (2010) Genome-wide large-scale genome-wide interaction study. PLOS
meta-analyses identify multiple loci associated with Genet [Internet] 11(10):e1005378. Available from:
smoking behavior. Nat Genet 42(5):441–447 https://doi.org/10.1371/journal.pgen.1005378
Turcot V, Lu Y, Highland HM, Schurmann C, Justice AE, World Health Organization (WHO). Obesity and Over-
Fine RS et al (2018) Protein-altering variants weight. [Internet]. [cited 2019 Nov 6]. Available
associated with body mass index implicate pathways from: https://www.who.int/news-room/factsheets/
that control energy intake and expenditure in obesity. detail/obesity-and-overweight
Nat Genet [Internet] 50(1):26–41. Available from: Wolfe D, Dudek S, Ritchie MD, Pendergrass SA (2013)
https://doi.org/10.1038/s41588-017-0011-x Visualizing genomic information across chromosomes
van der Harst P, Verweij N (2018 Feb 02) Identification of with PhenoGram. BioData Min [Internet] 6:18. Avail-
64 novel genetic loci provides an expanded view on the able from: https://www.ncbi.nlm.nih.gov/pmc/articles/
genetic architecture of coronary artery disease. Circ PMC4015356/ [cited 2019 Oct 30]
Res 122(3):433–443 Wu L, Xi B, Zhang M, Shen Y, Zhao X, Cheng H et al
Vimaleswaran KS, Tachmazidou I, Zhao JH, Hirschhorn (2010) Associations of six single nucleotide
JN, Dudbridge F, Loos RJ (2012) Candidate genes for polymorphisms in obesity-related genes with BMI
obesity-susceptibility show enriched association and risk of obesity in Chinese children. Diabetes [Inter-
within a large genome-wide association study for net] 59(12):3085–3089. Available from: http://
BMI. Hum Mol Genet [Internet] 21(20):4537–4542. europepmc.org/abstract/MED/20843981 [cited 2019
Available from: https://europepmc.org/articles/ Nov 4]
PMC3607467/ [cited 2019 Oct 30] Xi B, Cheng H, Shen Y, Chandak GR, Zhao X, Hou D et al
Visscher PM, Wray NR, Zhang Q, Sklar P, McCarthy MI, (2013) Study of 11 BMI-associated loci identified in
Brown MA et al (2017) 10 years of GWAS discovery: GWAS for associations with central obesity in the
biology, function, and translation. Am J Hum Genet Chinese children. PLoS One 8(2):e56472
[Internet] 101(1):5–22. Available from: http://www. Xu B, Xie X (2016) Neurotrophic factor control of satiety
sciencedirect.com/science/article/pii/ and body weight. Nat Rev Neurosci 17(5):282–292
S0002929717302409 [cited 2019 Oct 28] Yengo L, Sidorenko J, Kemper KE, Zheng Z, Wood AR,
Wang K, Li WD, Zhang CK, Wang Z, Glessner JT, Grant Weedon MN et al (2018) Meta-analysis of genome-
SF et al (2011) A genome-wide association study on wide association studies for height and body mass
obesity and obesity-related traits. PloS One [Internet] 6 index in 700000 individuals of European ancestry.
(4):e18939. Available from: https://europepmc.org/ Hum Mol Genet 27(20):3641–3649
articles/PMC3084240/ [cited 2019 Oct 30] Zandoná MR, Sangalli CN, Campagnolo PDB, Vitolo
Wang HJ, Hinney A, Song JY, Scherag A, Meng XR, MR, Almeida S, Mattevi VS (2017) Validation of
Grallert H et al (2016) Association of common variants obesity susceptibility loci identified by genome-wide
identified by recent genome-wide association studies association studies in early childhood in South
with obesity in Chinese children: a case-control study. Brazilian children. Pediatr Obes [Internet] 12
BMC Med Genet [Internet] 17:7. Available from: (1):85–92. Available from: https://onlinelibrary.wiley.
https://europepmc.org/articles/PMC4724138/ [cited com/doi/abs/10.1111/ijpo.12113 [cited 2019 Oct 31]
2019 Oct 31] Zhao J, Bradfield JP, Li M, Wang K, Zhang H, Kim CE
Wen W, Zheng W, Okada Y, Takeuchi F, Tabara Y, et al (2009 Dec) The role of obesity-associated loci
Hwang J-Y et al (2014) Meta-analysis of genome- identified in genome-wide association studies in the
wide association studies in East Asian-ancestry determination of pediatric BMI. Obes Silver Spring
populations identifies four new loci for body mass Md 17(12):2254–2257
The Science of Love: State of the Art
16
Donatella Marazziti, Stefania Palermo, and Federico Mucci

Abstract positive effects of love can be extremely bene-


In these last decades, emotions and feelings, ficial for both mental and physical health.
neglected for centuries by experimental However, available data are still limited,
sciences, have become the topic of extensive and the proposed models, although supported
neuroscientific research. Currently, love, the by converging data, should be considered
most typically human feeling, can be viewed speculative and oversimplified. The hope is
as the result of different phases (steps), each that neuroscience will permit to shed light on
regulated by evolutionary well-conserved and love, one of the most intriguing, and still
integrated neural substrates. We have pro- largely unknown mysteries of human nature.
posed that the early stage, generally called
romantic love, is the result of the activation
of the brain limbic structures regulating fear/
anxiety reactions leading to changes of major
Abbreviations
neurotransmitters, such as increased mono-
amine levels and decreased serotonin
BD bipolar disorder
concentrations. The second stage of love is
BDNF brain-derived neurotrophic factor
mainly underlain by the structures regulating
dACC dorsal anterior cingulate cortex
the attachment system and involving oxytocin
ECR Experiences in Close Relationships
and vasopressin neuropeptides and
fMRI functional magnetic resonance imaging
neurotrophins. This would explain why the
NGF nerve growth factor
NTs neurotrophins
OCD obsessive-compulsive disorder
D. Marazziti (*)
Department of Clinical and Experimental Medicine OT oxytocin
Section of Psychiatry, University of Pisa, Pisa, Italy ReHo regional homogeneity
UniCamillus - Saint Camillus International University of rsfMRI resting state functional magnetic
Health and Medical Sciences, Rome, Italy resonance imaging
e-mail: dmarazzi@psico.med.unipi.it VP vasopressin
S. Palermo 5-HT 5-hydroxytryptamine (serotonin)
Department of Clinical and Experimental Medicine
Section of Psychiatry, University of Pisa, Pisa, Italy
F. Mucci
Department of Biotechnology, Chemistry and Pharmacy,
University of Siena, Siena, Italy

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 249
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_16
250 D. Marazziti et al.

16.1 Introduction Nature follows its rules in all processes, that is


to say, accomplishment of its scopes. Therefore,
Love is an exquisite human attribute that is love is moral from the point of view of the nature,
embedded in human nature. Love springs and but while being regulated by the same and strong
develops because human beings are born with neurobiological systems involved in species sur-
the innate drive toward the reward intrinsic to vival, its intents may collide with human rules. If
loving and to be loved. Love is a process, specifi- love may represent the most extraordinary and
cally an integrated neurobiobehavioral process joyful experience that nature invented for
which promotes proximity between two unrelated humans, on the other side, it may provoke not
individuals, reproduction, and kids’ rearing. only extreme individual suffering, but also family
Moreover, it reduces feelings of stress and anxi- destructions, or disasters like wars, suicides, and
ety while triggering a deep sense of safety, joy, murders.
and reward. Therefore, love is a blend of brain The aim of the present paper is to provide a
mechanisms and related physical phenomena, as careful and updated literature review on the neu-
well as cultural and social factors that may shape robiology of love together with authors’ personal
some features and regulate its expression. If some contributions in this fascinating domain.
components of this complex process can be
also found in mammals, what makes love typical
of humans is the evidence that the formation of 16.1.1 The First Step of Love:
pair bonding in our species is related not only to Attraction
reproduction, but also to the creation of group
structures, social organizations, and interactions. Attraction is the biological term to denote what is
Nature should have provided us mechanisms of commonly called romantic love or falling in love.
increasing complexity for assuring that the focus The evidence that is experienced by every
is limited to one (or a few) partner(s) only, and the individual all over the world strongly indicates
sense of safety is formed and maintained, while that it is possibly underlaid by neurobiological
rewarding the individuals with that feeling of underpinnings (Jankowiak and Fischer 1992).
pleasure and completeness that we call love. The Available data suggest that generally it lasts
sum of emotions + behaviors + subjective aware- between 6 months and 2 years that is considered
ness of the whole processes constitutes, perhaps, the necessary time required to mate, to reach a
the essence of love (Marazziti 2009, 2015; De pregnancy, and to take care of the newborns
Boer et al. 2012). before they become mature to survive alone.
The consistency of love across different Attraction is independent from bonding, having
individuals suggests that it is regulated by shared sex, or being reciprocated.
biological mechanisms that only recently have Our hypothesis is that attraction is a basic
been investigated. Modern brain imaging and emotion that shares some neurobiological
neurobiological techniques have revealed that substrates with fear and anxiety (LeDoux 2000;
people in romantic relationships, the first step of Marazziti and Cassano 2003; Marazziti 2015). As
love, bear a striking similarity to those with psy- such, nobody can decide by his/her will to be
chiatric disorders, which is not surprising, just attracted by somebody; rather, it is driven by
considering that love involves the same systems casual and strong sensorial stimuli occurring
that regulate anxiety and fear responses, and more or less on the same time in two unrelated
related neurotransmitters (Marazziti et al. 1999; individuals. Indeed, following internal (such as
Bartels and Zeki 2000; Fisher 1992; Fisher et al. hormonal variations) and external (such as life
2016). The next step, that is, attachment, involves events) changes, our brain, in particular amyg-
the nonapeptide oxytocin and dopamine areas dala, a group of subcortical nuclei regulating
regulating the reward process. fight/flight responses fears and emotions,
becomes particularly predisposed to “normal”
16 The Science of Love: State of the Art 251

Fig. 16.1 The circuitry of


romantic love. (a) Shorter
pathway, (b) longer
pathway

stimuli coming from another person. These behaviors to gain his/her attention, while
stimuli are first processed by the thalamus, and suggesting a common alteration with obsessive-
then sent to amygdala through two pathways: a compulsive disorder (OCD) patients (Marazziti
direct and a longer one involving an intermediate et al. 1999).
step represented by the cortex (Fig. 16.1). The The temporary state of “madness” experienced
amygdala modulates the incoming stimuli while by people in the phase of romantic love may have
detecting its emotional quality (good or bad?) and an evolutionary purpose, as it pushes the individ-
organizes all outputs, such as neurovegetative ual to become more impulsive, to overcome fear
reactions and hormonal responses, in order to and neophobia, and to willingly accept to be close
prepare the individual to fight or flight. After to a stranger that becomes the most extraordinary
6 milliseconds, the activated cortex, through the one all over the world. That is to say, it allows the
longer pathway, provides the subjective aware- individual to get out of their comfort zone to
ness of the bodily changes that everybody can experience an emotional storm.
relate to being in love, in spite of the evidence Romantic love is a stressful event, as shown by
that, in terms of brain circuits, they are the same the hormonal changes it entails (Marazziti and
of fear or anxiety. Love can be considered, there- Canale 2004), and, as such it has the function of
fore, “a fear without fear” (Marazziti 2015). outgoing individual boundaries, creating new
Altered mental state and intrusive thoughts bonds, and evolving. However, there is a cost to
about the partner are the specific features of pay due to the significant changes of
attraction. Altered mental state resembles neurotransmitters and related brain structures: a
alternating phases of the bipolar disorder (BD): greater risk of developing a full-blown psychiatric
people can experience a raised energy, changes in disorder.
both the appetite and in the need of sleep, lower- In any case, human beings cannot escape from
ing of rational judgment, or mood swings falling in love: it is embedded in our nature that if
according to the reciprocation or not by the part- we love and will be loved, we will be rewarded by
ner. Such characteristics would suggest increased a deep joy and by increasing chance of survival.
functioning of the norepinephrine and dopamine Not surprisingly, some authors reported higher
systems, as well as increased opioid peptides and levels of two neurotrophins (NTs), specifically
a decrease in serotonin (5-HT). The lower func- nerve growth factor (NGF) and brain-derived
tioning of the 5-HT system may explain intrusive neurotrophic factor (BDNF) in plasma of roman-
thoughts regarding the partner and repetitive tic lovers (Emanuele et al. 2006).
252 D. Marazziti et al.

Unfortunately, all described hormonal and while showing activation of brain area rich in
neurobiological do not last forever, and the OT receptors when individuals look at pictures
“altered state of mind” returns to a more stable of the partners (Bartels and Zeki 2000; Fisher
and calm condition. In parallel, if the partner does et al. 2002). Subsequent authors confirmed these
not reciprocate or the emotions do not last, the findings while highlighting how RR is a strong
relation comes to an end; otherwise, the couple is motivational state that follows the neuronal cir-
ready to move toward experiencing the next cuitry of reward and pleasure (Aron et al. 2005),
step of love that is romantic attachment. and how long-term love activates the attachment
system (Acevedo et al. 2012).
Besides OT and VP, also NTs seem to play a
16.1.2 Romantic Attachment role in this phase, as shown in a study
investigating the possible relationship between
Romantic attachment is the social process BDNF plasma levels and romantic attachment in
keeping together two individuals for a long 24 healthy adults (12 men, 12 women), as
period, sometimes forever. It is believed generally assessed by the “Experiences in Close
that it is triggered and maintained by the neuro- Relationships” (ECR) questionnaire (Brennan
peptide oxytocin (OT) that seems to be involved et al. 1998). The results highlighted the presence
in the initiation and maintenance of pair bonding, of a strong negative correlation between BDNF
infant attachment, and maternal behavior, while levels and the ECR avoidance scale, while
vasopressin (VP) would show opposite activities suggesting a role of BDNF in promoting social
(Carter 1998; Insel and Young 2001). Both OT bonding through the decrease of fear and avoid-
and VP are produced in the hypothalamus and ance behaviors. On the contrary, this correlation
released by the posterior pituitary (Acher 1996). was not present among men. The authors
Oxytocin would be produced in the hypothalamus hypothesized that perhaps the role of BDNF is
when the attraction phase starts, and it is sup- gender-related and influenced by the interactions
posed to “transform” the “anxiety/fear” reactions between hormones and genes (Marazziti et al.
in a sense of well-being, reward, and joy (Kosfeld 2009). Interestingly, estrogens are supposed to
et al. 2005). This is possible through the reduction induce BDNF synthesis in different brain regions
of the stress systems, and the activation of the (Sasahara et al. 2007). Therefore, BDNF may be
reward processing regulated by the neurotrans- more active in women, which are basically more
mitter dopamine (Heinrichs et al. 2003). At the anxious than men (Kessler et al. 2001; Afifi
same time, OT is released in the bloodstream and 2007), so that NTs would protect neurons from
conveys information to peripheral organs, where stress-induced damage (Bergström et al. 2008).
it is also produced in some of them, and returns to According to some authors (Insel 2003; Fisher
the brain, closing a loop linking brain et al. 2016), love resembles an addiction, on the
mechanisms to periphery. According to the basis of a shared activation of dopamine-rich
polyvagal theory, the activity of OT on the regions involved in the reward system and drug
nucleus vagus, especially on the ventral complex, or behavioral addictions, and of overlapping
the so-called smart vagus would permit to provide symptoms, such as a sense of euphoria, craving,
an explanation of the different emotions and and physical and emotional dependence. Love
bodily changes occurring during a social bonding can be a positive addiction or a negative one
such as love (Porges and Furman 2011). One of when not reciprocated or when it becomes toxic,
our studies confirmed the relationship between for example, leading to depression, stalking, sui-
romantic attachment and OT in 45 healthy cide, homicide, and other crimes (Fisher et al.
subjects (Marazziti et al. 2006). In addition, the 2016).
first double-blind studies conducted with func- Song et al., using resting state functional mag-
tional magnetic resonance imaging (fMRI) con- netic resonance imaging (rsfMRI), aimed at
firmed the role of OT in romantic attachment investigating how romantic love may affect the
16 The Science of Love: State of the Art 253

Fig. 16.2 The


dopaminergic circuitry of
the social brain (VTA
ventral tegmental area,
NAcc nucleus accumbens)

brain and its functional architecture during rest. The scope of the science is not to destroy the
Participants consisted of 34 people (the “in-love poetry of love, as somebody claimed; on the
group”) intensely in love for 4–18 months, contrary, our opinion is that a deeper understand-
34 people who ended their romantic relationship ing of the neurobiology of love would permit to
recently (2–17 months earlier), and 32 people get access to our unique humanity, to love in a
who had never fallen in love. The “in-love more rewarding and joyful way, and perhaps to
group,” in comparison with the control group, prevent the suffering and dramatic consequences
showed an increased ReHo (regional homogene- of unrequited love or separations.
ity) on the left dorsal anterior cingulate cortex
(dACC), and an increased functional connectivity
within both the reward network (caudate, nucleus
References
accumbens, etc.) and the social cognition network
(temporoparietal junction, posterior cingulate cor- Acevedo BP, Aron A, Fisher HE, Brown LL (2012) Neu-
tex, etc.) (Fig. 16.2). All the mentioned increases ral correlates of long-term intense romantic love. Soc
were positively correlated with the duration of the Cogn Affect Neurosci 7(2):145–159. https://doi.org/
“being in love” state (Song et al. 2015). 10.1093/scan/nsq092
Acher R (1996) Molecular evolution of fish neurohypo-
physial hormones: neutral and selective evolutionary
mechanisms. Gen Comp Endocrinol 102(2):157–172.
https://doi.org/10.1006/gcen.1996.0057
16.2 Conclusions Afifi M (2007) Gender differences in mental health.
Singap Med J 48(5):385–391
The present paper is a review on some of the Aron A, Fisher HE, Mashek DJ, Strong G, Li H, Brown
LL (2005) Reward, motivation, and emotion systems
major findings on the neurobiology of love. This associated with early-stage intense romantic love. J
is an emerging and intriguing field of research Neurophysiol 94:327–337. https://doi.org/10.1152/jn.
that only recently has become the topic of inten- 00838.2004
sive scientific investigations and has received Bartels A, Zeki S (2000) The neural basis of romantic love.
Neuroreport 11(17):3829–3834. https://doi.org/10.
benefits from the application of the most
1097/00001756-200011270-00046
advanced methods provided by neuroscience. Bergström A, Jayatissa MN, Mørk A, Wiborg O (2008)
The scattered data that have been gathered and Stress sensitivity and resilience in the chronic mild
are accumulating at an increasing amount, albeit stress rat model of depression; an in situ hybridization
study. Brain Res 1196:41–52. https://doi.org/10.1016/
preliminary and fragmentary, permit to set up an
j.brainres.2007.12.025
initial framework and hypotheses that need to be Brennan KA, Clark CL, Shaver PR (1998) Self-report
tested. measurement of adult attachment: an integrative over-
view. In: Simpson JA, Rholes WS (eds) Attachment
254 D. Marazziti et al.

theory and close relationships. New York, Guilford Marazziti D (2009) Neurobiology and hormonal aspects of
Press, pp 46–76 romantic attachment. In: De Haan M, Gunnar MN
De Boer A, van Buel EM, Ter Horst GJ (2012) Love is (eds) Handbook of developmental and social neurosci-
more than just a kiss: a neurobiological perspective on ence. New York, Guilford Press, pp 265–280
love and affection. Neuroscience 201:114–124. https:// Marazziti D (2015) “Beyond emotion: love as an encoun-
doi.org/10.1016/j.neuroscience.2011.11.017 ter of myth and drive” by Lubomir Lamy. Emotion Rev
Carter CS (1998) Neuroendocrine perspectives on social 8(2):110–112. https://doi.org/10.1177/
attachment and love. Psychoneuroendocrinology 23 2F1754073915594437
(8):779–818. https://doi.org/10.1016/s0306-4530(98) Marazziti D, Canale D (2004) Hormonal changes when
00055-9 falling in love. Psychoneuroendocrinology 29
Emanuele E, Politi P, Bianchi M, Minoretti P, Bertona M, (7):931–936. https://doi.org/10.1016/j.psyneuen.2003.
Geroldi D (2006) Raised plasma nerve growth factor 08.006
levels associated with early-stage romantic love. Marazziti D, Cassano GB (2003) The neurobiology of
Psychoneuroendocrinology 31(3):288–294. https:// attraction. J Endocrinol Investig 26:58–61
doi.org/10.1016/j.psyneuen.2005.09.002 Marazziti D, Akiskal HS, Rossi A, Cassano GB (1999)
Fisher E (1992) Anatomy of love. Fawcett Columbine, Alteration of the platelet serotonin transporter in
New York romantic love. Psychol Med 29(3):741–745. https://
Fisher HE, Aron A, Mashek D, Li H, Brown LL (2002) doi.org/10.1017/s0033291798007946
Defining the brain systems of lust, romantic attraction, Marazziti D, Dell’Osso B, Baroni S, Mungai F, Catena M,
and attachment. Arch Sex Behav 31(5):413–419. Rucci P, Albanese F, Giannaccini G, Betti L,
https://doi.org/10.1023/a:1019888024255 Fabbrini L, Italiani P, Del Debbio A, Lucacchini A,
Fisher HE, Xu X, Aron A, Brown LL (2016) Intense, Dell’Osso L (2006) A relationship between oxytocin
passionate, romantic love: a natural addiction? How and anxiety of romantic attachment. Clin Pract
the fields that investigate romance and substance abuse Epidemiol Ment Health: CP & EMH 2:28. https://doi.
can inform each other. Front Psychol 7:687. https://doi. org/10.1186/1745-0179-2-28
org/10.3389/2Ffpsyg.2016.00687 Marazziti D, Roncaglia I, Del Debbio A, Bianchi C,
Heinrichs M, Baumgartner T, Kirschbaum C, Ehlert U Massimetti G, Origlia N, Domenici L, Piccinni A,
(2003) Social support and oxytocin interact to suppress Dell’Osso L (2009) Brain-derived neurotrophic factor
cortisol and subjective responses to psychosocial in romantic attachment. Psychol Med 39
stress. Biol Psychiatry 54(12):1389–1398. https://doi. (11):1927–1930. https://doi.org/10.1017/
org/10.1016/s0006-3223(03)00465-7 S0033291709990742
Insel TR (2003) Is social attachment an addictive disorder? Porges SW, Furman SA (2011) The early development of
Physiol Behav 79:351–357. https://doi.org/10.1016/ the autonomic nervous system provides a neural plat-
s0031-9384(03)00148-3 form for social behavior: a polyvagal perspective.
Insel TR, Young LJ (2001) The neurobiology of attach- Infant Child Dev 20(1):106–118. https://doi.org/10.
ment. Nat Rev Neurosci 2:129–136. https://doi.org/10. 1002/icd.688
1038/35053579 Sasahara K, Shikimi H, Haraguchi S, Sakamoto H,
Jankowiak WR, Fischer EF (1992) A cross-cultural per- Honda S, Harada N, Tsutsui K (2007) Mode of action
spective on romantic love. Ethnology 31(2):149–155. and functional significance of estrogen-inducing den-
https://doi.org/10.2307/3773618 dritic growth, spinogenesis, and synaptogenesis in the
Kessler RC, Keller MB, Wittchen HU (2001) The epide- developing Purkinje cell. J Neurosci 27:7408–7417.
miology of generalized anxiety disorder. Psychiatr Clin https://doi.org/10.1523/JNEUROSCI.0710-07.2007
North Am 24:19–39. https://doi.org/10.1016/s0193- Song H, Zou Z, Kou J, Liu Y, Yang L, Zilverstand A,
953x(05)70204-5 d’Oleire Uquillas F, Zhang X (2015) Love-related
Kosfeld M, Heinrichs M, Zak PJ, Fischbacher U, Fehr E changes in the brain: a resting-state functional mag-
(2005) Oxytocin increases trust in humans. Nature netic resonance imaging study. Front Hum Neurosci
435:673–676. https://doi.org/10.1038/nature03701 9:71. https://doi.org/10.3389/fnhum.2015.00071
LeDoux JE (2000) Emotion circuits in the brain. Annu Rev
Neurosci 23:155–184. https://doi.org/10.1146/
annurev.neuro.23.1.155
NGF and Retinitis Pigmentosa: Structural
and Molecular Studies 17
Maria Luisa Rocco, Laura Calzà, and Luigi Aloe

Abstract reduces/stops the progression of photoreceptor


Nerve growth factor (NGF) is a neuroprotective degeneration of rats with RP.
molecule performing not only on central and This study was carried out in vitro on
peripheral neurons but also on cells of the isolated photoreceptors to further investigate
visual system. Human retinitis pigmentosa the action on these cells and whether the action
(RP) is a major cause of blindness worldwide, is direct or mediated.
and a resolute therapy is still lacking. Recent The results indicate that ocular NGF admin-
studies have shown that ocular NGF adminis- istration can protect photoreceptors from
tration exerts a protective action on damaged degeneration into a model developing
retinal cells of mammalians, including human inherited RP and that the NGF action is direct.
beings, although whether NGF also protects In this regard, we observed that binding of
photoreceptors is not clear. NGF to its receptor modulates expression of
We used the Royal College of Surgeons rhodopsin, a specific biological marker for
(RCS) strain in this study. The RCS is a rodent photoreceptor survival and functionality.
affected by inherited retinitis pigmentosa Part of the data reported in this chapter has
(RP) during postnatal life. For this study, we been published in a previous study.
investigated whether ocular NGF treatment
Abbreviations
M. L. Rocco (*)
IRET Foundation, Ozzano Emilia, Bologna, Italy NGF Nerve Growth Factor
Institute of Translational Pharmacology, CNR, Rome, RCS Royal College of Surgeons
Italy RP Retinitis Pigmentosa
L. Calzà RPE Retinal Pigment Epithelium
Health Science and Technologies Interdepartmental TrkA Tyrosine Kinase Receptor
Center for Industrial Research (HST-ICIR), University of
Bologna, Ozzano Emilia, Bologna, Italy
Department of Pharmacy and Biotechnology, University 17.1 Introduction
of Bologna, Bologna, Italy
Montecatone Rehabilitation Institute, Montecatone, We used the Royal College of Surgeons (RCS)
Bologna, Italy
e-mail: laura.calza@unibo.it strain in this study. The RCS is a rodent affected
by inherited retinitis pigmentosa (RP) during
L. Aloe
IRET Foundation, Ozzano Emilia, Bologna, Italy postnatal life (D’Cruz et al. 2000). The primary

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 255
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_17
256 M. L. Rocco et al.

pathological event in many forms of retinal 17.2 Methods


degeneration is the degeneration of photoreceptor
cells, while one of the most common retinal NGF Isolation and Purification For these stud-
degenerations is retinitis pigmentosa (RP). RP is ies, the molecule 2.5S NGF was purified in our
a group of inherited human diseases characterized laboratory from adult CD1 mouse submaxillary
by progressive degeneration of photoreceptor gland (75gr). The biological activity of NGF is
cells and loss of photoreceptor function (Dowling tested on PC12 cells, plated on 24-well plates at a
and Sidman 1962; Faktorovich et al. 1990). A density of 10,000 cells/wells in Dulbecco’s
number of recent studies report that the differen- Modified Eagle Medium (DMEM), containing
tiation, survival, and function of retinal cells are 10% heat-inactivated horse serum and 5% bovine
regulated by survival growth factors, including serum. Cells plated on glass dishes, which had
neurotrophins (Amendola and Aloe 2002; previously coated with polylysine, were cultured
Amendola et al. 2003; Pagani et al. 2006). NGF in the presence of and with different
is a potent stimulant of the tropism and wound concentrations of NGF for 2–4 days.
healing process in the eye, and basically, all Figure 17.1a–b illustrates representative
structures of the eye can produce NGF and even photomicrographs showing PC12 cells cultured
upregulate NGF synthesis in the event of a lesion. in vitro for four consecutive days in the absence
In recent years, Aloe et al. have demonstrated that (Fig. 17.1a) and in the presence of 10 ng/ml of
NGF topical administration plays an important NGF (Fig. 17.1b). The presence of NGF
role in the survival of photoreceptor cells, as stimulates neuritis outgrowth. For further details
well as that NGF exercises a significant effect see Bocchini and Angeletti (1969).
on expression of its receptors, blood vessel distri-
bution, and the degeneration of the outer nuclear NGF and ANA Biological Activity
layer (ONL) (Aloe 2004; Aloe et al. 2008, 2012). on PC12 Before using the NGF on isolating
In this study, we investigated whether ocular photoreceptors, we first tested the biological
NGF treatment reduces/stops the progression of activity on PC12 cell line routinely used for test-
photoreceptor degeneration of rats with RP ing the biological activity of NGF. In the absence
(Rocco et al. 2015). During the early stages of of NGF, the cells are unable to produce neuritis
photoreceptor degeneration, rats were treated outgrowth, while in the presence of 50 ng/ml
with NGF eye drops (200 ug/ml) three times a NGF, they respond with neuritis outgrowth, and
day for 10 consecutive days. The controls were in the presence or 50 ng/ml NGF, preincubated
treated with doses of vehicle solution for the same with 500 ng/ml anti-NGF antibody (ANA) that
length of time (Rocco et al. 2015). At the end of inhibit the NGF biological activity, these cells are
the treatment, the eyes were surgically removed unable to produce neuritis. Together these
and used for histological, immunohistochemical, observations indicate that both NGF and ANA
biochemical, and molecular studies (Rocco et al. are biologically and functionally active (for the
2015). Compared to controls, it emerged that results see Rocco et al. (2015)).
NGF-treated retinas showed altered expression
of apoptotic markers, enhanced presence of Western Blotting Analysis For Western blot-
NGF receptors, and reduced loss of ting analysis, tissues were homogenized with
photoreceptors (Rocco et al. 2015). Our study ultrasonication in RIPA buffer (50 mM Tris,
was carried out in vitro on isolated photoreceptors pH 7.5; 150 mM NaCl; 5 mM EDTA; 1% Triton
to further investigate the action on these cells and X-100; 0.1% SDP; 0.5% DOC (sodium
whether the latter is direct or mediated (Rocco deoxycholate); 1 mM PMSF; 1 μg/ml leupeptin),
et al. 2015). centrifuged at 4  C for 20 min at 13000 rpm; then
the supernatant was stored at 20  C. Samples
(30 μg of total protein) were dissolved in loading
17 NGF and Retinitis Pigmentosa: Structural and Molecular Studies 257

Fig. 17.1 PC12 cells in the absence (untreated) or pres- rhodopsin (c red channel), NGF-treated RCS retinas. Rho-
ence of NGF. Untreated cells are unable to produce neuri- dopsin and DAPI immunoreactivity appeared in photore-
tis outgrowth, (a) while in the presence of 50 ng/ml NGF, ceptor cells was clearly increased and the outgrowth of
they respond with neuritis outgrowth (b red arrow). Scale neurites after NGF treatment (c yellow arrow). Scale bar:
bar: 15 μm. Phase-contrast images, 40/objective. Confo- 15 μm. 60/objective; zoom 5; 1024  1024 pixels; 10 Hz
cal microscopy images of DAPI (c green channel), acquisition speed

buffer (0.1 M Tris–HCl buffer, pH 6.8, containing Biotechnology, CA, USA) bands was used as a
0.2 M dithiothreitol (DTT), 4% Sodium dodecyl normalizing factor. For each gel blot, the
sulfate (SDS), 20% glycerol, and 0.1% normalized values were then expressed as per-
bromophenol blue), separated by 8% or 12% centage of relative normalized controls and used
SDP-PAGE, and electrophoretically transferred for statistical evaluation. Statistical evaluations
to PVDF membrane overnight. The membranes were performed using the GraphPad Prism pack-
were incubated for 1 h at room temperature with age for Windows and data expressed as means 
blocking buffer constituted by 5% nonfat dry SEM of eight different corneas (for the results see
milk in TBS-T (10 mM Tris, pH 7.5, 100 mM Rocco et al. (2015)).
NaCl, and 0.1% Tween-20). Membranes were
washed three times for 10 min each at room Relative Quantitative Real-Time PCR For
temperature in TBS-T followed by incubation at RT-PCR analysis of TrkA and p75NTR, total
4  C with primary antibodies overnight with the RNA from the cornea was extracted using SV
antibodies shown in the table. Membranes were Total Isolation System (Promega Italia, Milan,
washed three times for 10 min each at room Italy). RNA was quantified by spectrophotometer
temperature in TBS-T and incubated for 1 h at 260 nm and stored at 80  C before use. RNA
with horseradish peroxidase-conjugated was converted into cDNA in a 20 μl reverse
anti-rabbit IgG 1:4000 or horseradish transcription reaction containing 2γ of total
peroxidase-conjugated anti-mouse IgG as the sec- RNA; 1X reverse transcriptase buffer (50 mM
ondary antibody (Cell Signaling Technology, Tris–HCl, pH 8.3; 75 mM KCl; 3 mM MgCl2;
MA, USA) at room temperature. The blots were 10 mM DTT); 0.5 mM dNTPs; 200 ng oligo
developed with an ECL chemiluminescent horse- (dT) 15; 0.5 U RNasin ribonuclease inhibitor;
radish peroxidase (HRP) substrate as the chromo- and 200 U of M-MLV RT (Promega Italia,
phore (Millipore, MA, USA). The public ImageJ Milan, Italy). Reactions were incubated at 42  C
software was used to evaluate band density, for 60 min, heated at 95  C for 5 min, and then
which was expressed as arbitrary units of gray cooled at 4  C for a minimum of 5 min and a
level. The ImageJ program determines the optical maximum of 30 min. The polymerase chain reac-
density of the bands using a grayscale sharehold- tion (PCR) was analyzed using the 7900HT Fast
ing operation. The optical density of polyclonal Real-Time PCR System (PE Applied Biosystems)
rabbit anti-GAPDH 1:4000 (Santa Cruz and FAM-labeled probe specific for the NTrk1
258 M. L. Rocco et al.

(Rn00572130_m1) and Ngfr (Rn00561634_m1) microscope equipped with a 40X objective


(PE Applied Biosystems). The housekeeping (Rocco et al. 2015).
gene, glyceraldehyde-3-phosphate dehydroge-
nase (GAPDH) (4352338E) (PE Applied Immunofluorescence For immunofluorescence
Biosystems), was co-amplified with TrkA and eye globes of each experimental group were
p75NTR and used to normalize sample removed and fixed in 4% paraformaldehyde
variability. The cDNA was amplified under the dissolved in 0.1 M phosphate buffer, pH 7.4, for
following conditions: 1 cycle at 50  C for 2 min 24 h and then left overnight in the same buffer
and at 95  C for 10 min, followed by 40 cycles at containing 20% of sucrose. Coded sections of
95  C for 15 s and at 60  C for 1 min. The amount each eye were then sectioned with a cryostat, at
of mRNA of each gene was calculated using the 20 μm thickness. Sections were first washed with
standard curve method and adjusted for the 1% of PBS two times for 5 min and then
expression of GAPDH. Relative quantification incubated overnight at 4  C with polyclonal rabbit
was performed using the ΔΔCt method (for the anti-TrkA 1:100 (Santa Cruz, CA, USA) and
results see Rocco et al. (2015)). monoclonal mouse anti-rhodopsin 1:100
(Neomarkers, CA, USA). After washing with
Immunohistochemistry For immunohisto- PBST, the sections were incubated for 1 h at
chemistry eye globes of each experimental room temperature with Alexa Fluor 488 donkey
group were removed and fixed in 4% paraformal- anti-rabbit IgG (1: 200) and Alexa Fluor 555 don-
dehyde dissolved in 0.1 M phosphate buffer, key anti-mouse IgG (1: 200). After two rinses in
pH 7.4, for 24 h and then left overnight in the PBS and 10-min incubation with the DAPI solu-
same buffer containing 20% sucrose. Coded tion (Molecular Probes, Invitrogen, Italy) for
sections of each eye were then sectioned with a nuclei visualization, sections were coverslipped
cryostat, at 20 μm thickness. Sections were first and examined under a confocal laser scanning
exposed to 0.03% of hydrogen peroxide (H2O2) microscope (Leica SP5, Leica Microsystems,
and 10% of methanol for 20 min, followed by Germany) under a sequential mode to avoid
exposure to 0.1 M PBS containing 10% of horse cross talk between channels. The confocal image
or goat serum for 1 h, and then incubated over- acquisition was conducted so that all samples
night at 4  C with polyclonal rabbit anti-TrkA were imaged using consistent settings for laser
1:100 (Santa Cruz, CA, USA) and monoclonal power and detector gain (for the results see
mouse anti-p75 1:100 (Santa Cruz, CA, USA). Rocco et al. (2015)).
Sections, after having been washed, were then
exposed to biotinylated anti-mouse or anti-rabbit Cell Culture We first tested the biological activ-
IgG 1:300 (anti-IgG and avidin-conjugated horse- ity on PC12 cells as reported under Material and
radish peroxidase complex) which were pur- Method Section. As shown in Fig. 17.1a–b, it was
chased by Vector Laboratories (Burlingame, found that the presence of 10 ng of NGF/ml of
CA, USA) with 2% of goat or horse serum, culture medium was sufficient to stimulate neurite
depending on the animal in which the secondary outgrowth from these cells, whereas sister cells
antibody was produced, for 2 h at room tempera- cultured in the absence of NGF were unable to do
ture, and immunoperoxidase staining was it. Based on these observations, we tested the
performed using an ABC (1:100) (Avidin-Biotin effect of the purified NGF on photoreceptor
Complex Solution, Vectastain Elite Kit (Vector response and survival. For cell culture eye globes
Laboratories, CA, USA) for 2 h at room tempera- of each experimental group were removed, and
ture. Signals were visualized by DAB (3,3- the retinal was isolated. The cells were washed
0
-diaminobenzidine) as a chromate. Stained with 6 ml of 10 μg/ml in 0.1 M phosphate buffer
sections were visualized using a Zeiss Axiophot and, after that, were centrifuged for 1 min at
1000 rpm. After digestion in a 0.25% trypsin for
17 NGF and Retinitis Pigmentosa: Structural and Molecular Studies 259

30 min at 37  C, the trypsin action was stopped Sodium dodecyl sulfate (SDS), 20% glycerol,
with 10% of serum retinal, after was washed with and 0.1% bromophenol blue), separated by 8%
10 μg/ml in 0.1 M phosphate buffer, pelleted and or 12% SDP-PAGE, and electrophoretically
resuspended for 15 min in culture medium transferred to PVDF membrane overnight. The
(DMEM/F12, 1:1 Invitrogen), and supplemented membranes were incubated for 1 hr at room tem-
with 5% FBS and 1% penicillin/streptomycin. perature with blocking buffer constituted by 5%
The supernatant was released and the pellet was nonfat dry milk in TBS-T (10 mM Tris, pH 7.5,
resuspended with 1 ml of culture medium. 100 mM NaCl, and 0.1% Tween-20). Membranes
Finally, the two supernatants were centrifuged were washed three times for 10 min each at room
together for 10 min at 1000 rpm, and 6 ml of temperature in TBS-T followed by incubation at
culture medium was added with 6 ml of culture 4  C with Bax (B8429, Sigma, Missouri, USA)
medium. The cells (2.5  106 for plate) remained and Bcl-2 (sc-7382, Santa Cruz Biotechnology,
undisturbed at 37  C in a 5% CO2 atmosphere for Inc., Europe) antibodies overnight with the
48h before the treatments (for the results see antibodies shown in the table. Membranes were
Rocco et al. (2015)). washed three times for 10 min each at room
temperature in TBS-T and incubated for 1 h
Photoreceptor Sorting To verify the effect of with horseradish peroxidase-conjugated
NGF on the photoreceptor, the cells were selected anti-rabbit IgG 1:4000 or horseradish
and separated through from whole retinal cultures peroxidase-conjugated anti-mouse IgG as the sec-
by EasySep Magnet separation with the use of ondary antibody (Cell Signaling Technology,
tetrameric antibody complexes recognizing MA, USA) at room temperature. The blots were
rhodopsin-bearing photoreceptors, in the absence developed with an ECL chemiluminescent horse-
of columns (EasySep Magnet separation; radish peroxidase (HRP) substrate as the chromo-
Stemcell Technologies, Voden Medical phore (Millipore, MA, USA). The public ImageJ
Instruments SpA, Milan, Italy). Enriched software was used to evaluate band density,
photoreceptors (~80–90%) were cultured, treated, which was expressed as arbitrary units of gray
and analyzed as previously reported. Purity was level. The ImageJ program determines the optical
assessed by a digital-based flow cytometry plat- density of the bands using a grayscale sharehold-
form (MACSQuant Analyzer, Miltenyi, GmbH, ing operation. The optical density of polyclonal
Germany). Both untreated and NGF-treated cells rabbit anti-GAPDH 1:4000 (Santa Cruz Biotech-
(6 days) were postfixed in buffered 4% PFA and nology, CA, USA) bands was used as a
analyzed for trkANGFR and p75NTR expression normalizing factor. For each gel blot, the
(for the results see Rocco et al. (2015)). normalized values were then expressed as per-
centage of relative normalized controls and used
Cell Death Studies The cells (2.5  106 for for statistical evaluation. Statistical evaluations
plate) remained undisturbed at 37  C in a 5% were performed using the GraphPad Prism pack-
CO2 atmosphere for 48h before the treatments. age for Windows and data expressed as means 
After the treatment the cells were homogenized SEM of eight different corneas (for the results see
with ultrasonication in RIPA buffer (50 mM Tris, Rocco et al. (2015)).
pH 7.5; 150 mM NaCl; 5 mM EDTA; 1% Triton
X-100; 0.1% SDP; 0.5% DOC (sodium
deoxycholate); 1 mM PMSF; 1 μg/ml leupeptin),
17.3 Results
centrifuged at 4  C for 20 min at 13000 rpm;
then the supernatant was stored at 20  C.
This chapter presents only two previously unpub-
Samples (20 μg of total protein) were dissolved
lished figures, which summarize the essence of a
in loading buffer (0.1 M Tris–HCl buffer, pH 6.8,
previously published work (Rocco et al. 2015).
containing 0.2 M dithiothreitol (DTT), 4%
260 M. L. Rocco et al.

Animals were housed in animal facilities and through activation of NGF signaling on cell
treated according to the experimental procedure survival. This is further supported by increased
approved by the Ethical Commission on Animal neuritis outgrowth and rhodopsin expression after
Experimentation of the National Research Coun- treatment (Fig. 17.1c) (Rocco et al. 2015).
cil of our Institution (CNR, Rome). All efforts
were made to reduce the number of animals
used for experimentation, and to minimize animal 17.3.1 The Effect of NGF Exposure
suffering. For the in vivo study, rats were grouped on Survival of p10 Retinal Cells
as untreated (n ¼ 30; RCS) and NGF treated
(n ¼ 30; NGF-RCS). RCS rats were treated with We thereafter tested the effect of NGF on primary
NGF eye drops (200 μg/mL; 5 μL) for 3 times a cultures obtained from P10 retinas dissected out,
day over 21 consecutive days, starting at day dissociated, and plated for 12 h or 6d depending
20 of postnatal age. The NGF administration on the experiment. As a result, the cells quickly
enhanced both NGF mRNA and protein in the adhered to the plate (A, time 0 or 12 h), and some
retina. Similarly, both trkANGFR and p75NTR of them resulted in rhodopsin positive (Fig. 17.1c
levels significantly increased, as quantified by red staining). These rhodopsin-positive cells
Western blot analysis and gene expression. The showed a few elongations. Culturing was
phosphorylated trkANGFR form (p-trkANGFR) performed over 6d in medium alone, with the
was overexpressed compared to total trkANGFR result that a reduced number of rhodopsin-
indicating the activation of a specific receptor positive cells were detected compared to the ini-
(Rocco et al. 2015). To assess whether NGF acts tial plating. Upon 6d NGF exposure (Fig. 17.1c),
on the photoreceptor function, retinal sections of the rhodopsin-positive cell number was compara-
NGF-treated and untreated eyes were ble to that of plating, and interestingly,
immunostained with anti-rhodopsin, a specific elongations occurred in most of the rhodopsin-
marker of these cells and of both NGF receptors. positive cells. The effect is clearly visible when
The confocal microscopy revealed increased comparing NGF treated with untreated fields.
trkANGFR immunoreactivity in the retina of These observations indicated that NGF stimulates
NGF-treated RCS rats against untreated ones the survival of photoreceptors (for the results see
(Fig. 17.2a, b, c and d). As shown in Fig. 17.2a, Rocco et al. (2015)).
b, c, and d, the ONL thickness of rats treated with
ocular NGF (c–d) is higher and markedly stained
with rhodopsin (b–d), compared to the lower 17.3.2 The Effect of NGF
thickness and poor immunostaining in the ONL on Rhodopsin-Positive Neuritis
of control rats (a–b). A significant enhancement Outgrowth and trkANGFR
of both receptors was localized in the ON layer of Expression
NGF-treated RCS retinas, alongside
photoreceptors and in proximity of rhodopsin To validate the specificity of the NGF effect on
immunoreactivity (for the results see Rocco these retinal cells, primary cultures were seeded/
et al. (2015)). Overall, these observations cultured over 6d in medium alone or
revealed a critical protective link between the supplemented with NGF, neutralizing anti-NGF
NGF effect and photoreceptor survival. To ana- antibodies (ANA) or NGF preincubated with
lyze the effect of the NGF administration on cell ANA (NGF/ANA), highlighting the specificity
survival, expression of the apoptotic markers Bax of the addition of exogenous NGF on survival
and Bcl-2 – pro- and anti-apoptotic markers, and neuritis outgrowth of dissociated P10 retinal
respectively – was measured (for the results see cells as compared to controls. This NGF effect
Rocco et al. (2015)). All data produced in vivo was detectable neither in the presence of
showed a positive effect of NGF treatment on the neutralizing anti-NGF antibodies nor when the
retinal tissue and, above all, on the ON layer, cells were exposed to NGF preincubated with
17 NGF and Retinitis Pigmentosa: Structural and Molecular Studies 261

Fig. 17.2 Confocal


microscopy images of
DAPI (upper panel a–c) and
TrkANGFR (lower panel
b–d), in untreated (a–b) or
NGF-treated RCS retinas
(c–d). TrkANGFR (b–d)
and DAPI (a–c)
immunoreactivity appeared
mainly in the outer nuclear
layer (ONL) and in
surrounding cells was
clearly increased and the
thickness of the layer after
NGF treatment (c–d). Scale
bar: 15 μm. 60/objective;
1024  1024 pixels; 10 Hz
acquisition speed

ANA. Survival and neuritis outgrowth were


17.4 Discussion
quantified in three optic fields from 20 untreated
and NGF-treated wells (for the results see Rocco
The aim of this study was to provide further
et al. (2015)).
supporting data regarding the role of NGF on
The NGF-exposed rhodopsin-positive cells
the cells of the anterior and posterior segments
(photoreceptor) survived culturing over 6d,
of the eye. The first convincing data indicating the
implying that unless daily supply of NGF is
critical protective role of NGF on retinal cells was
provided, they will degenerate. This hypothesis
initially demonstrated in fish by Turner and
might be supported by the observation that these
Delaney (1979), and in mammalians’ visual
cells express trkANGFR. The NGF-exposed cells
cells a few years later. It emerged that NGF
co-expressed both trkANGFR and rhodopsin. A
administration induces modification of presynap-
widespread neuritis outgrowth and a higher num-
tic elements in adult visual system (Colafrancesco
ber of trkANGFR-positive cells were observed
et al. 2011; Conner and Varon 1994; Cuello et al.
upon NGF stimulation, as compared to untreated
2010; Cuello et al. 1992; Curtis et al. 1995);
or ANA- and/or ANA/NGF-treated cells. Indeed,
prevents the shift in ocular dominance distribu-
high trkANGFR and rhodopsin were
tion of visual cortical neurons and promotes func-
co-expressed in NGF-treated cultures and
tional recovery of RGCs after ischemia
co-localized in ANA-treated cultures. Moreover,
(Siliprandi et al. 1993); delays retinal cell degen-
the confocal analysis indicates that NGF
eration in rodents with inherited retinopathy
stimulates neuritis outgrowth from photoreceptor
(Amendola and Aloe 2002; Liang et al. 2012);
(for the results see Rocco et al. (2015)).
and reduces retinal damages in rabbits affected by
Part of the data reported in this chapter has
ocular hypertension, while NGF antibodies can
been published in a previous study (Rocco et al.
exacerbate the damaging action of RGCs
2015).
(Lambiase et al. 2007). A few years later, it was
reported that in the case of purified NGF topical
application on purified NGF patients with
262 M. L. Rocco et al.

advanced glaucoma running the imminent risk of administration for delivering αNGF to further
loss of visual functions (Coassin et al. 2008; investigate the structural, biochemical, and
Lambiase et al. 2009), NGF promoted progres- molecular role of retinal and corneal cells. Over-
sive improvement of the functionality of the inner all, the information resulting from this study is
retinal layer and of the parameters of the post- that under physiological conditions, NGF
retinal neural conduction becoming manifest dur- regulates not only normal survival of corneal
ing the treatment period, and maintained it for and retinal cells but also NGF-responsive cell
3 months after discontinuation of treatment; degeneration under NGF-deprivation conditions.
visual acuity significantly improved in patients To conclude with, our data demonstrated that
in whom it had remained unchanged after several cross talk between photoreceptor activity and
months’ follow-up. NGF signaling actually exists, and reinforced
To understand the role of NGF more deeply on the concept that NGF is crucial for the regular
the protective mechanisms of corneal and retinal activity of the visual system and, in particular, of
cells, we used intravitreal administration of photoreceptors.
purified αNGF and examined the effect induced
in corneal and retinal cells. The results showed Acknowledgments L.A. by Fondazione IRET, Ozzano
that the αNGF administration has no significant Emilia, Bologna, Italy.
effect on the endogenous presence of NGF in the
retina and considerably reduces the level of NGF
in the cornea. One possible interpretation of these References
different effects might be either that corneal cells
are more vulnerable than retinal cells to the pres- Aloe L (2004) NGF and molecules in health and disease.
ence of αNGF or alternatively that retinal cells Prog Brain Res 2004, 146
express large concentration of NGF. Western blot Aloe L, Tirassa P, Lambiase A (2008) The topical applica-
tion of nerve growth factor as a pharmacological tool
analysis and immunostaining analyses revealed for human corneal and skin ulcers. Pharmacol Res
that αNGF administration induces no significant 57:253–258
alteration of the high-affinity NGF receptor and Aloe L, Rocco ML, Bianchi P, Manni L (2012) Nerve
TrkANGFR in the retina and enhances expression growth factor: from the early discoveries to the poten-
tial clinical use. J Transl Med 10:239
of TrkANGFR protein in the cornea, thus Amendola T, Aloe L (2002) Developmental expression of
suggesting different vulnerability of retinal and nerve growth factor in the eye of rats affected by
corneal cells to αNGF exposure. However, the inherited retinopathy: correlative aspects with retinal
possibility that αNGF administration induces structural degeneration. Arch Ital Biol 140:81–90
Amendola T, Fiore M, Aloe L (2003) Postnatal changes in
changes in other retinal and corneal biological nerve growth factor and brain derived neurotrophic
mediators cannot be excluded. Indeed, it was factor levels in the retina, visual cortex, and geniculate
found that the expression of rhodopsin and nucleus in rats with retinitis pigmentosa. Neurosci Lett
inflammatory markers in the retina is significantly 345:37–40
Bocchini V, Angeletti PU (1969) The nerve growth factor:
downregulated as a result of αNGF administra- purification as a 30,000-molecular-weight protein.
tion. The observed significant decrease in some Proc Natl Acad Sci U S A 64:787–794
cytokine expression has no explanation. Interest- Coassin M, Lambiase A, Sposato V, Micera A, Bonini S,
ingly, the overexpression to IL-21 may possibly Aloe L (2008) Retinal p75 and bax overexpression is
associated with retinal ganglion cells apoptosis in a rat
be related to the activation of Muller cells, which model of glaucoma. Graefes Arch Clin Exp
needs to be investigated to better understand Ophthalmol 246:1743–1749
αNGF action on visual cells. These findings indi- Colafrancesco V, Coassin M, Rossi S, Aloe L (2011)
cate that αNGF administration reaches both cor- Effect of eye NGF administration on two animal
models of retinal ganglion cells degeneration. Ann Ist
neal and retinal cells and assert that the latter can Super Sanita 47:284–289
cause downregulation of endogenous NGF Conner JM, Varon S (1994) Nerve growth factor
expression and retinal cell deficit, thus providing influences the distribution of sympathetic sprouting
a useful experimental approach of noninvasive
17 NGF and Retinitis Pigmentosa: Structural and Molecular Studies 263

into the hippocampal formation by implanted superior Lambiase A, Aloe L, Centofanti M, Parisi V, Bao SN,
cervical ganglia. Exp Neurol 130:15–23 Mantelli F, Colafrancesco V, Manni GL, Bucci MG,
Cuello AC, Maysinger D, Garofalo L (1992) Trophic Bonini S et al (2009) Experimental and clinical evi-
factor effects on cholinergic innervation in the cerebral dence of neuroprotection by nerve growth factor eye
cortex of the adult rat brain. Mol Neurobiol 6:451–461 drops: implications for glaucoma. Proc Natl Acad Sci
Cuello AC, Bruno MA, Allard S, Leon W, Iulita MF U S A 106:13469–13474
(2010) Cholinergic involvement in Alzheimer's dis- Liang XL, Li J, Chen F, Ding XY, Yang XX, Long LX
ease. A link with NGF maturation and degradation. J (2012) A comparing study of quantitative staining
Mol Neurosci 40:230–235 techniques for retinal neovascularization in a mouse
Curtis R, Adryan KM, Stark JL, Park JS, Compton DL, model of oxygen-induced retinopathy. Int J
Weskamp G, Huber LJ, Chao MV, Jaenisch R, Lee KF Ophthalmol 5:1–6
et al (1995) Differential role of the low affinity Pagani L, Manni L, Aloe L (2006) Effects of electroacu-
neurotrophin receptor (p75) in retrograde axonal trans- puncture on retinal nerve growth factor and brain-
port of the neurotrophins. Neuron 14:1201–1211 derived neurotrophic factor expression in a rat model
D'Cruz PM, Yasumura D, Weir J, Matthes MT, of retinitis pigmentosa. Brain Res 1092:198–206
Abderrahim H, LaVail MM, Vollrath D (2000) Muta- Rocco ML, Balzamino BO, Petrocchi Passeri P, Micera A,
tion of the receptor tyrosine kinase gene Mertk in the Aloe L (2015) Effect of purified murine NGF on
retinal dystrophic RCS rat. Hum Mol Genet 9:645–651 isolated photoreceptors of a rodent developing retinitis
Dowling JE, Sidman RL (1962) Inherited retinal dystro- pigmentosa. PlosOne 21:10
phy in the rat. J Cell Biol 14:73–109 Siliprandi R, Canella R, Carmignoto G (1993) Nerve
Faktorovich EG, Steinberg RH, Yasumura D, Matthes growth factor promotes functional recovery of retinal
MT, LaVail MM (1990) Photoreceptor degeneration ganglion cells after ischemia. Invest Ophthalmol Vis
in inherited retinal dystrophy delayed by basic fibro- Sci 34:3232–3245
blast growth factor. Nature 347:83–86 Turner JE, Delaney RK (1979) Retinal ganglion cell
Lambiase A, Coassin M, Sposato V, Micera A, response to axotomy and nerve growth factor antise-
Sacchetti M, Bonini S, Aloe L (2007) NGF topical rum in the regenerating visual system of the newt
application in patients with corneal ulcer does not (Notophthalmus viridescens): an ultrastructural mor-
generate circulating NGF antibodies. Pharmacol Res phometric analysis. Brain Res 177:35–47
56:65–69
NGF in Inflammatory
and Neurodegenerative Diseases 18
of the Eye: New Findings Supporting
Neuroprotection and Proper Tissue
Remodeling in Vitreoretinal Disorders

Graziana Esposito, Bijorn Omar Balzamino, Luca Bruno,


Andrea Cacciamani, and Alessandra Micera

Abstract disease-associated biomarkers and allow the


Nerve growth factor (NGF) plays a crucial role monitoring of retinal impairments with sustain
in retinal disorders, as suggested by in vitro/ to the retinal imaging. Coupled to other solu-
in vivo models. The major effect embraces the ble mediators, NGF has been quantified in
neuroprotective activity on retinal ganglion aqueous (slightly expressed) from diabetic
cells (RGCs) undergoing degeneration, as retinopathy-suffering patients (cataract sur-
observed in experimental diabetic retinopathy, gery) and vitreal reflux (significantly impaired)
age-related and diabetic macular degeneration, of diabetic macular degeneration-suffering
and some vitreoretinal diseases. Focused patients (intravitreal surgery). Although the
experiments suggested that locally applied reasons of these NGF impairments are not
NGF (intravitreal delivery) not only allowed fully comprehended, some retinal cells (glial
the counteraction of RGC degeneration but cells, bipolar neurons, and RGCs) have been
also provided data for a whole retina restora- recognized partially responsible for these local
tion. The currently available retinal microsur- changes.
gery allows the collection of human aqueous Taken together, the recent progress in the
and more interesting vitreous (vitreal reflux) ocular microsurgeries might be associated with
humors. The recent biomolecular analysis sampling of small amount of ocular humors,
highlights the possibility to identify allowing the collection of biochemical informa-
tion about diseased retina and the monitoring of
treatment. The chance to detect NGF and like-
G. Esposito · B. O. Balzamino · L. Bruno · A. Cacciamani wise other neuroprotective or pro-/anti-inflam-
Research and Development Laboratory for Biochemical, matory factors in these fluids would open to the
Molecular and Cellular Applications in Ophthalmological
possibility to identify biomarkers of early diag-
Sciences IRCCS – Fondazione Bietti, Rome, Italy
e-mail: graziana.esposito@fondazionebietti.it; bijorn. nosis or monitoring of retinal disease evolution/
balzamino@fondazionebietti.it; luca. therapy (precision medicine).
bruno@fondazionebietti.it; andrea.
cacciamani@fondazionebietti.it
A. Micera (*)
Research and Development Laboratory for Biochemical,
Molecular and Cellular Applications in Ophthalmological 18.1 Introduction
Sciences IRCCS – Fondazione Bietti, Rome, Italy
Head of Research Laboratories in Ophthalmology, Since the discovery occurred in 1950 by Rita
IRCCS – Fondazione Bietti, Rome, Italy Levi-Montalcini and Stanley Cohen, nerve
e-mail: alessandra.micera@fondazionebietti.it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 265
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_18
266 G. Esposito et al.

growth factor (NGF) has represented the proto- amacrine, photoreceptors, and glial cells synthe-
type of the neurotrophic family (neurotrophins) size and release into the microenvironment the
(Aloe and Chaldakov 2013). The signal pathway, mature NGF form for autocrine/paracrine cell
a peculiar aspect of this NT family (NGF, BDNF, function, homeostatic, as well as survival/protec-
NT-3, NT-4, and NT-5), is driven by two distinct tive effects under physiopathological states (Aloe
receptors (the tyrosine kinase receptors trkANGFR, and Chaldakov 2013). The release of NGF into
trkBBDNFR, trkCNTR and the pan-neurotrophin ocular fluids occurs locally by structural, acces-
p75NTR receptor), working in concert (high- sory, and local or infiltrating immune cells (Aloe
affinity binding; heterodimers) or alone (single and Chaldakov 2013). A pre-proNGF protein
binding; homodimers) to downstream the indica- (140 kDa) is quickly cleaved to a proNGF form
tion inside the cell. Both survival and death (32–25 kDa), the result of specific protease action
signals can be produced to regulate tissue homeo- (intracellularly by furins and extracellularly by
stasis and functional parainflammation, mainly several proteases including metalloproteinases
for NGF (Micera et al. 2007). (MMP)-7, which in turn can be cleaved to pro-
duce a mature NGF (13.5 kDa) (Masoudi et al.
2009). Intracellularly, trkANGFR-NGF binding
18.1.1 NGF and Eye: The Prototype activates mainly pro-survival pathways, while
of a Prodigious Family p75NTR-proNGF triggers inflammatory and apo-
ptotic pathways. NGF prefers the high-affinity
Originally, NGF was believed to stimulate the binding which includes trkANGFR- and p75NTR-
survival, growth, and differentiation of cells coupled receptor, although a ratio between hetero
populating the central nervous system and periph- and homo binding can also occur producing mod-
eral nervous system (CNS and PNS) throughout ulation in the final cell activity. Finally, the
development and adulthood. Later, NGF was mature NGF form binds specifically the tyrosine
demarcated as a pleiotropic factor exerting kinase receptor (trkANGFR), with or without
biological effects to non-neuronal cells, opening cooperation of both receptors, while the proNGF
to new considerations for a modulatory role of form binds and activates selectively the
NGF alongside the neuro-immuno-endocrine pan-receptor p75NTR (Micera et al. 2007).
systems (Aloe and Chaldakov 2013). Over the NGF expression inside retinal layers has been
last decades, several aspects in NGF biological reported in subsequent years and strongly
effects were highlighted, and those related to the associated with protective tasks, especially for
visual system, from the cornea to the conjunctiva, RGC. A general concept was produced after the
with respect to epithelial-stromal cross talk in vitro/in vivo/ex vivo findings of NGF
(keratocytes, fibroblast/myofibroblast), were applications on the visual system, regarding the
highlighted as opening the way to the application possibility of NGF to reduce or counteract
in those human corneal diseases orphan of tangi- neurodegeneration of photoreceptors, neuron
ble treatment (Micera et al. 2004). loss, and Müller cell gliosis. Different strategies
aimed at increasing endogenous NGF availability
by exogenous NGF administration (Sivilia et al.
18.1.2 NGF and NGF Receptors: 2009). The possibility for exogenous NGF to
Distribution and Function reach both the retina and optic nerve was tested
in Posterior Eye Segment in animal models (Lambiase et al. 2005). Modern
techniques allowed to specifically trace the topi-
NGF and receptors (trkANGFR-p75NTR) were cal administration of NGF, confirming the possi-
observed in several ocular fluids and tissues, bility of a retinal targeting by a noninvasive
including aqueous/vitreous, posterior lens, retina, approach. The benefits of local instillation of
and optic nerve (Micera et al. 2007). Retinal NGF eye drops could be twice: a dual effect in
ganglion cells (RGCs), bipolar neurons, improving the quality of a “healthy” ocular
18 NGF in Inflammatory and Neurodegenerative Diseases of the Eye: New. . . 267

surface epithelium and reaching the retina. Infor- were observed for NGF levels, as discussed in
mation from animal models opened to a wider detail below (Cacciamani et al. 2019).
usage of topical NGF applications inside the eye From experimental studies, NGF changes
upon pathological states. Other recent experimen- were observed in the vitreous from animal
tal studies opened to the use of intravitreal NGF models, including reeler (defected retinogenesis)
administration to counteract retina degeneration mice and RCS (diabetic retina) rats (Balzamino
(Rocco et al. 2017). More recent studies et al. 2014; Rocco et al. 2017). Vitreous from
highlighted the NGF ability to promote survival DR-suffering patients showed changes in the nor-
of retinal and even brain cells when NGF was mal levels of NGF, BDNF, NT-3, and NT-4, as
applied intraocularly and/or topically (Aloe and observed from the analysis of vitreous/vitreal
Chaldakov 2013). reflux samples collected during the intravitreal
injections or vitrectomy (therapeutic vitreoretinal
surgery) (Cacciamani et al. 2016). A correlation
18.2 NGF and Vitreoretinal between vitreal NGF and inflammatory mediators
Disorders was reported in these patients and further
associated with inflammatory/oxidative stress
Vitreoretinal disorders are increasing in the last conditions (Cacciamani et al. 2019).
decade. The vitreous and retina can be affected by Retinal cell layers are structurally organized to
a variety of insults, and a number of vitreoretinal accomplish the vision function in a proper and
surgery techniques have been developed in the smart way. As the principal glia of the retina,
last 10 years, to counteract vision loss. Not all Müller cells keep in touch with all neuronal
vitreoretinal impairments are age-related subsets, providing a unique support in neuronal
diseases: a direct consequence of certain diseases networking and allowing a link with vessels
(diabetes, myopia) or even trauma (crash, colli- through their processes, forming/maintaining the
sion) or lifestyling (amateur activities/sport) can BRB, regulating retinal glutamate levels, and
exert a major influence, resulting in sight- stabilizing blood flow. Müller cells represent an
threatening and severe impact on person’s daily important source of VEGF, NGF, basic fibroblast
activities and quality of life. At the biomolecular growth factor-2 (bFGF2), tumor necrosis factor,
level, diabetic retinopathy (DR) and age-related and other soluble mediators with neuroprotective
macular degeneration (AMD) are directly effects (Wang et al. 2016). From experimental
associated with alterations of the blood-retinal in vitro/in vivo studies, both NGF receptors are
barrier (BRB), merely the inner BRB for DR differentially expressed by resting and activated
and the outer BRB for AMD (Vieira-Potter et al. Müller cells, sustaining the involvement of NGF
2016). Dysfunction of BRB occurs by increased signaling in Müller cells’ functions (Balzamino
microvascular permeability and culminates in et al. 2014).
diabetic macular edema (DME), also leading to When infiltration of inflammatory/immune
decreased visual acuity (Vieira-Potter et al. 2016). cells occurs inside the retinal layers, the choroid
At the extracellular level, neovascularization and represents the “main” entrance implying that the
increase of several pro-/antiangiogenic factors “systemic inflammatory status” can severely
have been detected, including vascular endothe- affect the retinal one. The plethora of soluble
lial growth factor (VEGF) (Wang et al. 2016; mediators includes growth and angiogenic-
Vujosevic et al. 2016). angiostatic factors, neurogenic components, as
Current treatments for DR and DME are well as neurotrophins, in addition to the common
repeated anti-VEGF injections into vitreal cham- inflammatory path (cytokines and chemokines)
ber to reduce the high VEGF levels (Vieira-Potter (Wang et al. 2016; Vujosevic et al. 2015;
et al. 2016). Those injections have been Cacciamani et al. 2019). This wide-range influx
associated with decrease of VEGF and increase can severely perturbates the conservative asset of
of other mediators, while no significant changes retinal layers, and the local parainflammation can
268 G. Esposito et al.

quickly shift to inflammation with angiogenesis Upon a chronic insult, NGF levels occur highly
and neovascularization, opening to harmful expressed at beginning while lowing at late states
consequence. of disease. This NGF expression is often
associated with inflammation, erroneously
pointed as inducing inflammation. By itself,
18.2.1 NGF as Bystander or Partaker NGF does not trigger inflammation, although
in the Cross Talk Between being a proliferating factor for different inflam-
Resident and Infiltrating Cells matory cells (Aloe and Chaldakov 2013). Even if
a protein signature is quite visible for DR, DME,
Inflamed retina is a suitable microenvironment to AMD, and vitreoretinal diseases in general, actu-
study the interplay between inflammation, ally no biomarker or a few biomarker signatures
neuroprotection, and matrix remodeling. All detectable in ocular fluids can predict the ongoing
these steps can discern a simple and reversible of an inflamed retina (Wang et al. 2016). There-
damage from a neurodegenerative one. Diabetes fore, indications in changes of NGF and VEGF
is a neurodegenerative disease characterized by levels in vitreal fluids might represent an addi-
chronic hyperglycemia that can cause damage of tional information for monitoring the current ther-
body microvasculature and thereafter silent apy (Cacciamani et al. 2019).
insults to the retinal microvasculature with vision In recent years, increasing data support the
deficits (Vieira-Potter et al. 2016). The long- contribution of matrix enzymes in the remodeling
lasting insults occurring at the retinal layers, due of inflamed retina. The well-known process of
to non-stabilized glycemia, can offend continu- conversion from pro- to mature NGF is driven
ously the retinal layers, with a reduced retinal by furins and MMP-7, intracellularly localized.
thickness, neurofilament abnormality, and An impaired MMP-7 activity might cause an
declined ERG, all steps culminating in retinal imbalanced expression between these two forms
cell death and blindness (Vieira-Potter et al. in both the retina and circulating, as observed for
2016). diabetic retinopathy (Ali et al. 2011). As the early
The eye comprises two fluid systems, whose retinal neurodegeneration is linked to a decrease
fluid density is regulated by circulation and drain- of NGF expression, the exogenous supply might
age, guaranteeing homeostasis and perfect func- avoid retinal damage as reported for animal
tion (Pizzirani and Gong 2015). One is in front of models of diabetes (Ali et al. 2011). The NGF
aqueous and the other behind of vitreous lens, content in aqueous and vitreous from diabetic
which compositions and clearness are the result subjects is shown in Fig. 18.1.
of gender, age, systemic and local factors, and
life-styling features. Ciliary body system
contributes massively to aqueous composition, 18.2.2 Neuroprotection: NGF-VEGF
keeping functioned both sclerocorneal and retinal Race for RGC, Photoreceptor,
structures. So, glucose and other systemic Bipolar, and Müller Cell
components (complement, albumin, insulin, etc.) Protection
are equilibrated and drained (Vieira-Potter et al.
2016). Supported by several clinical trials and a current
The early expression/production of NTs might routine approach, anti-VEGF remains a mainstay
represent an attempt to trigger neuronal survival to control posterior segment conditions like
signaling to quickly compensate the neuronal neovascular (or wet) age-related macular degen-
insult and counteract as early as the initial neuro- eration (nAMD) (Cacciamani et al. 2019). While
degenerative process. The association between various molecules suitable for retinal protection
DR and altered NGF, BDNF, NT-3, and NT-4 have shown efficacy in maintaining vision and
levels and/or expression is consistent with the preventing disease progression, there is still
hypothesis (Mohamed and El-Remessy 2015). much debate on the current treatment and ideal
18 NGF in Inflammatory and Neurodegenerative Diseases of the Eye: New. . . 269

Fig. 18.1 NGF levels in vitreoretinal disorders. The anti- condition (DR). Briefly, vitreal reflux was collected during
VEGF intravitreal injection represents a routine therapeu- intravitreal surgery, while vitreous was sampled at the time
tic approach in several retinal disorders, to delay but not of pars plana vitrectomy, a commonly employed technique
reverse/cure the damage of the retina. (a) Representative in vitreoretinal surgery. (c) NGF concentration in the
SD-OCT (spectral domain optical coherence tomography) vitreal samples from pucker and macular holes. Vitreal
scan image from a DME retina. Briefly, OCT is a nonin- samples were collected during pars plana vitrectomy.
vasive, noncontact imaging modality used to visualize and NGF levels were quantified by ELISA, with antibodies
monitor changes of retinal layers. (b) The higher expres- that do not recognize the other neurotrophins (b–c)
sion in vitreal reflux is consistent with the inflammatory

regimen/most efficacious intervals of signal-regulated kinase) and finally the VEGF


administration. gene activation (Wang et al. 2016).
New indications suggest that Müller cells are The neuronal counterpart of the retinal tissue
active partakers in DR and vitreoretinal diseases interacts with vasculature through a complex net-
(Zhang et al. 2019). Essential during work responsible for guaranteeing tissue homeo-
retinogenesis and adult homeostasis, Müller stasis and preserving retinal network functionality
cells are present in the entire depth of neural (Segatto et al. 2019). Among those factors,
retina (Zhang et al. 2019). As observed in vitro, VEGF, by means of VEGFR-1 and VEGFR-2-
Müller cell-derived NGF can increase VEGF receptors, and NGF, through specific and
expression, providing an active boost for physio- pan-neurotrophin receptors, might be additional
logical retinal neovascularization and cell prolif- main players. Some few studies on treated
eration via activating ERK1/2 and AKT signaling patients indicate that anti-VEGF treatment nega-
(Wang et al. 2016). The retinal layers, mainly tively influences the survival of retinal neuronal
crossed by Müller cells, are continuously per- and the chronic neutralization of local VEGF
fused by a variety of neurotropic factors (Zhang might prime neuron loss (Fogli et al. 2018). This
et al. 2019). A new insight does not exclude would suggest that the neuroprotective effect of
Müller-derived NGF as an ancillary VEGF is actually a debate: it is quite clear that a
neuroprotective agent. Merely, NGF release into complete neutralization of VEGF is followed by
retinal microenvironment can sustain neurons and other tissue alterations as well as angiogenesis
other retinal subsets (Rocco et al. 2015). In an and neovascularization can offend seriously reti-
autocrine/paracrine manner, NGF can sustain nal network. VEGF acts as a potent trophic factor
Müller cell activation and consequently different for retinal neurons, facilitating both proliferation
signaling pathways involved also in Müller cells and differentiation of retinal progenitors into
spreading through the specific trkANGFR signal specialized neurons, while its overt production
pathway (PI3K, phosphatidylinositol 3-kinase; promotes new vessel formations with deleterious
Akt, protein kinase B; and ERK1/2, extracellular effects on whole retinal network (Fogli et al.
2018). Recently, NGF has been proposed as
270 G. Esposito et al.

important angiogenic growth factors, as direct cell degeneration (Zhou et al. 2014). Both degen-
enhancers of VEGF expression (Nakamura et al. eration and Cyr61 overexpression in Müller cells
2011). While NGF stimulates VEGF production, take critical parts in the pathogenesis of
there is no evidence for VEGF induction of NGF, DR. Elevated Cyr61 levels were quantified in
highlighting the importance of NGF in clinical the vitreous from PDR patients (Zhou et al.
applications. Anti-VEGF therapy is widely used 2014). Recently, our group observed a correlation
as the primary treatment for counteracting retinal between NGF, VEGF, OPN, and cyr61 at least in
neovascularization (DR, AMD, CNV, and retinal ERMs (Dinice et al. 2020). This association
vein occlusion) (Cacciamani et al. 2016). By the should be verified in Müller cells exposed to
way, experimental data showed a risen apoptosis vitreal samples from DR, including the
of retinal cells after anti-VEGF administration, proliferative forms, AMD and CNV.
clearly visible with a reduced thickness and
NGF production (Fogli et al. 2018). Thus, the
purpose of our studies was to evaluate the 18.3 Conclusion and Future
human vitreous reflux, obtained during anti- Perspectives
VEGF administration, in the modulation of dif-
ferent biomarkers (Cacciamani et al. 2016). Several lines of evidence highlight a protective
effect of NGF against retinal cell loss, with spe-
cific emphasis on reduction of retinal ganglion
18.2.3 Tissue Remodeling: NGF cell degeneration, photoreceptor death, and
and Extracellular-Matrix Müller cell gliosis. A better understanding of the
Interplay mechanisms underlying the effects of NGF and
proNGF as well as NGF/BDNF/NT4/p75NTR in
Chronic (Th1- and Th2-mediated) inflammatory the modulation of retinal neurodegeneration and
conditions might influence the local innervation gliosis will help to develop efficient therapeutic
as well as trigger the development of complex strategies for various retinal diseases. Different
neurogenic mechanisms including the release of strategies, including increasing the availability
neuromediators (VIP, NPY, and CGRP), of endogenous NGF, administration of exoge-
representing a novel mechanism in the pathogen- nous NGF, activation of trkANGFR, and inhibition
esis of ocular surface (Micera et al. 2004). The of p75NTR, have been reviewed highlighting the
same mechanisms might be activated in autoim- potential aspects in NGF neuroprotection. Our
mune and neurodegenerative states, including studies showing the increased NGF levels inside
type I and type II diabetes. In experimental the vitreal chamber would suggest an initial
models of DR, NGF and its pro-forms and own attempt of NGF to protect neuronal cells from
receptors were found overexpressed in degeneration. A potential therapeutic intervention
GFAP- and Iba1-bearing cells, in association with NGF for the treatment of retinal neurodegen-
with several intracellular pathways (Rocco et al. erative diseases can be therefore suggested as an
2015). Other tissue remodeling proteins have implementation of endogenous NGF. The major
been associated with retinal remodeling upon limitation of NT treatment for CNS diseases is
inflammation. A specific function for OPN in represented by the difficulty of crossing the
homeostatic processes is to facilitate the survival blood-brain barrier and the impossibility to
of stressed endothelial cells, possibly by deliver NTs systemically, particularly NGF
occupying un-ligated integrins and suppressing (Giannaccini et al. 2018). Alternatives for exoge-
integrin-mediated death (Dinice et al. 2020). In nous delivery of NTs has been proposed for the
other studies, on Müller cells, exposure to high treatment of retinal disorders: intravitreal injec-
glucose increased Cyr61 expression, inhibited tion and the nanoscale MNP-protein hybrid rep-
cell viability/migration, and induced apoptosis, resent excellent tools for bypassing the limit of
suggesting the potential role of Cyr61 in Müller BRB and implementing ocular drug delivery
18 NGF in Inflammatory and Neurodegenerative Diseases of the Eye: New. . . 271

Fig. 18.2 Different routes of drug administration in the The periocular route includes subconjunctival, subtenon,
eye: a glimpse to NGF. The draw summarizes all accessi- peribulbar, and retrobulbar sites of injection. Except for
ble intraocular sites as indicated: topical absorption (eye the systemic route, all the others have been tested for NGF
drops and emulsions/gels) in the anterior segment and and anti-NGF at both experimental and clinical levels.
related to the posterior segment, the intravitreal injections Highlighted in red are future directions on experimental
(ophthalmic solutions/suspensions, implants, nanotech- animals, comprising the anti-VEGF/NGF-combined
nology), periocular administration (implants or injections), intravitreal injection or the use of NGF nanoparticles lay-
and finally the systemic delivery (oral and intravenous). ered in vitreal chamber

strategies for neuroprotection and therapy touch by fluid circulation. The poor permeability
(Giannaccini et al. 2018). As above introduced, of the cornea/sclera and BRB makes the systemic
the eye has two fluid systems and own barriers, administration of NTs ineffective in reaching the
providing a limited and immune-privileged sys- retina as well as the short half-life of NTs ideally
tem (Pizzirani and Gong 2015). The anterior seg- bypassed by multiple injections (Gaudana et al.
ment includes tear film, conjunctiva, cornea, 2010). For this reason, several attempts were car-
aqueous, and iris, and the posterior segment ried out to find out alternatives to reach the retina,
contains the vitreous humor and the underneath such as topical application as well as intravitreal,
retina with retinal pigment epithelium (RPE) and periocular, subretinal, or systemic administration
choroid. The lens represents the physical edge (Fig. 18.2). To combine anti-VEGF/NGF therapy
between those two compartments that are in or use NGF nanoparticle delivery represents the
272 G. Esposito et al.

starting points for our future investigations in growth factor in the retina and optic nerve of adult
experimental models. rats. Invest Ophthalmol Vis Sci 46(10):3800–3806.
https://doi.org/10.1167/iovs.05-0301
Masoudi R, Ioannou MS, Coughlin MD et al (2009)
Acknowledgments Many thanks to Tiziana Pacchetti for Biological activity of nerve growth factor precursor is
precious assistance in collecting fluids and tissues at sur- dependent upon relative levels of its receptors. J Biol
gery and Angelica Napoli for drawing Fig. 17.2. This work Chem 284(27):18424–18433. https://doi.org/10.1074/
was partially supported by MaBIOS Project (Regione jbc.M109.007104
Lazio; LR13/2015; FILAS-RU-2014-112), Italian Minis- Micera A, Lambiase A, Aloe L, Bonini S, Levi-Schaffer F,
try of Health (ricerca corrente), and 5xMille (2015 and Bonini S (2004) Nerve growth factor involvement in
2016) to IRCCS Fondazione Bietti. the visual system: implications in allergic and neuro-
degenerative diseases. Cytokine Growth Factor Rev 15
(6):411–417. https://doi.org/10.1016/j.cytogfr.2004.
09.003
References Micera A, Lambiase A, Stampachiacchiere B et al (2007)
Nerve growth factor and tissue repair remodeling: trkA
Ali TK, Al-Gayyar MMH, Matragoon S et al (2011) Dia- (NGFR) and p75(NTR), two receptors one fate. Cyto-
betes induced peroxynitrite impairs the balance of kine Growth Factor Rev 18(3–4):245–256. https://doi.
pro-nerve growth factor and nerve growth factor, and org/10.1016/j.cytogfr.2007.04.004
causes neurovascular injury. Diabetologia 54(3): Mohamed R, El-Remessy AB (2015) Imbalance of the
657–668 nerve growth factor and its precursor: implication in
Aloe L, Chaldakov GN (2013) The multiple life of nerve diabetic retinopathy. J Clin Exp Ophthalmol 6(5):483.
growth factor: tribute to rita levi-montalcini https://doi.org/10.4172/2155-9570.1000483
(1909–2012). Balkan Med J 30(1):4–7 Nakamura K, Tan F, Li Z, Thiele CJ (2011) NGF activa-
Balzamino BO, Biamonte F, Esposito G et al (2014) tion of TrkA induces vascular endothelial growth fac-
Characterization of NGF, trkA (NGFR), and p75 tor expression via induction of hypoxia-inducible
(NTR) in retina of mice lacking Reelin glycoprotein. factor-1α. Mol Cell Neurosci 46(2):498–506. https://
Int J Cell Biol 2014:725928. https://doi.org/10.1155/ doi.org/10.1016/j.mcn.2010.12.002
2014/725928 Pizzirani S, Gong H (2015) Functional anatomy of the
Cacciamani A, Parravano M, Scarinci F et al (2016) A outflow facilities. Vet Clin North Am Small Anim
simple spontaneous Vitreal reflux collecting procedure Pract 45(6):1101–1126. https://doi.org/10.1016/j.
during intravitreal injection: set-up and validation stud- cvsm.2015.06.005
ies. Curr Eye Res 41(7):971–976. https://doi.org/10. Rocco ML, Balzamino BO, Petrocchi Passeri P, Micera A,
3109/02713683.2015.1080282 Aloe L (2015) Effect of purified murine NGF on
Cacciamani A, Esposito G, Scarinci F, Parravano M, isolated photoreceptors of a rodent developing retinitis
Dinice L, Di Nicola M, Micera A (2019) Inflammatory pigmentosa. PLoS One 10(4):e0124810. https://doi.
mediators in the vitreal reflux of patients with diabetic org/10.1371/journal.pone.0124810
macular edema. Graefes Arch Clin Exp Ophthalmol Rocco ML, Balzamino BO, Esposito G et al (2017) NGF/
257(1):187–197. https://doi.org/10.1007/s00417-018- anti-VEGF combined exposure protects RCS retinal
4169-4 cells and photoreceptors that underwent a local wors-
Dinice L, Cacciamani A, Esposito G et al (2020) ening of inflammation. Graefes Arch Clin Exp
Osteopontin in vitreous and idiopathic epiretinal Ophthalmol 255(3):567–574. https://doi.org/10.1007/
membranes. Graefes Arch Clin Exp Ophthalmol 258 s00417-016-3567-8
(7):1503–1513. https://doi.org/10.1007/s00417-020- Segatto M, Fico E, Gharbiya M et al (2019) VEGF inhibi-
04685-w tion alters neurotrophin signalling pathways and
Fogli S, Del Re M, Rofi E, Posarelli C, Figus M, Danesi R induces caspase-3 activation and autophagy in rabbit
(2018) Clinical pharmacology of intravitreal anti- retina. J Cell Physiol 234(10):18297–18307. https://
VEGF drugs. Eye (Lond) 32(6):1010–1020. https:// doi.org/10.1002/jcp.28462
doi.org/10.1038/s41433-018-0021-7 Sivilia S, Giuliani A, Fernández M, Turba ME, Forni M,
Gaudana R, Ananthula HK, Parenky A, Mitra AK (2010) Massella A, De Sordi N, Giardino L, Calzà L
Ocular drug delivery. AAPS J 12(3):348–360. https:// (2009 May 27) Intravitreal NGF administration
doi.org/10.1208/s12248-010-9183-3 counteracts retina degeneration after permanent carotid
Giannaccini M, Usai A, Chiellini F, Guadagni V, artery occlusion in rat. BMC Neurosci 10:52. https://
Andreazzoli M, Ori M, Pasqualetti M, Dente L, Raffa doi.org/10.1186/1471-2202-10-52
V (2018) Neurotrophin-conjugated nanoparticles pre- Vieira-Potter VJ, Karamichos D, Lee DJ (2016) Ocular
vent retina damage induced by oxidative stress. Cell complications of diabetes and therapeutic approaches.
Mol Life Sci 75(7):1255–1267. https://doi.org/10. Biomed Res Int 2016:3801570. https://doi.org/10.
1007/s00018-017-2691-x 1155/2016/3801570
Lambiase A, Tirassa P, Micera A, Aloe L, Bonini S (2005)
Pharmacokinetics of conjunctivally applied nerve
18 NGF in Inflammatory and Neurodegenerative Diseases of the Eye: New. . . 273

Vujosevic S, Micera A, Bini S, Berton M, Esposito G, proliferation via ERK1/2 and AKT signaling in Müller
Midena E (2015) Aqueous humor biomarkers of cells. Mol Vis 22:254–263. eCollection 2016
Müller cell activation in diabetic eyes. Invest Zhang L, Li X, Lin X, Wu M (2019) Nerve growth factor
Ophthalmol Vis Sci 56(6):3913–3918. https://doi.org/ promotes the proliferation of Müller cells
10.1167/iovs.15-16554 co-cultured with internal limiting membrane by
Vujosevic S, Micera A, Bini S, Berton M, Esposito G, regulating cell cycle via Trk-a/PI3K/Akt pathway.
Midena E (2016) Proteome analysis of retinal glia BMC Ophthalmol 19(1):130. https://doi.org/10.
cells-related inflammatory cytokines in the aqueous 1186/s12886-019-1142-x
humour of diabetic patients. Acta Ophthalmol 94 Zhou F, Zhang Y, Chen D et al (2014) Potential role of
(1):56–64. https://doi.org/10.1111/aos.12812 Cyr61 induced degeneration of human Müller cells in
Wang J, He C, Zhou T, Huang Z, Zhou L, Liu X (2016) diabetic retinopathy. PLoS One 9(10):e109418. https://
NGF increases VEGF expression and promotes cell doi.org/10.1371/journal.pone.0109418
Part VI
Veterinary Medicine
Nerve Growth Factor (NGF) and Animal
Reproduction 19
Margherita Maranesi, Cristiano Boiti, and Massimo Zerani

Abstract luteotrophic action, while, in induced


Stimuli that lead to the release of ovulators it is the main ovulation-inducing
gonadotropin-releasing hormone (GnRH) and factor. A better understanding of the role of
pituitary gonadotropins and, consequently, NGF in reproduction would be of great inter-
ovulation in mammals fall into two broad est, since it could help finding innovative ther-
categories. In the first, high plasma oestrogen apeutic aids to improve mammalian fertility.
concentrations induce the events that trigger
ovulation, a characteristic of spontaneous
ovulators. In the second, nerve stimuli occur-
ring during mating reach the hypothalamus 19.1 Introduction
and trigger the release of GnRH and ovulation
with a neuroendocrine reflex that characterizes Ovulation in domestic animals can be induced
induced ovulators. (reflex) or spontaneous. The hypothalamic-
In this review, we will give an overview of pituitary-gonadal (HPG) axis is involved in both
the distribution of NGF and its expression in cases. In spontaneous ovulation species, the
the different tissues of the male accessory sex oestrogens cyclically produced by ovarian
glands, the main sites of NGF production. follicles exert a positive feedback on the hypotha-
Next, we will highlight the role of NGF in lamic nuclei that are responsible for the produc-
sperm function and its potential tion of GnRH, determining the preovulatory peak
cryopreserving role in artificial insemination that leads to ovulation. In induced ovulation spe-
techniques. Finally, we will evaluate the cies, the sexual act triggers, by means of afferent
functions of NGF in ovulation, particularly in neuronal pathways, the activation of the HPG axis
induced ovulators. Overall, the information leading to ovulation (Seibel 1986; Milligan
obtained so far indicates that NGF is widely 1982). In the induced ovulation species such as
distributed in organs that regulate the repro- rabbits, cats, camelids, squirrels, and ferrets, there
ductive activity, in both males and females. In is not a real oestrous cycle. Different signals,
spontaneous ovulators, NGF exerts mainly a which may be tactile, olfactory, and visual, assist
the ovulatory response in these species. However,
the stimulation of the genital tract during mating
M. Maranesi (*) · C. Boiti · M. Zerani
Dipartimento di Medicina Veterinaria, Università di turns out to be decisive for triggering the LH peak
Perugia, Perugia, PG, Italy and ovulation (Bakker and Baum 2000).
e-mail: margherita.maranesi@unipg.it; massimo.
zerani@unipg.it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 277
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_19
278 M. Maranesi et al.

Despite our understanding of the factors to be widely distributed in central neurons, NGF
inducing ovulation in these species that has was subsequently found to be also expressed in
improved in recent years, much of the intricate several other cell types and organs such as the
mechanistic details still remains elusive and immune (Ehrhard et al. 1993), vascular endothe-
represents one of the most fascinating themes of lial, and muscular smooth cells (Yamamoto et al.
reproductive physiology. 1996), the lung (Ricci et al. 2007), the adrenal,
The nerve growth factor (NGF) was the first and the thyroid (Saruta et al. 2010; Ceccanti et al.
member of the neurotrophin family to be discov- 2013). Remarkably, among the numerous body
ered (Levi-Montalcini 1987), followed by brain- sources of NGF, the pivotal paper of Ratto et al. in
derived growth factor (BDNF) and other 2012 demonstrated the NGF is highly conserved
neurotrophins (NT3, NT4). Together, these among species in the seminal fluid identified as
neurotrophins support survival, development, dif- the OIF.
ferentiation, and synaptic plasticity of both cen-
tral and peripheral neurons and, therefore, play an
essential biological role (Thoenen and Barde
19.2 Male
1980; Snider 1994). Recently, however, several
studies have shown that NGF also influences the
19.2.1 Expression of NGF in Male
reproductive function of both males and females
Accessory Glands
(Bogle et al. 2018; Ratto et al. 2019).
In the mid-1980s, an ovulation-inducing factor
In 1980, Harper and Thoenen firstly described the
(OIF) was identified in the seminal plasma of
distribution of NGF in mammalian male sex
camelids, which are induced ovulators (Chen
organs and its relevance for male reproductive
et al. 1985). Later, OIF was found in the seminal
functions. From there on, several works on this
plasma of other induced ovulators such as koalas
topic were realized (see, e.g. Adams et al. 2016;
(Johnston et al. 2004), rabbits (Silva et al. 2011),
Bogle et al. 2018).
alpacas, llamas, and further camelids (Adams
NGF and its receptors have been detected in
et al. 2005; Silva et al. 2014; Ulloa-Leal et al.
different organs of the male reproductive tract
2014; Berland et al. 2016; El Allali et al. 2017).
(Shikata et al. 1984; MacGrogan et al. 1991; Li
OIF was even detected in the seminal plasma of
et al. 2005; Wang et al. 2011). In a recent work,
species with spontaneous ovulation, such as cat-
Adams et al. (2016) evidenced the immunoreac-
tle, pigs, and horses (Adams et al. 2016).
tivity of NGF in the secretory epithelium of sev-
Subsequent biochemical studies identified NGF
eral male accessory glands: the ampulla, prostate,
as the major OIF component of seminal plasma
bulbourethral gland, vesicular gland, and
that induced ovulation in llamas (Silva et al.
coagulating gland. Then, Bogle et al. (2018)
2014) and alpacas (Kershaw-Young et al. 2012).
extended this research to other organs and tissues
Two classes of receptors mediate the
including the testis, epididymis, ductus deferens,
biological effects of NGF. The first is a high-
ampulla, and penis of llamas, cattle, bison, elks,
affinity-specific 140-kDa transmembrane
white-tailed deer, and rats. They reported that
neurotrophic tyrosine kinase receptor
differences in the NGF expression among species
1 (NTRK1; also known as tropomyosin receptor
were likely related to differences in the anatomy
kinase A—TrKA) and the second a low-affinity
and function of accessory sex glands (Adams
75-kDa receptor, p75, or nerve growth factor
et al. 2005; Ratto et al. 2012).
receptor (NGFR) belonging to the tumour necro-
The perinuclear distribution of NGF in several
sis factor family receptor (Johnson et al. 1986;
cell types of the male genital apparatus,
Kaplan et al. 1991; Dechant and Barde 1997;
demonstrates protein translation of NGF and stor-
Schecterson and Bothwell 2010). Although
age into secretory vesicles (Bogle et al. 2018;
initially investigated extensively for its nerve
Wang et al. 2011).
growth-promoting effects and then demonstrated
19 Nerve Growth Factor (NGF) and Animal Reproduction 279

In llamas, NGF is particularly expressed in the species displays a positivity in the connective
prostate gland rather than in other accessory tissue and smooth muscle (Bogle et al. 2018). In
glands (Bogle et al. 2018). In testicles of llamas, rabbits, no positivity for NGF was found in the
NGF immunoreactivity was localized only in the epididymis at different ages of sexual maturity
vascular endothelium and connective tissue of the (22 and 37 weeks); conversely, TrKA immunore-
stroma and rete testes, but not in the interstitial activity was evidenced only in younger rabbits at
cells (Bogle et al. 2018). 22 weeks, within stereocilia and epithelial cells
In rats, NGF was mainly expressed in (Sanchez-Rodriguez et al. 2019). In the testis of
coagulating glands (especially in the connective rabbits, intense immunostaining for NGF and its
tissue), but a strong staining was observed also in receptors was observed in the perinuclear region
interstitial cells of the testes. In the same species, and cytoplasm of Leydig, Sertoli, and germinal
Squillacioti et al. (2009) found that the prostate cells at different stages of development (Maranesi
and seminal vesicles were the major sites of NGF et al. 2015).
expression. One of the most intriguing findings was the
In bulls, NGF mRNA abundance was highest NGF positivity of vascular endothelium of the
in the vesicular gland, intermediate in the male genital apparatus (Bogle et al. 2018). NGF
ampulla, and lowest in the bulbourethral and could play an important role in the vascularization
prostate glands. By immunohistochemistry, the of genital tissues, as shown for female llamas,
most intense NGF staining was found in the where NGF extracted from seminal plasma
ampulla and vesicular glands (Hofmann and increased luteal vasculature (Silva et al. 2017),
Unsicker 1982; Adams et al. 2016; Stewart et al. or female rats, where NGF contributed to ovarian
2018) with a strong immune reaction in the apical vascularization via the vascular endothelial
region of the glandular epithelium (Bogle et al. growth factor (VEGF—Julio-Pieper et al. 2006).
2018). On the other hand, there was a lesser NGF
staining intensity in the prostate and
bulbourethral glands (Harper and Thoenen 1980; 19.2.2 NGF and Sperm Function
Harper et al. 1982; Bogle et al. 2018).
In rabbits, the highest levels of NGF and Several authors have assessed the role of NGF
NTRK1 transcripts were found in the prostate, and its receptors in spermatogenesis and sperm
while intermediate expressions were found in function of several animal species and humans
the testis. In these animals, the main source of (Li and Zhou 2013; Adams et al. 2016; Bogle
NGF is the prostate as suggested by the higher et al. 2018; Jin et al. 2006; Levine et al. 2000).
NGF biological activity (Harper and Thoenen Strong immune reactions for NGF and NGFR
1980), the widespread distribution of NGF in all were found in germinal cells and spermatozoa,
prostate glandular cells, and its relatively high suggesting an important intervention of these
mRNA abundance (Maranesi et al. 2015; molecules in all steps of spermatogenesis
Sanchez-Rodriguez et al. 2019). Based on immu- (Maranesi et al. 2015). Transcript corresponding
nohistochemical and molecular studies, the pros- to both NGF and TrkA has been detected in
tate gland is thought to be the main source of human foetal testis, and the NGF protein was
seminal NGF also in llamas, alpacas, and guinea mainly localized to Sertoli and interstitial cells
pigs, while the ampulla and prostate are richer in (Robinson et al. 2003). Functional assays
elks and white-tailed deer, respectively, and the performed with a mouse tumour Leydig cell line
ampulla and vesicular glands in cattle and bison have revealed that NGF increased cellular steroid
(Adams et al. 2016; Bogle et al. 2018). production, indicating that the neurotrophin could
In the epididymis, the caudal region seems to regulate testis morphogenesis (Müller et al.
have greater NGF expression than the other 2006). This NGF role in promoting proliferation
segments (Jin et al. 2010), especially in the con- and differentiation of Leydig cells as well as
nective tissue. Similarly, the penis of different testosterone production was demonstrated also
280 M. Maranesi et al.

in rats by Zhang et al. (2013). In addition, Perrard seasonal changes found in male rabbits agree with
et al. (2007) proposed that NGF has a role also in the findings of Schneidgenova et al. (2011) that
regulating the meiosis of rat spermatocytes. showed less sperm motility and concentration
Remarkably, the seminal plasma of oligoasthe- during the winter season and may reflect the nat-
nozoospermic men had lower NGF content than ural fluctuation of breeding capability during
that of fertile men (Li et al. 2010a, b), suggesting the year.
a relevant role of NGF in sperm function. Using an immunofluorescent technique, NGF
Increased plasma concentrations of NGF report- immunoreactivity was localized to the sperm
edly affect the pattern and intensity of TrkA and head and tail, whereas that of TrkA was detected
p75 in rat testicles, suggesting that the in the acrosomal cap, nucleus, and tail regions. In
NGF/TrkA/p75 system has a role in male repro- addition, in vitro exogenous NGF administration
ductive physiology (Levanti et al. 2006). significantly affected the secretion of leptin, cell
In all species insofar studied, NGF stimulates viability, and sperm apoptosis (Li et al. 2010a).
sperm motility (Lin et al. 2015; Shi et al. 2012; Squillacioti et al. (2009) observed that testicular
Saeednia et al. 2015; Castellini et al. 2019) and steroids likely regulated the expressions of NGF
viability (Li et al. 2010a) and also facilitates and NTRK1 receptor in the accessory sex glands
sperm cell acrosome reactions (Li and Zhou because castration enhanced their expression.
2013). These NGF functions are mediated in a NGF increased Leydig cell steroid production in
time- and dose-dependent manner (Jin et al. an in vitro system (Müller et al. 2006). By con-
2010). trast, NGF expression and concentration in semi-
The great differences in the abundance of nal plasma and blood plasma remain constant
OIF/NGF among species suggest a greater physi- throughout the maturation process, independently
ological importance in camelids. In fact, NGF of testosterone level (Sanchez-Rodriguez et al.
represented approximately 27% of total protein 2019).
in llama/alpaca ejaculates (1.2  0.21 mg/dl;
Bogle et al. 2018), 1.5% of total protein in rabbits
(ranging from 1894  277 pg/ml to 19.2.3 NGF and Preservation
151.9  9.25μg/ml; Maranesi et al. 2015, 2018), of Spermatozoa
and less than 1% of total protein in bovine
ejaculates (0.10  0.03 mg/dl; Bogle et al. 2018). Spermatozoa are highly sensitive to several types
In a recent study, Bezerra et al. (2019) of chemical/physical stress due to reactive oxy-
described the proteome of seminal plasma in gen species, osmotic changes, and different
rabbits. Interestingly, in this work sperm motility, temperatures applied during cryopreservation or
viability, and morphology were associated with freeze-drying procedures (Koshimoto and Mazur
specific seminal proteins. The in silico analysis 2002). Semen conservation methods can affect
pointed out to NGF interactions with some DNA methylation, thus influencing gene expres-
proteins involved in acrosome reaction and sion and animal fertility (Aurich et al. 2016;
sperm capacitation (Bezerra et al. 2019). The Verma et al. 2014). For this reason, the addition
NGF content in rabbit seminal plasma during of chelating or antioxidant agents is mandatory to
winter decreases four times compared with the improve and maintain sperm quality during the
other seasons (Casares-Crespo et al. 2018). preservation procedures and to avoid the deleteri-
Zhang et al. (2015) reported similar results in ous effects of ROS (Gadea et al. 2011; Kalthur
wild ground squirrels where the production of et al. 2011; Mercati et al. 2020).
NGF by testes decreased during the nonbreeding Previous studies demonstrated that addition of
season and increased in the breeding season. exogenous NGF to the incubation medium had
Interestingly, also in squirrel females, the ovarian significant effects on rabbit (Castellini et al.
NGF transcript was higher in the breeding than in 2019), bovine (Li et al. 2010a) and human
nonbreeding season (Maranesi et al. 2020). Such (Saeednia et al. 2016) sperm cell viability, and
19 Nerve Growth Factor (NGF) and Animal Reproduction 281

also exhibits trophic activity in the rescue of (Shi et al. 2006; Mirshokraei et al. 2013; Lobos
Sertoli cell viability mediated by the TrkA recep- et al. 2005; Bjorling et al. 2002).
tor protein (Chen et al. 1997). The rabbit uterus expresses NGF and its cog-
In seminal plasma, there was an abundance of nate receptors, NTRK1 as well as NGFR
NGF mRNA positively associated with the main- (Maranesi et al. 2016, 2018). In addition, NGF
tenance of post-thaw functional membrane integ- induces the synthesis and secretion of both
rity in bull spermatozoa, indicating that it could PGF2alpha and PGE2 by the uterus in vitro, via
be used to assess cryotolerance (Shilpa et al. selective modulation of nitric oxide synthase and
2017). In another study, there were greater PGE2–9-ketoreductase activities (Maranesi et al.
abundances of spermatozoa NGF mRNA in 2016).
bulls with greater quality semen (<25% Evidence documented in cattle suggests that
discarded ejaculates) compared with those with NGF may have a role as a luteotrophic factor in
lesser quality semen (>40% discarded spontaneous ovulators (Carrasco et al. 2016;
ejaculates); relative abundances of NGF mRNA Tribulo et al. 2015) and intramuscular injection
were also positively associated with pre-freeze at the time of artificial insemination of purified
mitochondrial membrane potential and post- ß-NGF from bovine seminal plasma increased
thaw sperm velocity variables in bulls (Parthipan pregnancy rate up to 16% (Stewart et al. 2018).
et al. 2017). In both normozoospermic and In induced ovulators in which OIF/NGF was
asthenozoospermic men, supplementation of first described (llamas and alpacas), an endocrine
freezing extender with NGF at 0.5 ng/mL mechanism was postulated to explain the ovula-
improved sperm viability and motility and tory response induced by deposition of fresh
decreased DNA fragmentation (Saeednia et al. homologous seminal plasma into the genital
2015, 2016). Consistently, treatment of frozen- tract (Ratto et al. 2012). According to this mecha-
thawed bull sperm cells with 40–80 ng/mL NGF nism, seminal plasma NGF is absorbed through
increased both leptin secretion and sperm mem- the genital tract and reaches the pituitary and/or
brane integrity (Li et al. 2010a). Based on all the the hypothalamus via the bloodstream to elicit the
previous findings, NGF may be an optimal candi- release of LH that, in turn, induces ovulation
date protein to enhance sperm cryopreservation (Adams et al. 2005). Indeed, it has been recently
techniques in domestic animals and men, a prop- reported that, in llamas, significant increases
osition that should be better analysed in the occur in circulating NGF levels 15 min after the
future. intrauterine deposition of seminal plasma,
followed by a LH surge 1 hour later (Berland
et al. 2016). More intriguingly, Berland et al.
19.3 Female (2016) using urethrostomized males, for the first
time, unequivocally demonstrated that copulation
19.3.1 NGF and Ovulation in Females alone did not induce ovulation in the absence of
seminal plasma, thus challenging the concept of
Recently, NGF and its cognate receptors, NTRK1 reflex ovulation in llamas.
and NGFR, as well as BDNF, were detected in the The observation that seminal plasma OIF
uterus of several spontaneous ovulatory species, elicited LH release from the pituitary in vitro
including rodents, chiropters, and farm animals and induced ovulation in various mammalian spe-
such as rat, guinea pig, mouse, bat, pig, and horse cies (Adams et al. 2016) further supported the
(Varol et al. 2000; Brauer et al. 2000; Wessels concept that the endocrine mechanisms involved
et al. 2014). The uterine expression of in spontaneous ovulation also included stimuli
the neurotrophin system varied greatly across derived from the male reproductive tract. More-
the oestrous cycle and during pregnancy under over, a study using recombinant human NGF in
the influence of oestrogen and progesterone alpacas confirmed a dose-related effect on ovula-
(P4) in golden hamsters, ewes, rat, and mouse tion rate (Stuart et al. 2015), similar to that
282 M. Maranesi et al.

obtained by exogenous GnRH administration ovulated, and again no preovulatory elevation in


(Silva et al. 2012). blood plasma LH was detected. In the same study,
In a recent work, Ratto et al. (2019) described mechanical stimulation of the vagina increased
the presence NGF immunoreactivity in tanycytes the ovulation rate by 50% in NGF-treated females
of the median eminence in llamas, where GnRH (García-García et al. 2017), suggesting that
nerve terminals are concentrated. In the same besides OIF/NGF, these females need physical
work, a greater relative abundance of kisspeptin stimulation to ovulate.
fibres was detected in association with GnRH Similarly, Maranesi et al. (2018) reported ovu-
fibres in the median eminence region. Moreover, lation rates of 67% (4/6) and 17% (1/6) in female
NGF may act through the hypothalamic rabbits inseminated with raw semen in
kisspeptin neurons to elicit the preovulatory LH non-anaesthetized and lumbar-anaesthetized
surge and ovulation and may be similarly impor- females, respectively; in all these females, ovula-
tant in the spontaneous and induced ovulators tion was preceded by an increase in plasma LH
(Inoue et al. 2011; El Allali et al. 2017). concentration during the 2 h after artificial insem-
Studies conducted in rabbits, however, have ination. Lumbar anaesthesia or administration of
yielded contradictory results. COX inhibitors before artificial insemination
Recently, several studies (Maranesi et al. blocked the increase in plasma LH concentration,
2015; García-García et al. 2017; Casares-Crespo attenuated the systemic blood rise of NGF, and
et al. 2018; Maranesi et al. 2018; Castellini et al. reduced the ovulation rate. These authors
2019) have confirmed the presence of NGF in postulated that other seminal plasma cytokines,
rabbit semen. This molecule was found at rela- besides NGF, might contribute to local stimula-
tively high concentrations in rabbit seminal tion of the female reproductive tract and ovulation
plasma, ranging from 1894  277 pg/ml to in the rabbit (Maranesi et al. 2018).
151.9  9.25μg/ml (Maranesi et al. 2015, 2018), Based on these findings, the same group pro-
closer to those in bovine seminal plasma posed a novel paracrine mechanism driven by raw
(100  30μg/mL), but much lower than those semen OIF, likely NGF, in the uterus/cervix,
reported in lama/alpaca 1.2  0.21 mg/ml which reinforces the neuroendocrine reflex pro-
(Bogle et al. 2018). voked by vaginal stimuli during natural mating
Despite these concentrations, Silva et al. (Fig. 19.1). According to such a mechanism,
(2011) did not detect ovulation in rabbits after (a) semen-derived NGF stimulates de novo syn-
intramuscular injections of rabbit seminal plasma thesis of NGF in the uterine wall; (b) both seminal
at different doses, although the same procedure NGF and uterine NGF are absorbed into the
induced ovulation in llamas. Conversely, bloodstream and act directly on the ovary rather
Cervantes et al. (2015) reported that the intramus- than on the pituitary and/or hypothalamus; and
cular injection of rabbit seminal plasma induced (c) semen-derived NGF and locally synthesized
ovulation in group-housed, but not in individually NGF stimulate uterine/cervix sensory neurons,
housed rabbits. An experiment conducted by which trigger GnRH neurons in the hypothala-
Rebollar et al. (2012) confirmed ovulation in mus. In sensory neuron stimulation (nervous
75% of rabbits after intravaginal deposition of pathway), prostaglandins and nitric oxide are
raw semen without treatment with a GnRH ana- synthesized by the uterus after binding of NGF
logue. No increase in blood plasma LH concen- to NTRK1 and/or NGFR receptors. NGF-induced
tration was observed. Quite surprisingly, prostaglandins would then stimulate directly or
however, there was no ovulation in those indirectly (via local chemical mediators) uterine/
inseminated with raw semen after lumbar intra- cervix neurons that reach, via spinal cord afferent
epidural anaesthesia. pathways, the hypothalamic centres responsible
A recent study (García-García et al. 2017) for the LH surge that induces ovulation. Addition-
reported that only one of the six female rabbits ally, NGF could directly stimulate uterine/cervix
treated intramuscularly with 24μg of murine NGF terminals of visceral afferent fibres through
19 Nerve Growth Factor (NGF) and Animal Reproduction 283

Fig. 19.1 Schematic representation of the suggested the circulating blood and/or act on sensory nervous
endocrine (left yellow area) and nervous (right green terminals. The question marks indicate putative target or
area) NGF-/OIF-mediated pathways involved in inducing way. GnRH gonadotropin-releasing hormone, LH
ovulation in rabbits. The inserts magnify NGF/OIF mech- luteinizing hormone, NGF nerve growth factor, NTRK1
anism at the uterine level: line thickness represents the neurotrophic tyrosine kinase receptor 1, OIF ovulation-
different NGF amounts of either exogenous (seminal inducing factor. Figure from Maranesi et al. (2018)
plasma) or endogenous (uterus/cervix) origin that enter
284 M. Maranesi et al.

binding to NTRK1 receptors and/or indirectly via There are increasing evidences that several
downstream activation of prostaglandin and/or components of the seminal plasma are involved
nitric oxide synthesis (Maranesi et al. 2016). in both male and female reproductive processes
This experimental evidence suggests that the (Maranesi et al., unpublished results). Besides
neuro reflex triggered by coitus and a seminal NGF, a well-known ovulation-inducing factor,
plasma male factor stimulating the endocrine other seminal plasma cytokines may stimulate
mechanism are both involved in inducing ovula- the female reproductive tract and induce ovula-
tion during natural mating or artificial insemina- tion, thus enhancing the reproductive success. A
tion in rabbits. better knowledge of these complex biological
Finally, it is worth recollecting here that NGF mechanisms could be useful to improve reproduc-
and its cognate receptors, NTRK1 and NGFR, tive performance of farm animals as well as
were immunolocalized in several rabbit ovarian humans.
structures, including corpus luteum (Zerani et al.
2021), granulosa and theca cells (Maranesi et al.
2018), similar to results previously reported for References
the ovary of Japanese Shiba goats (Ren et al.
2005) and other species (review: Linher-Melville Adams GP, Ratto MH, Huanca W, Singh J (2005)
Ovulation-inducing factor in the seminal plasma of
and Li 2013). The wide distribution of NGF
alpacas and llamas. Biol Reprod 73(3):452–457
receptors in ovarian structures provides the Adams GP, Ratto MH, Silva ME, Carrasco RA (2016)
grounds for a local paracrine and/or endocrine Ovulation-inducing factor (OIF/NGF) in seminal
action of NGF on the ovary. Indeed, Silva et al. plasma: a review and update. Reprod Dom Anim
51:4–17
(2011) reported an increase in the total number of
Aurich C, Schreiner B, Ille N, Alvarenga M, Scarlet D
antral and haemorrhagic anovulatory follicles, but (2016) Cytosine methylation of sperm DNA in horse
no ovulation, in rabbits treated with an intramus- semen after cryopreservation. Theriogenology 86
cular dose of rabbit seminal plasma compared to (5):1347–1352
Bakker J, Baum MJ (2000) Neuroendocrine regulation of
control those treated with PBS. Similar
GnRH release in induce ovulators. Front
haemorrhagic follicles were obtained with murine Neuroendocrinol 21:220–262
NGF (García-García et al. 2017) or after super- Berland MA, Ulloa-Leal C, Barría M, Wright H, Dissen
ovulation of those with eCG (García-Ximénez GA, Silva ME et al (2016) Seminal plasma induces
ovulation in llamas in the absence of a copulatory
and Vicente 1990).
stimulus: role of nerve growth factor as an ovulation-
inducing factor 1. Endocrinology 57(8):3224–3232
Bezerra MJB, Arruda-Alencar JM, Martins JAM, Viana
19.4 Conclusions AGA, Viana Neto AM, Rêgo JPA et al (2019) Major
seminal plasma proteome of rabbits and associations
with sperm quality. Theriogenology 2019
NGF and its cognate receptors are expressed, (128):156–166
although differently, in all male accessory sex Bjorling DE, Beckman M, Clayton MK, Wang ZY (2002)
glands, in the genital tract of several females in Modulation of nerve growth factor in peripheral organs
by estrogen and progesterone. Neuroscience
the hypothalamus, pituitary, and ovary of all spe-
110:155–167
cies insofar studied. These findings, together with Bogle OA, Carrasco RA, Ratto MH, Singh J, Adams GP
results from in vitro studies, indicate that the NGF (2018) Source and localization of ovulation-inducing
system can regulate male and female reproduction factor/nerve growth factor in male reproductive tissues
among mammalian species. Biol Reprod 99
with different mechanistic processes. In addition,
(6):1194–1204
the relatively high concentrations of NGF in the Brauer MM, Shockley KP, Chávez R, Richeri A,
seminal plasma in all species examined so far Cowen T, Crutcher KA (2000) The role of NGF in
suggest that this neurotrophin can exert direct pregnancy-induced degeneration and regeneration of
sympathetic nerves in the Guinea pig uterus. J Auton
and/or indirect effects at several levels of the
Nerv Syst 79:19–27
female reproductive axis acting on ovarian Carrasco R, Singh J, Adams GP (2016) The dynamics of
structures and/or on the release of gonadotropins. trkA expression in the bovine ovary are associated with
19 Nerve Growth Factor (NGF) and Animal Reproduction 285

a luteotrophic effect of ovulation inducing factor/nerve Hofmann H-D, Unsicker K (1982) The seminal vesicle of
growth factor (OIF/NGF). Reprod Biol Endocrinol the bull: a new and very rich source of nerve growth
14:47–57 factor. Eur J Biochem 128:421–426
Casares-Crespo L, Fernandez-Serrano P, Vicente JS, Inoue N, Sasagawa K, Ikai K, Sasaki Y, Tomikawa J,
Marco-Marco-Jimenez F, Viudes-Castro MP (2018) Oishi S et al (2011) Kisspeptin neurons mediate reflex
Rabbit seminal plasma proteome: the importance of ovulation in the musk shrew (Suncus murinus). Proc
the genetic origin. Anim Reprod Sci 189:30e42 Natl Acad Sci U S A 108(42):17527–17532
Castellini C, Mattioli S, Dal Bosco A, Collodel G, Jin W, Arai KY, Shimizu K, Kojima C, Itoh M, Watanabe
Pistilli A, Stabile AM et al (2019) In vitro effect of G et al (2006) Cellular localization of NGF and its
nerve growth factor on the main traits of rabbit sperm. receptors trkA and p75LNGFR in male reproductive
Reprod Biol Endocrinol 17(1):93 organs of the Japanese monkey, Macaca fuscata
Ceccanti M, De Nicolo S, Mancinelli R, Chaldakov G, fuscata. Endocrine 29(1):155–160
Carito V, Ceccanti M et al (2013) NGF and BDNF Jin W, Tanaka A, Watanabe G, Matsuda H, Taya K (2010)
long-term variations in the thyroid, testis and adrenal Effect of NGF on the motility and acrosome reaction of
glands of a mouse model of fetal alcohol spectrum golden hamster spermatozoa in vitro. J Reprod Dev
disorders. Ann Ist Super Sanita 49:383–390 56:437–443
Cervantes MP, Palomino JM, Adams GP (2015) In vivo Johnson D, Lanahan A, Buck CR, Sehgal A, Morgan C,
imaging in the rabbit as a model for the study of Mercer E et al (1986) Expression and structure of the
ovulation-inducing factors. Lab Anim 49:1–9 human NGF receptor. Cell 47:545–554
Chen BX, Yuen ZX, Pan G (1985) Semen-induced ovula- Johnston SD, O’Callaghan P, Nilsson K, Tzipori G,
tion in the Bactrian camel (Camelus bactrianus). Curlewis JD (2004) Semen-induced luteal phase and
Reproduction 73(2):335–339 identification of a LH surge in the koala (Phascolarctos
Chen Y, Dicou E, Djakiew D (1997) Characterization of cinereus). Reproduction 128:629–634
nerve growth factor precursor protein expression in rat Julio-Pieper M, Lara HE, Bravo JA, Romero C (2006)
round spermatids and the trophic effects of nerve Effects of nerve growth factor (NGF) on blood vessels
growth factor in the maintenance of Sertoli cell viabil- area and expression of the angiogenic factors VEGF
ity. Mol Cell Endocrinol 127(2):129–136 and TGFbeta1 in the rat ovary. Reprod Biol Endocrinol
Dechant G, Barde YA (1997) Signalling through the 4:57
neurotrophin receptor p75NTR. Curr Opin Neurobiol Kalthur G, Raj S, Thiyagarajan A, Kumar S, Kumar P,
7:413–418 Adiga SK (2011) Vitamin E supplementation in
Ehrhard PB, Erb P, Graumann U, Otten U (1993) Expres- semen-freezing medium improves the motility and
sion of nerve growth factor and nerve growth factor protects sperm from freeze-thaw-induced DNA dam-
receptor tyrosine kinase Trk in activated CD4-positive age. Fertil Steril 95(3):1149–1151
T cell clones. Proc Natl Acad Sci U S A Kaplan DR, Martin-Zanca D, Parada LF (1991) Tyrosine
90:10984–10988 phosphorylation and tyrosine kinase activity of the trk
El Allali K, El Bousmaki N, Ainani H, Simonneaux V protooncogene product induced by NGF. Nature
(2017) Effect of the camelid’s seminal plasma 350:158–160
ovulation-inducing factor/ß-NGF: a kisspeptin target Kershaw-Young CM, Druart X, Vaughan J, Maxwell WM
hypothesis. Front Vet Sci 4:99 (2012) ß-nerve growth factor is a major component of
Gadea J, Molla M, Selles E, Marco MA, Garcia-Vazquez alpaca seminal plasma and induces ovulation in female
FA, Gardon JC (2011) Reduced glutathione content in alpacas. Reprod Fertil Dev 24(8):1093–1097
human sperm is decreased after cryopreservation: Koshimoto C, Mazur P (2002) Effects of warming rate,
effect of the addition of reduced glutathione to the temperature, and antifreeze proteins on the survival of
freezing and thawing extenders. Cryobiology 62 mouse spermatozoa frozen at an optimal rate. Cryobi-
(1):40–46 ology 45(1):49–59
García-García RM, Masdeu M, Sánchez-Rodriguez A, Levanti MB, Germanà A, de Carlos F, Ciriaco E, Vega JA,
Millán P, Arias Alvarez M, Sakr OG et al (2017) Germanà G (2006) Effects of increased nerve growth
ß-nerve growth factor identification in male rabbit gen- factor plasma levels on the expression of TrkA and p75
ital tract and seminal plasma and its role in ovulation in rat testicles. J Anat 208:373–379
induction in rabbit does. Ital J Anim Sci 17:442–453 Levi-Montalcini R (1987) The nerve growth factor
García-Ximénez F, Vicente JS (1990) Effect of PMSG 35 years later. Science 237:1154–1162
treatment to mating interval in the superovulatory Levine E, Cupp AS, Skinner MK (2000) Role of
response of primiparous rabbits. J Appl Rabbit Res neurotropins in rat embryonic testis morphogenesis
13:71–73 (cord formation). Biol Reprod 62(1):132–142
Harper GP, Thoenen H (1980) The distribution of nerve Li C, Zhou X (2013) The potential roles of neurotrophins
growth factor in the male sex organs of mammals. J in male reproduction. Reproduction 145(4):R89–R95
Neurochem 34:893e903 Li C, Watanabe G, Weng Q, Jin W, Furuta C, Suzuki AK
Harper GP, Glanville RW, Thoenen H (1982) The purifi- et al (2005) Expression of nerve growth factor (NGF),
cation of nerve growth factor from bovine seminal and its receptors TrkA and p75 in the reproductive
plasma. J Biol Chem 275:8541e8 organs of the adult male rats. Zool Sci 22(8):933–937
286 M. Maranesi et al.

Li C, Sun Y, Yi K, Ma Y, Sun Y, Zhang W et al (2010a) TrkB and p75 in the uterus and oviduct of pregnant and
Detection of nerve growth factor (NGF) and its specific non-pregnant ewes. Res Vet Sci 95:164–168
receptor (TrkA) in ejaculated bovine sperm, and the Müller D, Davidoff MS, Bargheer O, Paust HJ, Pusch W,
effects of NGF on sperm function. Theriogenology 74 Koeva Y et al (2006) The expression of neurotrophins
(9):1615–1622 and their receptors in the prenatal and adult human
Li C, Zheng L, Wang C, Zhou X (2010b) Absence of nerve testis: evidence for functions in Leydig cells.
growth factor and comparison of tyrosine kinase recep- Histochem Cell Biol 126:199–211
tor a levels in mature spermatozoa from oligoastheno- Parthipan S, Selvaraju S, Somashekar L, Arangasamy A,
zoospermic, asthenozoospermic and fertile men. Clin Sivaram M, Ravindra JP (2017) Spermatozoal
Chim Acta 411(19–20):1482–1486. https://doi.org/10. transcripts expression levels are predictive of semen
1016/j.cca.2010.06.002. Epub 2010 Jun 11. PMID: quality and conception rate in bulls (Bos taurus).
20542022 Theriogenology 98:41–49
Lin K, Ding XF, Shi CG, Zeng D, QuZong S, Liu SH et al Perrard MH, Vigier M, Damestoy A, Chapat C,
(2015) Nerve growth factor promotes human sperm Silandre D, Rudkin BB et al (2007) Beta-nerve growth
motility in vitro by increasing the movement distance factor participates in an auto/paracrine pathway of
and the number of a grade spermatozoa. Andrologia 47 regulation of the meiotic differentiation of rat
(9):1041–1046 spermatocytes. J Cell Physiol 210:51e62
Linher-Melville K, Li J (2013) The roles of glial cell line- Ratto MH, Leduc YA, Valderrama XP, van Straaten KE,
derived neurotrophic factor, brain-derived Delbaere LT, Pierson RA et al (2012) The nerve of
neurotrophic factor and nerve growth factor during ovulation-inducing factor in semen. Proc Natl Acad Sci
the final stage of folliculogenesis: a focus on oocyte U S A 109:15042–15047
maturation. Reproduction 145(2):R43–R54 Ratto MH, Berland MA, Silva ME, Adams G (2019) New
Lobos E, Gebhardt C, Kluge A, Spanel-Borowski K insights of the role of β-NGF in the ovulation mecha-
(2005) Expression of nerve growth factor (NGF) nism of induced ovulating species. Reproduction 157
isoforms in the rat uterus during pregnancy: accumula- (5):R199–R207
tion of precursor proNGF. Endocrinology Rebollar PG, Dal Bosco A, Millan P, Cardinali R,
146:1922–1929 Brecchia G, Sylla L et al (2012) Ovulating induction
MacGrogan D, Desprès G, Romand R, Dicou E (1991) methods in rabbit does: the pituitary and ovarian
Expression of the beta-nerve growth factor gene in responses. Theriogenology 77:292–298
male sex organs of the mouse, rat, and Guinea pig. J Ren L, Medan MS, Weng Q, Jin W, Li C, Watanabe G et al
Neurosci Res 28(4):567–573 (2005) Immunolocalization of nerve growth factor
Maranesi M, Zerani M, Leonardi L, Pistilli A, Arruda- (NGF) and its receptors (TrkA and p75LNGFR) in
Alencar J, Stabile AM et al (2015) Gene expression the reproductive organs of shiba goats. J Reprod Dev
and localization of NGF and its cognate receptors 51:399–404
NTRK1 and NGFR in the sex organs of male rabbits. Ricci A, Graziano P, Bronzetti E, Saltini C,
Reprod Domest Anim 50(6):918–925 Sciacchitano S, Cherubini E et al (2007) Increased
Maranesi M, Parillo F, Leonardi L, Rebollar PG, pulmonary neurotrophin protein expression in idio-
Alonso B, Petrucci L et al (2016) Expression of nerve pathic interstitial pneumonias. Sarcoidosis Vasc Dif-
growth factor and its receptors in the uterus of rabbits: fuse Lung Dis 24(1):13–23
functional involvement in prostaglandin synthesis. Robinson LL, Townsend J, Anderson RA (2003) The
Domest Anim Endocrinol 56:20–28 human fetal testis is a site of expression of
Maranesi M, Petrucci L, Leonardi L, Piro F, Rebollar PG, neurotrophins and their receptors: regulation of the
Millán P et al (2018) New insights on a NGF-mediated germ cell and peritubular cell population. J Clin
pathway to induce ovulation in rabbits (Oryctolagus Endocrinol Metab 88:3943–3951
cuniculus). Biol Reprod 98(5):634–643 Saeednia S, Bahadoran H, Amidi F, Asadi MH, Naji M,
Maranesi M, Palermo FA, Bufalari A, Mercati F, Fallahi P et al (2015) Nerve growth factor in human
Paoloni D, Cocci P et al (2020) Seasonal expression semen: effect of nerve growth factor on the
of NGF and its cognate receptors in the ovaries of Grey normozoospermic men during cryopreservation pro-
squirrels (Sciurus carolinensis). Animals (Basel) cess. Iran J Basic Med Sci 18(3):292–299
10:1558 Saeednia S, Shabani Nashtaei M, Bahadoran H,
Mercati F, Domingo P, Pasquariello R, Dall'Aglio C, Di Aleyasin A, Amidi F et al (2016) Effect of nerve
Michele A, Forti K et al (2020) Effect of chelating and growth factor on sperm quality in asthenozoosprmic
antioxidant agents on morphology and DNA methyla- men during cryopreservation. Reprod Biol Endocrinol
tion in freeze-drying rabbit (Oryctolagus cuniculus) 14(1):29
spermatozoa. Reprod Domest Anim 55(1):29–37 Sanchez-Rodriguez A, Arias-Alvarez M, Timón P,
Milligan SR (1982) Induced ovulation in mammals. In: Bautista JM, Rebollar PG, Lorenzo PL et al (2019)
Finn CA (ed) Oxford review in reproductive biology. Characterization of β-nerve growth factor-TrkA system
Clarendon press, London, pp 1–46 in male reproductive tract of rabbit and the relationship
Mirshokraei P, Hassanpour H, Rahnama A, Foster WG between β-NGF and testosterone levels with seminal
(2013) Gene expression of BDNF and its receptors, quality during sexual maturation. Theriogenology
126:206–213
19 Nerve Growth Factor (NGF) and Animal Reproduction 287

Saruta J, Sato S, Tsukinoki K (2010) The role of Stewart JL, Mercadante VRG, Diaz NW, Canisso IF,
neurotrophins related to stress in saliva and salivary Yau P, Imai B et al (2018) Nerve growth factor-beta,
glands. Histol Histopathol 25:1317–1330 purified from bull seminal plasma, enhances corpus
Schecterson LC, Bothwell M (2010) Neurotrophin luteum formation and conceptus development in Bos
receptors: old friends with new partners. Dev taurus cows. Theriogenology 106:30–38
Neurobiol 70:332–338 Stuart CC, Vaughan JL, Kershaw-Young CM,
Schneidgenova M, Vašíček J, Cupka P, Chrenek P (2011) Wilkinson J, Bathgate R, de Graaf SP (2015) Effects
Is it necessary to control seasonal quality of the rabbit of varying doses of β-nerve growth factor on the timing
ejaculate? Slovak J Anim Sci 44:48–51 of ovulation, plasma progesterone concentration and
Seibel MM (1986) Luteinizing hormone and ovulation corpus luteum size in female alpacas (Vicugna pacos).
timing. J Reprod Med 31(8):754–759 Reprod Fertil Dev 27(8):1181–1186
Shi Z, Arai KY, Jin W, Weng Q, Watanabe G, Suzuki AK Thoenen H, Barde YA (1980) Physiology of nerve growth
et al (2006) Expression of nerve growth factor and its factor. Physiol Rev 60:1284–1335
receptors NTRK1 and TNFRSF1B is regulated by Tribulo P, Bogle O, Mapletoft RJ, Adams GP (2015)
estrogen and progesterone in the uteri of golden Bioactivity of ovulation inducing factor (or nerve
hamsters. Biol Reprod 74:850–856 growth factor) in bovine seminal plasma and its effects
Shi C-G, Lin K, Xu X-B, Zhang S-C, Wang N, Fan M on ovarian function in cattle. Theriogenology 83
(2012) Evidence for the involvement of NGF in human (9):1394–1401
sperm motility. J Biomed Sci Eng 5(9):534–541 Ulloa-Leal C, Bogle OA, Adams GP, Ratto MH (2014)
Shikata H, Utsumi N, Hiramatsu M, Minami N, Luteotrophic effect of ovulation-inducing factor/nerve
Nemoto N, Shikata T (1984) Immunohistochemical growth factor present in the seminal plasma of llamas.
localization of nerve growth factor and epidermal Theriogenology 81(8):1101–1107.e1
growth factor in Guinea pig prostate gland. Histochem- Varol FG, Duchemin AM, Neff NH, Hadjiconstantinou M
istry 80(4):411–413 (2000) Nerve growth factor (NGF) and NGF mRNA
Shilpa M, Selvaraju S, GirishKumar V, Parthipan S, change in rat uterus during pregnancy. Neurosci Lett
Binsila KB, Arangasamy A et al (2017) Novel insights 294:58–62
into the role of cell-free seminal mRNAs on semen Verma A, Rajput S, De S, Kumar R, Chakravarty AK,
quality and cryotolerance of spermatozoa in bulls Datta TK (2014) Genome-wide profiling of sperm
(Bos taurus). Reprod Fertil Dev 29(12):2446–2456 DNA methylation in relation to buffalo (Bubalus
Silva M, Niño A, Guerra M, Letelier C, Valderrama XP, bubalis) bull fertility. Theriogenology 82(5):750-9.e1
Adams GP et al (2011) Is an ovulation-inducing factor Wang H, Dong Y, Chen W, Hei J, Dong C (2011) Expres-
(OIF) present in the seminal plasma of rabbits? Anim sion and localization of nerve growth factor (NGF) in
Reprod Sci 127(3–4):213–221 the testis of alpaca (llama pacos). Folia Histochem
Silva ME, Recabarren MP, Recabarren SE, Adams GP, Cytobiol 49:55–61
Ratto MH (2012) Ovarian estradiol modulates the Wessels JM, Wu L, Leyland NA, Wang H, Foster WG
stimulatory effect of ovulation inducing factor (OIF) (2014) The brain uterus connection: brain derived
on pituitary LH secretion in llamas. Theriogenology neurotrophic factor (BDNF) and its receptor (Ntrk2)
77:1873–1882 are conserved in the mammalian uterus. PLoS One 9:
Silva M, Ulloa-Leal C, Norambuena C, Fernandez A, e94036
Adams GP, Ratto MH (2014) Ovulation-inducing fac- Yamamoto M, Sobue G, Yamamoto K, Terao S, Mitsuma
tor (OIF/NGF) from seminal plasma origin enhances T (1996) Expression of mRNAs neurotrophic factors
Corpus luteum function in llamas regardless the pre- (NGF, BDNF, NT-3 and GDNF) and their receptors
ovulatory follicle diameter. Anim Reprod Sci 148 (p75NGFR, NTRK1, TrkB, and Trkc) in the adult
(3–4):221–227 human peripheral nervous system and non-neural
Silva M, Ulloa-Leal C, Valderrama XP, Bogle OA, Adams tissues. Neurochem Res 21:929–938
GP, Ratto MH (2017) Nerve growth factor from semi- Zhang L, Wang H, Yang Y, Liu H, Zhang Q, Xiang Q et al
nal plasma origin (spβ-NGF) increases CL vasculari- (2013) NGF induces adult stem Leydig cells to prolif-
zation and level of mRNA expression of steroidogenic erate and differentiate during Leydig cell regeneration.
enzymes during the early stage of Corpus luteum Biochem Biophys Res Commun 436(2):300–305
development in llamas. Theriogenology 103:69–75 Zhang H, Wang Y, Zhang J, Wang L, Li Q, Sheng X et al
Snider WD (1994) Functions of the neurotrophins during (2015) Testicular expression of NGF, TrkA and p75
nervous system development: what the knockouts are during seasonal spermatogenesis of the wild ground
teaching us. Cell 77:627–638 squirrel (Citellus dauricus Brandt). Eur J Histochem 59
Squillacioti C, De Luca A, Paino S, Langella E, Mirabella (3):2522
N (2009) Effect of castration on the expression of the Zerani M, Polisca A, Boiti C, Maranesi M (2021) Current
NGF and NTRK1 in the vas deferens and accessory knowledge on the multifactorial regulation of corpora
male genital glands of the rat. Eur J of Histochem lutea lifespan: the rabbit model. Animals (Basel) 11
53:239–248 (2):296
Neurotrophins in the Brain of Teleost
Fish: The State of the Art 20
Paolo de Girolamo and Livia D’Angelo

Abstract multifaceted roles of neurotrophins in the


Neurotrophins are evolutionary well- brain of fish and other vertebrates.
conserved molecules, and fish constitute valu-
able vertebrate models to explore their pleio-
tropic role in the brain. In addition to an
introduction on the evolutionary importance 20.1 Introduction
of using fish in biomedicine and their neuro-
anatomy in comparison with mammals, here The discrete degree of molecular and functional
we review the available literature on the conservation of neurotrophins in teleostean fish
molecular evolution of neurotrophins and (Heinrich and Lum 2000) has attracted numerous
their receptors in teleost fish as well as their researchers to address studies on the related
role in the fish brain, from the early stages of conserved functional roles during the develop-
development until adulthood and aging. ment of the fish central nervous system (CNS)
Among neurotrophins, BDNF is the most as well as its maintenance in adult and aged brain.
well studied in the brain of teleost fish, and Molecular evolution studies of neurotrophins and
we report data on the functional involvement their receptors represent a clear example of stud-
of the BDNF/TrkB system in the development ies for analyzing coevolution processes of these
of the visual system and in the mechanisms of gene families and their correlation with the
adult brain regeneration. With the exception of increased complexity of the vertebrate nervous
neuroanatomical expression, much less is system. Fish share enough commonalities to jus-
known about the role of the other members tify conducting research relevant to humans,
of neurotrophin family in fish brain. We hope although fish diverged from humans more than
that this chapter opens new avenues leading to 400 million years ago. A vast amount of literature
a better understanding of the complex and has convincingly demonstrated the usefulness of
fish for improving our understanding of the
molecular and cellular mechanisms leading to
pathological conditions, and for the development
of new diagnostic and therapeutic tools (Schartl
2014); Aleström et al. 2020).
P. de Girolamo · L. D’Angelo (*)
Department of Veterinary Medicine and Animal
Production, University of Naples Federico II, Naples, Italy
e-mail: paolo.degirolamo@unina.it; livia.dangelo@unina.
it

# The Author(s), under exclusive license to Springer Nature Switzerland AG 2021 289
L. Calzà et al. (eds.), Recent Advances in NGF and Related Molecules, Cellular Neuroscience, Neural
Circuits and Systems Neuroscience 1331, https://doi.org/10.1007/978-3-030-74046-7_20
290 P. de Girolamo and L. D’Angelo

20.1.1 Evolutionary Aspects of Fish Evolutionary mutant models can be further


as Model Organisms categorized into two main classes:
in Biomedicine (a) evolutionary mutants with adaptive disease
phenotypes and (b) fish that are afflicted with
The most powerful and today used laboratory fish the same disease as humans, and thus develop a
species is zebrafish (Danio rerio), thanks also to true illness (Albertson et al. 2009; Schartl 2014).
the advanced and refined development of genetic Examples of models in the first category are
techniques for modeling human-acquired disease. represented by Antarctic icefish, mimicking
These freshwater fish provide a number of excep- “spontaneous” anemia-like disease and
tional advantages as laboratory organisms. First, osteopenia. The Antarctic icefish family is unique
they are small, can be bred and maintained in among vertebrates which do not express the
large numbers, and are at low cost (Aleström developmental program for erythrocyte forma-
et al. 2020). Second, they offer the opportunity tion. They do not make hemoglobin, nor they
to combine the analytical clarity of developmental produce erythrocytes, and therefore they model
biology with the power of genetics, and trans- harmful human blood diseases, including
genic lines can be easily and quickly obtained anemias, hemoglobinopathies, and thalassemias.
(Mann and Bhatia 2019). Furthermore, they Comparative genomic analysis with closely
offer the great advantage of high-throughput related species, i.e., zebrafish, which produce
approaches such as whole-genome mutagenesis hemoglobin and have erythrocytes, has led to
or chemical library drug screens (Cornet et al. the identification of candidate genes (Detrich
2018). Despite these undisputable advantages, and Yergeau 2004) also for human anemia. Fur-
the zebrafish as “genetically domesticated ther examples, among others, are represented
models” has also some limitations, particularly by the blind cavefish and the short-lived annual
in studies of pleiotropy, multigenic inheritance, killifish. The former evolved degenerated lens
variable penetrance, and so on. (Schartl 2014). and retinas, similar to humans with retinal degen-
Furthermore, the selective breeding of laboratory eration diseases (Jeffery 2009), the latter will be
animals makes very difficult or impossible to discussed more in detailed in this chapter.
observe and investigate the effect of the natural Examples of medical condition models include
environment on human model phenotypes those fish species that naturally develop the same
(Albertson et al. 2009). Indeed, natural selection disease in humans. Examples are the
is a potent generator of variation, and in this Xiphophorus which develops true malignant
perspective, other fish models are emerging as skin tumors from the naturally occurring large
“evolutionary mutant models” (Albertson et al. pigment spots (Fernandez and Bowser 2010)
2019). This concept was postulated on the and eels which spontaneously in the wild develop
evidences that in species or populations of many Wilms’ tumor, a malignant kidney tumor that
animal groups, including fish, the adaptation to a affects children at early age (Hu et al. 2011).
specific environment may result in an adaptive Beyond the opportunity of choosing between
phenotype, which resemble maladaptive human laboratory fish and evolutionary mutant model
diseases (Albertson et al. 2009). This phenome- according to the scientific need, when dealing
non constitutes a complementary approach to the with fish it should be taken into account their
genetic manipulation of a “domesticated” labora- evolutionary history, a key and relevant aspect.
tory organism, to discover genes and mechanisms Indeed, during evolution, teleost fish have
involved in human diseases. In addition, in this undergone a whole-genome duplication so that
sense, teleost fish, being the largest group of fish have two copies of many genes of which
vertebrates, offer great opportunities residing in tetrapods have only one (Meyer and Schartl
the biodiversity of the different environment and 1999; Postlethwait et al. 1998). Those duplicated
adaptive morphophysiological features. genes may be (1) orthologs, pairs of genes (one in
each of two different species) that evolved from a
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 291

single gene in the last common ancestor of those reaches the brain primarily via specialized cranial
two species, and/or (2) paralogs, genes that are nerves (trigeminus, facialis, vagus, and the three
related to each other by gene duplication events lateral line nerves), rather than through ascending
occurring within a lineage. fibers of the spinal cord (Kotrschal et al. 1998).
The duplication has resulted in altered gene The brainstem houses primarily the represen-
expression patterns or protein functions, such tation centers for all receiving somatosensory
that the complement of the expression domains stimuli, except for olfaction and vision. Somato-
or protein functions of both fish paralogs is equiv- sensory target areas are generally displaced dor-
alent to the single ortholog in other vertebrates. sally and organized in columns, whereas motor
For instance, if a mouse gene is expressed in the fibers originated from ventrally located centers. In
liver and kidney, one copy of the equivalent sep- certain fish species, i.e., Cyprinids, characterized
arate fish gene would be expressed in the liver, by a well-developed taste system made up of
and the second copy would be expressed in the internal and skin taste buds, the areas of the dorsal
kidney. This evolutionary process, which has column form prominent bulges: the facial lobe
been termed subfunction partitioning or emerges from the fourth ventricle as a singular
subfunctionalization (Force et al. 1999), has lobe, and the vagal lobe is a pair of dorso-caudal
far-reaching implications for functional studies ridges along the fourth ventricle receiving tactile/
of many genes. For example, when a mutation chemosensory nerves. In addition to several
in mouse inactivates a gene that has a role in early ascending and descending fiber systems, the
development in addition to an adult organ- brainstem houses the reticular formation, a ven-
specific function, it becomes impossible to study trally located network of neurons that is essential
the function of the adult organ in such mutants. In for the vital functions.
fish, both functions can be partitioned between The mesencephalic tegmentum and dienceph-
duplicates, making it possible to separate both alon house integrative systems for the brain
phenotypes. Thus, the teleost whole-genome structures arising from the cerebellum, optic tec-
duplication provides unique opportunities for tum, and forebrain. In a rostrocaudal direction,
studies on genes involved in disease development the tegmental third ventricle changes from a slit-
(Schartl 2014). like gap to a narrow channel before opening into
the fourth ventricle. Several structures develop as
extensions to this ventricle. The inferior lobes of
the hypothalamus are paired, constitute the
20.1.2 The Organization of the Brain
ventro-caudal part of diencephalon, and serve as
of Teleostean Fish
multimodal integration centers. As in all
vertebrates, the hypothalamus converts sensory
The brain of fish exhibits the typical arrangements
inputs into hormonal and behavioral responses.
of subdivisions typical of most vertebrates: the
The cerebellum is a discretely well-developed
spinal cord extends rostrally with the
structure, organized in a corpus and a valvula, the
(1) brainstem, including the tegmentum of mes-
latter appearing as a rostral extension beneath the
encephalon and diencephalon; (2) cerebellum and
optic tectum. Cerebellar structures are intimately
(3) pair of optic lobes which cap the mesenceph-
connected among each other and intervene in
alon; and (4) telencephalon consisting of paired
spatial orientation, proprioception, motor coordi-
hemispheres with olfactory bulbs in ventro-rostral
nation, and eye movement. The ventral cerebellar
position. The motor system mainly resides in the
surface area, the granular area, represents the
spinal cord, except for few a pathways
central acoustic area (Popper and Fay 1993). On
descending from the brain and some prominent
both sides of the corpus of cerebellum, there is a
brainstem reflexes, i.e., the Mauthner neuron sys-
parvocellular area called eminentia granularis,
tem for escape. The somatosensory information
292 P. de Girolamo and L. D’Angelo

which receives inputs from the inner ear and from telencephalon as the pallium, and the ventral
lateral line, whereas caudal and in continuation part as the subpallium (Nieuwenhuys and Meek
with the molecular layer of the corpus, there is the 1990). According to this theory, the rostral neural
crista cerebelli, predominantly involved in tube is thought to bend outward resulting in two
processing lateral line input (Davis and Northcutt telencephalic hemispheres separated by an
1983). unpaired ventricle and covered by a thin roof
The optic tectum consists of paired, dorsal plate, thus referred to as “everted” (Folgueira
hemispheres, separated from the tegmentum by et al. 2015). This eversion process results in an
the ventricular space, receives projections from unpaired ventricle and a different arrangement of
contralateral retinal ganglion cells, and the homologous of pallium. The pallium in
participates in the bidirectional communication teleosts has generally been subdivided into a
with the brainstem. Below the optic lobes, in the medial (Dm), dorsal (Dd), central (Dc), lateral
paramedian position, the torus longitudinalis (Dl), and posterior (Dp) part (Nieuwenhuys
extends into the subtectal ventricle as a pair of 2009). However, the precise and exact
longitudinal cylinders. Its presumed functions homologies with the pallium of tetrapods are
include postural control, detection of luminance quite debated, because several models of homol-
levels, and monitoring of saccadic movements ogy have been proposed based on connectional,
(Northmore et al. 1983). It has also a role as a neurochemical, gene expression, and functional
premotor center between the telencephalon and data (Ganz et al. 2012; Mueller 2011; Wullimann
the brainstem (Wullimann 1994). and Mueller 2004). More recently, a new pallial
The telencephalon arises from the rostral por- subdivision and related homology to pallial nuclei
tion of the embryonic neural tube forming two in tetrapods have been proposed, based on the
hemispheres, which present a T-shaped ventricle expression of conserved marker genes in
separating the two halves up to the dorsolateral zebrafish (Ganz et al. 2015). According to the
surface. Centrally, the two hemispheres are authors, Dm is homologous to the pallial amyg-
closely attached to each other and may even be dala in tetrapods, and the dorsal subdivision of Dl
fused. In addition to secondary olfactory is homologous to part of the hippocampal forma-
tract which terminates throughout the entire struc- tion in mouse (Ganz et al. 2015). The combina-
ture, virtually all sensory modalities project to its tion of ablation and behavioral experiments
dorsal portion through lemniscal pathways (Fin- documented that animals fed, grew, and behaved
ger 1983), and hypothalamic as well as primary normally in most respects, but exhibited signifi-
olfactory input is received at the ventral forebrain. cantly diminished rates of learning and were
The latter also contains the anterior commissure unable to perform more complex social tasks
with a peduncle of decussating tracts for a (Broglio et al. 2005; Rodriguez et al. 2002). An
two-way flow of information between the telen- analogous approach, based on molecular marker
cephalon and the diencephalon as well as within analysis, has been used to also identify homolo-
the telencephalon itself. gous areas of the subpallium, the ventral part of
The telencephalon has attracted the attention telencephalic hemispheres. The nuclei homolo-
of numerous neuroscientists about its develop- gous of striatum and pallidum are stretched
mental process, which has consequences on the along the rostrocaudal axis of the telencephalon,
neuromorphological organization of homologous also caudal to the anterior commissure (Ganz
telencephalic brain areas of tetrapods. More et al. 2012). Subpallial homologies have been
recently, developmental and gene expression identified as follows: a striatal domain in ventro-
studies have confirmed the theory of the “everted dorsal (Vd) and the dorsal part of ventro-ventral
telencephalon” (Nieuwenhuys 2009; Folgueira (Vv) and a striatal-septal domain (dorsal and ven-
et al. 2015), and identified the dorsal part of tral part of Vv) in the rostral telencephalon and
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 293

striatum (Vd, dorsal part of Vv), pallidum, and a A remarkable difference between fish and
pallidal-septal domain (ventral part of Vv) in the mammals is the absence of stellate astrocytes in
mid-telencephalon, caudal to anterior commis- the former, and it has been hypothesized that
sure (Ganz et al. 2012). radial glia in fish may serve some specialized
roles of astrocytes in mammals. For example,
zebrafish radial glia (1) express aquaporin-4,
20.1.3 Comparative Histological which is localized within the end feet of mamma-
Aspects Between the Brain lian astrocytes at the blood-brain barrier, although
of Teleostean Fish in fish the localization is not polarized (Grupp
and Mammals et al. 2010), and (2) express the glutamate trans-
porter Eaat2b, suggesting that radial glia in
The adult teleost brain resembles more to the zebrafish may serve functions related to
embryonic than to the adult mammalian brain. neurotransmitters, as in mammals (McKeown
Studies on neurogenic and regenerative capacity et al. 2012). Alternatively, some features in the
in the adult throughout the rostrocaudal axis of “reactive astrocyte” response of mammalian brain
the brain largely and accurately have upon injury are conserved in fish glia (Neve et al.
demonstrated this aspect. It has been suggested 2012) under similar experimental conditions. In
that the addition and/or replacement/turnover of the course of injury, also zebrafish glia
neuronal subtypes is necessary for expanding the overexpresses rat lipocalin-2, an autocrine medi-
existing sensory systems concomitantly with the ator of reactive astrocytosis in rodents, which
allometric growth and related increase in primary induces changes in the GFAP-expressing radial
sensory input (Kaslin et al. 2008). The great neu- glia (Lee et al. 2009).
rogenic ability of fish brain resides mainly in the
persistence of neuronal progenitors through the 20.1.3.2 Oligodendrocytes
entire life. The myelinating cells of the CNS of fish present
evolutionary conserved features common to all
20.1.3.1 Radial Glia vertebrates (Baraban et al. 2016). The mature
The neuronal progenitors have a radial glia mor- morphology of oligodendrocytes is plastic and
phology, and express radial glia markers, such as determined by the axonal environment. Indeed,
the glial fibrillary acidic protein (GFAP) (Lyons neuronal activity can affect distinct stages of oli-
et al. 2003). Cells expressing radial glia morphol- godendrocyte development and the process of
ogy and markers act as progenitors over the entire myelination. Increasing evidences show that
life. However, many radial progenitor cells, new myelin sheaths can be made on previously
which remain proliferative, do not express unmyelinated axons into adult life and there is a
GFAP as radial glia marker (März et al. 2010), certain degree of myelin sheet turnover through
but rather those of neuroepithelial cells, i.e., life (Lyons and Talbot 2014). Molecular signature
nestin (Mahler and Driever 2007). during development and differentiation of
Neural stem and progenitor cells are located oligodendrocytes, as well as in the myelination
within the brain in distinct neurogenic niches process, has been investigated, and
differing in their location, cellular composition, commonalities with mammals have been con-
and proliferative behavior. Proliferating radial firmed (Baraban et al. 2016). Altogether, these
glia, quiescent radial glia, and studies are pivotal to define therapeutic strategies
neuroepithelial-like cells (Lindsey et al. 2018) for the treatment of demyelinating diseases.
mainly characterize heterogeneity in the neural
stem and progenitor cell populations. This hetero- 20.1.3.3 Schwann Cells
geneity is hypothesized to reflect varying Developmental mechanisms regulating the cor-
capacities for neurogenesis, plasticity, and repair rect differentiation and myelination process have
between different neurogenic zones. been studied in zebrafish (Lyons and Talbot
294 P. de Girolamo and L. D’Angelo

2014). Precursors of Schwann cells, the neuromediator systems, including transmitters,


myelinating glia of peripheral nerves, derive receptors, transporters, and the enzymes required
form the neuronal crest, and migrate with grow- for synthesis and metabolism of these mediators
ing axons during nerve development. After (Stewart et al. 2014). Remarkably, the spatial and
migration, the process of radial sorting begins, temporal distribution of these systems has been
and Schwann cells associate with individual well characterized in both embryo/larva and adult
axons in the bundle of the developing nerve zebrafish for glutamate, γ-aminobutyric acid
(Jessen and Mirsky 2005; Raphael and Talbot (GABA), acetylcholine, and the aminergic
2011). As in mammals, in zebrafish Swann cells neurotransmitters dopamine (DA), noradrenaline
form myelin by wrapping their cell membranes (NA), serotonin (5-hydroxytryptamine (5-HT)),
around axons to generate a multilayered membra- and histamine (Stewart et al. 2015). Similar
nous sheath that insulates and supports the axons anatomical distribution of dopaminergic, norad-
(Lyons and Talbot 2014). Myelinating Schwann renergic, and cholinergic neurons has been
cells are postmitotic, but, in response to injury, reported in N. furzeri (Matsui et al. 2019; de
they lose contact with axons and undergo a pro- Girolamo et al. 2020). Neuroanatomical studies
cess of dedifferentiation (Glenn and Talbot 2013). have documented that these major neurotransmit-
ter systems are overall localized in homologous
20.1.3.4 Microglia brain areas of mammals, and drug treatment has
Microglia represent the tissue-resident macro- confirmed the conserved functions in the adult
phage population of the fish brain, alike in CNS (Stewart et al. 2014). The pattern of distri-
mammals. In zebrafish, a subpopulation of primi- bution of dopamine is slightly different when
tive macrophages from the yolk sac colonize the compared to homologous brain areas of zebrafish,
brain early during development from 48 h post- because the gene encoding th is duplicated, and
fertilization onward. Once in the brain, these th1 and th2 are differentially expressed
macrophages then rapidly differentiate into early (Yamamoto et al. 2010). Additionally, dopami-
microglia over the next 24 h post-fertilization nergic neurons have not been identified in the
(hpf) (Herbomel et al. 2001). At 3 and 4 days mesencephalon (Holzschuh et al. 2001) and ven-
post-fertilization (dpf), microglia are fully func- tral midbrain (Tay et al. 2011). Finally, it is
tional (Sieger et al. 2012), and share a significant important to consider that the fish-specific
number of expressed genes with their adult genome duplication resulted in expressing more
counterparts in zebrafish as well as with mouse neurotransmitter receptors than in other classes of
and human microglia (Mazzolini et al. 2019). vertebrates. For example, zebrafish have an
Mature microglia are present throughout the estimated 122 G-protein-coupled receptors for
parenchyma of the brain and spinal cord of fish biogenic amines (such as 5-HT, NA, DA, HA,
(Sieger and Peri 2013), with cell bodies largely adrenaline, and trace amines), compared with
stationary. They actively extend and retract their 57 in mouse and 44 in humans (Fredriksson and
processes in the presence of perturbation in the Schioth 2005).
environment, becoming activated (Svahn et al.
2013).
20.2 The Two Mainstream Fish
Models in Neurotrophin
20.1.4 Neuromediator Systems Research: Zebrafish
of Teleostean Fish and the African Turquoise
Killifish
Vertebrate neurochemistry is generally highly
conserved. Numerous and very detailed studies, The existence of functional NGF-related
mainly addressed on zebrafish, have neurotrophins in invertebrate lineages is
demonstrated fish possess all primary supported by studies in Drosophila and
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 295

C. elegans (Jaaro et al. 2001) as well as in scientists to investigate the regulatory


Lymnaea (Getz et al. 2020). Among nonmamma- mechanisms underlying the remarkable adult neu-
lian vertebrates, two mainstream model species rogenic and regenerative capacity throughout the
have been used to explore the role of rostrocaudal axis of the brain (Ganz and Brand
neurotrophins in the CNS: zebrafish and the 2016).
annual turquoise killifish, N. furzeri, both tropical N. furzeri, a clear example of evolutionary
freshwater organisms belonging to the class of mutant model, is characterized by a remarkable
Actinopterygii. While zebrafish is cyprinid, short lifespan (condensed in a few months), dur-
originated from ponds and standing water bodies, ing which it recapitulates typical hallmarks of
often connected to rice cultivation in India vertebrate aging (Harel et al. 2016). Interestingly,
(Parichy 2015), N. furzeri is a Cyprinodon- the short lifespan can be modulated by pharma-
tiformes living in the ephemeral pond, which cological, dietary, and lifestyle interventions
can be found during the monsoon season in (Cellerino et al. 2016). In the last few years, a
South-Eastern Africa (Mozambique and number of tools to work with this model have
Zimbabwe) (Reichard and Polačik 2019). The been developed. First, the genome has been
origin of their different habitat results in a strong sequenced and assembled (Valenzano et al.
evolutive adaptation, with different morphophy- 2015; Reichwald et al. 2015). Second,
siological features (D’Angelo et al. 2016a). transcriptomes for a number of tissues (Petzold
Zebrafish is the canonical fish model for study- et al. 2013; Baumgart et al. 2014) are available.
ing human diseases. It offers the advantage of Third, several inbred lines are housed under lab
using either the larval or the adult forms. At the condition (Hu and Brunet 2018). Fourth, genome-
larval stage, it has the unique benefit of the optical editing techniques are widely adopted to rapidly
clarity, allowing visualization of individual genes produce stable transgenic lines (Harel et al. 2016).
(fluorescently labeled or dyed) throughout the Therefore, this species, which emerged as excel-
developmental process using noninvasive imag- lent model for aging studies, is now broadly used
ing techniques. The embryonic transparency also in disparate research fields, such as evolutionary
helps in case of genetic manipulations. The genomics (Reichwald et al. 2015), regenerative
sequencing of genome has been completed medicine (Wendler et al. 2015; Wang et al.
(Howe et al. 2013), and numerous protocols for 2020a, b), developmental biology (Hu and Brunet
transgenesis, mutagenesis, and genome editing 2018), and ecotoxicology (Philippe et al. 2018).
are available (Albadri et al. 2017). Owing to the However, N. furzeri remains as dominant model
small size of the larvae, high-throughput screen- organism for aging studies, because of the unique
ing of neuroactive compounds can be easily so far described feature of a rapid as well as
performed (Saleem and Kannan 2018). Adult spontaneous aging process. The brain of
zebrafish have an integrated nervous system, N. furzeri has been studied with reference to its
which is proposed to contain homologous brain morphology (D’Angelo 2013). The major cellular
structures to those found in humans, as well as phenotypes observed during aging are (1) a dra-
equivalent cellular and synaptic structure and matic reduction of stem cell activity and
functions (Imperatore et al. 2018). Zebrafish dis- age-dependent reduction of adult neurogenesis
play (1) evolutionarily conserved nature of many (Tozzini et al. 2012); (2) glial hypertrophy
behaviors to resemble those of other species, (gliosis), visualized as overexpression of GFAP
(2) simple and relatively complex forms of (Tozzini et al. 2012); and (3) neuronal degenera-
learning, and good performance on cognitive tion, as measured by Fluoro-Jade B staining and
tasks dependent on short-term and long-term accumulation of lipofuscin (Terzibasi Tozzini
memory (Gerlai 2016). Furthermore, zebrafish, et al. 2012). At transcriptomic level, comparison
as all other teleosts, has attracted numerous with human brain datasets revealed conserved
296 P. de Girolamo and L. D’Angelo

upregulation of ribosome and lysosome, and expression suggests that subfunctionalization


complement activation and conserved and, to some degree, neo-functionalization have
downregulation of synapse, mitochondrion, occurred after the duplication (Dethleffsen et al.
proteasome, and spliceosome (Baumgart et al. 2003).
2014). Based on tyrosine kinase domain comparisons
of the zebrafish and mammalian receptors, one of
the fish genes was most related to TrkA, two to
TrkB, and the remaining two to TrkC. Consis-
20.2.1 Evolutionary Aspects
tently, the correspondence between the zebrafish
of Neurotrophins in Teleost Fish
and the mammalian receptors was maintained
when analyzing their extracellular domains,
Neurotrophins and their high-affinity Trk
except for the TrkA, whose extracellular domain
receptors have been found in each class of
is yet unknown (Martin et al. 1995). Similarly, in
vertebrates (Heinrich and Lum 2000). Accord-
fully sequenced genome of the pufferfish
ingly, neurotrophins evolved early in vertebrate
Tetraodon (Chiu et al. 2004) and N. furzeri
history during two rounds of genome duplication
(Reichwald et al. 2015), there are also five Trk
that characterized the origin and evolution of the
receptors, one of the A class, and two of each B
subphylum (Lundin et al. 2003; Hallböök et al.
and C classes. This situation correlates well with
2006). The phylogeny of Trk receptors is consis-
the doubling of the genome that occurred early
tent with this general pattern as well (Hallböök
during the evolution of teleost fishes (Benito-
1999). More in details, orthologs of NGF, BDNF,
Gutiérrez et al. 2006) and implies that one of the
NT-3, and NT-4/5 as well as of TrkA, TrkB, and
duplicated TrkA genes was lost early in the fish
TrkC are known in teleost fish (Götz et al. 1994;
lineage. Two paralogs of TrkB and of TrkC
Martin et al. 1995; Hallböök 1999; Dethleffsen
receptor have been identified in zebrafish (Martin
et al. 2003). A complete set of the “core” four
et al. 1995), pufferfish (Crollius 2006), and the
neurotrophins were originally retrieved in two
African turquoise killifish (N. furzeri) (Reichwald
pufferfish (Takifugu rubripes and Tetraodon
et al. 2015; Leggieri et al. 2019).
nigroviridis) and the zebrafish (Danio rerio)
Differently from Trk receptor, the nerve
genome assemblies. Recently, they were
growth factor receptor (Ngfr), alias p75 NT recep-
identified also in N. furzeri.
tor, represents a real “case study” in molecular
First phylogenetic analyses revealed the exis-
evolution. The teleost ngfr- and ngfr-related
tence of bony fishes’ neurotrophins not known in
genes have been discretely characterized mainly
other vertebrates: neurotrophin-6 (NT-6) in
during development (Brosamle and Halpern
platyfish (Xiphophorus maculatus and
2009; Han et al. 2014; Pinzón-Olejua et al.
Xiphophorus helleri) (Götz et al. 1994) and
2014), and to a less extent in adult animals
neurotrophin-7 (NT-7) in zebrafish (Nilsson
recapitulating some humanlike disease states,
et al. 1998) and carp (Cyprinus carpio) (Lai
such as anxiety behavior associated with type
et al. 1998). Identification of just one NT-6/NT-
2 diabetes (Wang et al. 2020a, b). From an
7 ortholog in the two pufferfish as well as in
evolutive perspective, the most recent theory,
zebrafish genome assemblies shows that NT-6
based on elegant synteny and phylogenetic stud-
and NT-7 are orthologs, and the gene should be
ies, supports that after the divergence of the rayfin
denoted NT-6/7 in analogy with the NT-4/5
and lobefin lineage, the whole-genome duplica-
nomenclature (Hallböök et al. 2006; Dethleffsen
tion resulted in duplication of both ngfr1 and
et al. 2003). The NT-6/7 is similar to NGF and is
ngfr2. This then gave rise to ngfr1a and ngfr1b,
phylogenetically distant from the human pseudo
which both persist in zebrafish, and to ngfr2a and
NT-6 genes, closely related to NT-4/5. NT-6/7
ngfr2b, the latter of which was lost in the teleost
and fish NGF bind and activate TrkA, and their
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 297

lineage (Catchen et al. 2011). However, in the (hpf). Their abundance relative to total transcripts
case of the African turquoise killifish, ngfr has increased sixfolds between 0 day and 2 dpf and
been annotated in single copy (de Girolamo et al. again onefold by 6 dpf (Hashimoto and Heinrich
2020). 1997). The expression of bdnf in the developing
brain seemed to be structure-specific (De Felice
et al. 2014): genetic markers (shh, pax2a, emx1,
20.2.2 Neurotrophins in the Brain krox20, lhx2b, and lhx9) were instrumental to
of Teleost Fish identify major topological domains of bdnf-posi-
tive cells in the pallium, hypothalamus, posterior
The transcription activity and phenotypic charac- tuberculum, and optic tectum (De Felice et al.
terization of neurotrophins have been investigated 2014; Sahu et al. 2019). However, the expression
in developing as well as mature neural of bdnf in the 5 days post fertilization (dpf)
phenotypes. The most well-investigated member embryos likely depended upon the strain, as it
of the neurotrophin family is BDNF and its was higher in the larvae of AB strain compared
related receptor, TrkB. However, data on tran- to larvae of the strain Tubingen long fin (van den
scriptional activity and phenotypic characteriza- Bos et al. 2017). De Felice et al. (2014) also
tion of all other related members are available identified the relative timing of BDNF transcrip-
either in the developing or in the adult brain of tion in the eye and optic tectum, hypothesizing an
fish. A summary of the brain localization of the involvement of BDNF in a mechanism for coor-
different neurotrophins in various fish species is dinated development of the retinotectal system. A
reported in Table 20.1, and an overview of locali- few years later, Hall and Tropepe (2018)
zation in the adult brain of N. furzeri is depicted in demonstrated the role of BDNF in a zebrafish
Fig. 20.1. model of in vivo visual experience-dependent
neuronal survival. The authors found that
restricting the visual experience, by exposing
20.2.3 bdnf/trkB embryo at dim light or treating larvae with a
glutamate antagonist, reduced BDNF expression
The gene encoding for bdnf in zebrafish is about in the periventricular gray zone. However,
90% identical to its mammalian counterpart supplementing larvae with exogenous BDNF
(Hashimoto and Heinrich 1997), and it is more from the early dim light treatment prevented
compact and complex in its exon organization subsequent neuronal loss in the following days,
and primary transcript processing. Five suggesting that adaptive neurogenic growth in the
promoters, and possibly as many as six, have zebrafish optic tectum exhibits a similar mecha-
been identified in zebrafish (Heinrich and nism of sensory input-mediated neuronal sur-
Pagtakhan 2004). Of the seven 50 noncoding vival, via neural activity-dependent BDNF
exons associated with promoters, two have been production (Hall and Tropepe 2018).
found only in the embryo before transcription In the larval (7 dpf) and adult stages, bdnf is
starts (they must be parts of maternally transmit- widely expressed throughout the whole brain of
ted transcripts); the other two, promoters 1b and zebrafish. It is mainly localized in the forebrain
1d, could not be detected at any stage or in any (dorsal telencephalon, preoptic area, dorsal thala-
adult organ. The four active promoters appeared mus, posterior tuberculum, hypothalamus,
differentially expressed in the adult organs synencephalon), optic tectum, cerebellum, and
(Heinrich and Pagtakhan 2004). At amino acid medulla oblongata (Gatta et al. 2016; Cacialli
levels, the degree of conservation between teleos- et al. 2016; Sahu et al. 2019), in regions that are
tean fish and mammals is very high (Lanave et al. homolog to bdnf-expressing areas in the mamma-
2007; Tettamanti et al. 2010). lian brain. Most remarkably, when investigating
In developing zebrafish, BDNF transcripts the cell phenotype expressing bdnf, transcripts
were first measurable 13 hours post fertilization were observed in mature neurons (MAP2 and
298 P. de Girolamo and L. D’Angelo

Table 20.1 Summary of the brain localization of the different neurotrophins and Trk receptors in Danio rerio,
N. furzeri, and Xiphophorus reported in embryonic and adult stages
Nothobranchius
Danio rerio furzeri Xiphophorus
Embryo Adult Adult Embryo Adult
bdnf mRNA Telencephalon/ Telencephalon/ Telencephalon/ N/A N/A
diencephalon diencephalon diencephalon
Optic tectum Optic tectum Optic tectum
Cerebellum and Cerebellum and Cerebellum and
medulla oblongata medulla oblongata medulla oblongata
Protein N/A Cerebellum Telencephalon/ N/A N/A
diencephalon
Mesencephalon/
optic tectum
Cerebellum and
medulla oblongata
trkB mRNA Telencephalon/ N/A N/A N/A
diencephalon
Optic tectum
Cerebellum and
medulla oblongata
Protein N/A Cerebellum Telencephalon/ N/A N/A
diencephalon
Cerebellum
ngf mRNA Optic tectum Telencephalon/ Telencephalon/ N/A N/A
diencephalon diencephalon
Optic tectum Mesencephalon/
optic tectum
Cerebellum and Cerebellum and
medulla oblongata medulla oblongata
Protein N/A Cerebellum Telencephalon/ N/A N/A
diencephalon
Mesencephalon/
optic tectum
Cerebellum and
medulla oblongata
nt-6 mRNA N/A N/A Telencephalon/ Cerebellum Telencephalon/
diencephalon diencephalon
Mesencephalon/ Mesencephalon/
optic tectum optic tectum
Cerebellum and Cerebellum and
medulla oblongata medulla oblongata
Protein N/A N/A N/A N/A N/A
trkA mRNA Hindbrain N/A Telencephalon/ N/A N/A
diencephalon
Cerebellum and
medulla oblongata
Protein N/A Cerebellum Telencephalon/ N/A N/A
diencephalon
nt-3 mRNA Telencephalon/ N/A N/A N/A N/A
diencephalon
Optic tectum
Cerebellum and
medulla oblongata
Protein N/A N/A N/A N/A
(continued)
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 299

Table 20.1 (continued)


Nothobranchius
Danio rerio furzeri Xiphophorus
Embryo Adult Adult Embryo Adult
trkC mRNA Diencephalon N/A N/A N/A N/A
Hindbrain
Protein N/A Cerebellum Telencephalon/ N/A N/A
diencephalon
nt-4 mRNA N/A N/A Telencephalon/ N/A N/A
diencephalon
Cerebellum and
medulla oblongata
Protein N/A N/A N/A N/A N/A
N/A: not available in literature

acetylated-tubulin markers) and never expressed specific role for this neurotrophic factor in
by radial glial cells (aromatase B marker) or controlling cell proliferation. Studies that are
proliferating cells (PCNA marker) (Cacialli et al. more recent have contributed to reinforce the
2016). Similar to zebrafish, the identification and hypothesis that bdnf/trkb system is highly
characterization of the cDNA fragment encoding involved in the neurogenic as well as reparative
the BDNF protein and the localization of BDNF abilities of adult fish brain (Lucini et al. 2018). An
mRNA and protein have been investigated in the experimental model of injured telencephalon in
whole brain of adult brain of N. furzeri (D’Angelo the adult zebrafish has documented that bdnf
et al. 2014b). For protein distribution, a sensitive levels increased significantly the first day post-
immunohistochemical technique, along with lesion in mature neurons around the lesion area,
highly specific affinity-purified antibodies to and then sharply decreased from the fourth day
BDNF, was employed. Both BDNF mRNA and post-lesion onward. The opposite regulation was
protein were detected in neurons and identifiable reported in the contralateral uninjured telence-
glial cells. Interestingly, both in zebrafish and phalic hemisphere: few mature neurons expressed
N. furzeri, BDNF was also localized in the brain bdnf 1 day post-lesion, and then they progres-
areas involved in adult neurogenesis, suggesting a sively and significantly increased until 15 days

Fig. 20.1 Schematic


localization of the different
neurotrophins and Trk
receptors in the adult brain
of N. furzeri
300 P. de Girolamo and L. D’Angelo

post-lesion (Cacialli et al. 2018). The expression trkB1 and trkB2 expression in the peripheral sym-
of bdnf, however, was not seen in proliferating pathetic ganglia at 5 dpf and, only at 6 dpf,
cells (PCNA), and notably the timing of expres- observed staining in the brain, with the isoform
sion peak of pcna occurred later than bdnf peak trkB2 more abundantly distributed unlike trkB1.
(Cacialli et al. 2018). Thus, trkB2 expression started early and
The role of bdnf in brain regenerative ability of corresponded to bdnf expression pattern,
zebrafish has been also indirectly demonstrated suggesting that trkB2 is the key receptor for
by using ANA-12, an antagonist of Trkb, the BDNF in the zebrafish brain (Sahu et al. 2019).
BDNF-specific tyrosine kinase receptor (Anand The central role of bdnf/trkb2 during develop-
and Mondal 2018). Intraperitoneally ment was also investigated by a double approach,
pre-treatment of ANA-12 before telencephalic including a mutant and morphant zebrafish model
stab injury reduced significantly the expression for trkB2, to unravel the function of this receptor
levels of PCNA over different telencephalic in the maintenance of the two major aminergic
areas (Anand and Mondal 2018). Sato et al. systems (serotonin and dopamine) (Sahu et al.
(2017) have proposed another evidence of 2019). In both experimental models, specific
involvement of bdnf/trkB system in brain regen- subsets of the dopaminergic and serotonergic
eration by using a different experimental model, neuronal populations reduced drastically with
the optic nerve crush. In this experimental para- consequences at behavioral level. Overexpression
digm, the proliferative neuroepithelial-like neural of the full-length trkB2 mRNA reversed the
stem cells in the optic tectum decreased or effect. Overall, these results confirmed that, also
increased proportionally to the projections of the in zebrafish, the appropriate development and
optic nerves from adult zebrafish retinas. maintenance of dopamine and serotonin neuronal
After crush, while bdnf expression decreased populations rely on the bdnf/trkB system (Sahu
in the optic tectum in the operation side, trkb was et al. 2019).
expressed in both sides after nerve crush (Sato
et al. 2017) without significant differences. Treat-
ment with 7,8-DHF, a Trkb agonist, increased cell 20.2.4 ngf and nt-6/trkA
proliferation in the mitotic region of the optic
tectum. This confirmed that the lesion-induced The genomic organization and transcript structure
decrease of neuroepithelial-like neural stem cell of ngf and nt-6 in the teleost zebrafish share a high
proliferation in the optic tectum was rescued by similarity with the mouse ngf and suggest that
TrkB signal activation (Sato et al. 2017). teleost NT-6 has evolved from a common ances-
Concerning trkB expression, the two paralogs tor after a single fish-specific duplication of NGF
(trkB1 and trkB2) were expressed very early in (Dethleffsen et al. 2003). Furthermore, NGF
the somitogenesis and brain development (Martin proteins in zebrafish and turquoise killifish were
et al. 1995; Nittoli et al. 2018). trkB1 occurred in reported in mature and two precursor forms
several neurons in the posterior telencephalon, (D’Angelo et al. 2014a; Gatta et al. 2016; Cacialli
hypothalamus, ventral thalamus, ventral tegmen- et al. 2019).
tum, ventral hindbrain, cranial ganglia, and spinal In the developing brain of zebrafish, first posi-
cord at 24 hpf (Nittoli et al. 2018). trkB2 was tive ngf neurons appeared at 24 hpf in the optic
expressed in the posterior telencephalon, ventro- tectum, and later the expression reduced and was
rostral thalamus, ventral tegmentum, and ventral manly observed in the pharyngeal arches (Nittoli
hindbrain at 24 hpf. At 96 hpf the expression et al. 2018). In the adult brain, both NGF mRNA
increased remarkably over the entire brain (Nittoli and protein were detected in the perikaryon of
et al. 2018). According to Nittoli and colleagues, mature neurons in all brain areas, including
the pattern of expression of trkB1 was quite Purkinje cells of the valvula of cerebellum
overlapping to bdnf, and both were co-expressed (Gatta et al. 2016), whereas only pro-NGF protein
in cranial nerves. Sahu et al. (2019) detected both was distributed in the neuronal projections
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 301

(Cacialli et al. 2019). The majority of in the forebrain of N. furzeri (D’Angelo et al.
NGF-positive cells were indeed mature neurons 2012). In addition, in the African turquoise killi-
(MAP2 positive). NGF was not distributed in fish, trkA appeared more expressed than ngfr in
radial glial cells (aromatase B cells) nor in young and old animals, without a significant
proliferating cells (PCNA marker), although age-dependent change. trkA and ngfr receptors
PCNA and aromatase B cells were closely were not expressed in cholinergic neurons, as
intermingled with NGF cells. Therefore, it widely demonstrated in mammals (de Girolamo
seemed that mature neurons in the zebrafish et al. 2020).
brain were expressing NGF mRNA and storing
pro-NGF (Cacialli et al. 2019).
Consistently with the observations in 20.2.5 nt-3/trkC
zebrafish, in the teleost fish, N. furzeri NGF was
morphologically detected in neurons widespread Phylogenetic analysis based on nucleotides and
throughout all brain regions. On the other hand, mature amino acid sequences of NT-3 confirmed
only some glial cells lining the mesencephalic that this neurotrophin evolved in teleost fish from
and rhombencephalic ventricles expressed NGF the common ancestor of NGF and NT-6. In
(D’Angelo et al. 2014a). zebrafish, the NT-3 protein was expressed as
The expression of fish-specific neurotrophin mature (Auer et al. 2015) and pro-NT-3 (Gatta
nt-6 appeared in the developing brain of zebrafish et al. 2016).
at 16 hpf in two clusters of cells adjacent to the nt-3 expression appeared at 12 hpf in
anterior and posterior of the inner ear primordium zebrafish, and was distributed in different devel-
and persisted until 48 hpf in some otic vesicle oping organs, including the sense organs. How-
patches. After, expression seemed to be lost ever, at 72 hpf a discrete bilateral group of
(Nittoli et al. 2018). In the brain of N. furzeri, positive neuronal cell types was observed in the
nt-6 expression persisted during adult as well as brain and cranial nerve region (Nittoli et al.
old stages. Remarkably, the expression levels 2018). It has been elegantly demonstrated that in
were comparable between young and old animals, larvae mutated for Kif5A, characterized by a dis-
and nt-6 was discretely expressed in mature ruption of presynaptic activity, there is an
neurons of all brain regions (Leggieri et al. upregulation of nt-3 in postsynaptic tectal cells.
2019). Similarly to ngf, the cellular phenotype This, in turn, promoted exuberant branching of
of nt-6 expression was represented by mature retinal axons by signaling through the TrkC
neurons (MAP2 marker) but not by developing receptor. The increased production of nt-3 was
neurons or glial cells (Leggieri et al. 2019). NT-6 also observed when neuronal activity was
mRNA was documented in the valvula of cere- blocked, for example, by toxins. The lack of
bellum during the developing stages of neuronal activity in retinal axons therefore
Xiphophorus, a phylogenetically close fish spe- seemed to increase the production of nt-3, which
cies, and the expression was confirmed in the then stimulated axonal branching (Auer et al.
fully adult brain (Götz et al. 1994). 2015). In the adult brain of zebrafish, NT-3 was
In spite of the teleost-specific duplication, the distributed in the majority of Purkinje cells of
genomes of fish contain only one trkA gene. Dur- valvular and body of cerebellum, less in the
ing development of zebrafish, the expression of molecular and granular layers (Gatta et al. 2016).
trkA appeared at 24 hpf in two domains of cranial The two paralogs of trkC, trkC1, and trkC2
nerve ganglia flanking the hindbrain, and in the displayed a different expression. trkC1 expres-
spinal cord (Nittoli et al. 2018). At 6 dpf, ntrkA sion started at 1 dpf and was localized in cells of
was expressed in the trigeminal ganglion and in telencephalon including also diencephalon, pitui-
the rostral hindbrain (Martin et al. 1995). In the tary gland, hindbrain, cranial ganglia, otic vesicle,
adult brain, the TrkA protein has been reported in and spinal cord (Martin et al. 1995; Auer et al.
the cerebellum of zebrafish (Gatta et al. 2016) and 2015; Nittoli et al. 2018). The localization in the
302 P. de Girolamo and L. D’Angelo

trigeminal ganglia is still debated (Pan et al. 2012; organization in fish. This is fundamental to
Nittoli et al. 2018). Between 48 and 96 hpf, trkC1 translate novel insights to mammals and helpful
was expressed diffusely in the brain (Nittoli et al. to elucidate complex and unknown mechanisms
2018). trkC2 had a more restricted expression at of the neurobiology of disease and aging.
24 hpf, specifically in a small group of neurons in 2. Neurotrophins are evolutionary well-
the telencephalon and midbrain, whereas in the conserved molecules, in terms of nucleotides
hindbrain it appeared at 48 hpf (Nittoli et al. and amino acid sequences. This is particularly
2018). In the adult brain of zebrafish, a faint true for BDNF.
immunoreactivity to TrkC protein has been 3. All neurotrophins and their specific receptors
reported in the molecular layer of cerebellum are expressed at the early stages of brain devel-
(Gatta et al. 2016), whereas in the adult brain of opment and in adult brain. There are
N. furzeri, TrkC positivity has been observed in differences in the spatiotemporal distribution
the most aboral region of the telencephalon, of all neurotrophins in zebrafish and N. furzeri.
magnocellular preoptic nucleus, and few neurons However, it seems that in both species, the cell
of hypothalamus (D’Angelo et al. 2012). phenotype expressing neurotrophins is
represented by mature neurons.
4. In zebrafish, the BDNF/TrkB system regulates
20.2.6 nt-4/5 the appropriate development of visual system
and participates to the regenerative ability of
nt-4/5 is the less studied neurotrophin in fish. It the brain after traumatic injury.
was expressed during zebrafish development More studies are needed to better explore the
(Auer et al. 2015; Nittoli et al. 2018), and the functional role also of other members of
first expression appeared during somitogenesis, neurotrophin family. Data available on NGF,
at 12 hpf. With the exception of a signal in the NT-3, NT-4/5, and NT-6 in the brain are mainly
cranial nerves at 24 hpf, this neurotrophin seemed limited to the neuroanatomical expression,
more localized in non-neuronal tissues during whereas very little is known about their functional
development (Nittoli et al. 2018). In the adult role. Future studies are thus mandatory to better
brain, nt-4/5 was studied only in N. furzeri, characterize their involvement during develop-
where it was observed in neurons and glial cells ment and neuronal maintenance in adult brain.
of the forebrain and hindbrain, and very scarcely The great variety of morphophysiological
in the midbrain (D’Angelo et al. 2016b). features of fish species combined to the growing
availability of genetic and imaging assays can
help to identify neural pathways and circuits
20.3 Conclusion and Future implicated in neurotrophin-related physiology
Perspectives and pathological states in vertebrates. Remark-
ably, fish offer the opportunity to dissect and
The last two decades have seen an increasing better comprehend the regulatory mechanisms of
interest in the use of teleost fish, mainly zebrafish neurotrophins in the brain during developmental
and the African turquoise killifish, as valuable and adult stages, as well as to conduct longitudi-
model organisms to explore the pleiotropic action nal studies over aging in short time (Baumgart
of neurotrophins in the CNS during development, et al. 2016). In addition, pharmacological and
adulthood, and aging. This is corroborated by environmental interventions can help to comple-
several observations: ment analyses and may have implications for
1. The CNS of fish and mammals displays translational neuroscience research and modeling
several homologies, despite some specific CNS disorders (Stewart et al. 2014).
morphological features and a less complex
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 303

References telencephalon of adult zebrafish. J Comp Neurol 526


(4):569–582
Cacialli P, Gatta C, D’Angelo L, Leggieri A, Palladino A,
Albadri S, Del Bene F, Revenu C (2017) Genome editing
de Girolamo P, Pellegrini E, Lucini C (2019) Nerve
using CRISPR/Cas9-based knock-in approaches in
growth factor is expressed and stored in central
zebrafish. Methods 121–122:77–85
neurons of adult zebrafish. J Anat 235(1):167–179
Albertson RC, Cresko W, Detrich HW 3rd, Postlethwait
Catchen JM, Braasch I, Postlethwait JH (2011) Conserved
JH (2009) Evolutionary mutant models for human
synteny and the zebrafish genome. Methods Cell Biol
disease. Trends Genet 25(2):74–81
104:259–285
Aleström P, D’Angelo L, Midtlyng PJ, Schorderet DF,
Cellerino A, Valenzano DR, Reichard M (2016) From the
Schulte-Merker S, Sohm F, Warner S (2020)
bush to the bench: the annual Nothobranchius fishes as
Zebrafish: Housing and husbandry recommendations.
a new model system in biology. Biol Rev 91:511–533
Lab Anim 54(3):213–224. https://doi.org/10.1177/
Chiu CH, Dewar K, Wagner GP, Takahashi K, Ruddle F,
0023677219869037
Ledje C, Bartsch P, Scemama JL, Stellwag E, Fried C,
Anand SK, Mondal AC (2018) TrkB receptor antagonism
Prohaska SJ, Stadler PF, Amemiya CT (2004) Bichir
inhibits stab injury induced proliferative response in
HoxA cluster sequence reveals surprising trends in
adult zebrafish (Danio rerio) brain. Neurosci Lett
ray-finned fish genomic evolution. Genome Res 14
672:28–33
(1):11–17
Auer TO, Xiao T, Bercier V, Gebhardt C, Duroure K,
Cornet C, Di Donato V, Terriente J (2018) Combining
Concordet JP, Wyart C, Suster M, Kawakami K,
zebrafish and CRISPR/Cas9: toward a more efficient
Wittbrodt J, Baier H, Del Bene F (2015) Deletion of
drug discovery pipeline. Front Pharmacol 9:703
a kinesin I motor unmasks a mechanism of homeostatic
Crollius HR (2006) The tetraodon genome. Genome Dyn
branching control by neurotrophin-3. elife 15:4. https://
2:154–164
doi.org/10.7554/eLife.0506
D’Angelo L (2013) Brain atlas of an emerging teleostean
Baraban M, Mensch S, Lyons DA (2016) Adaptive
model: Nothobranchius furzeri. Anat Rec (Hoboken)
myelination from fish to man. Brain Res 1641
296(4):681–691
(Pt A):149–161
D’Angelo L, de Girolamo P, Cellerino A, Tozzini ET,
Baumgart M, Groth M, Priebe S, Savino A, Testa G,
Castaldo L, Lucini C (2012) Neurotrophin Trk
Dix A, Ripa R, Spallotta F, Gaetano C, Ori M,
receptors in the brain of a teleost fish, Nothobranchius
Terzibasi Tozzini E, Guthke R, Platzer M, Cellerino
furzeri. Microsc Res Tech 75(1):81–88
A (2014) RNA-seq of the aging brain in the short-lived
D’Angelo L, Castaldo L, Cellerino A et al (2014a) Nerve
fish N. furzeri - conserved pathways and novel genes
growth factor in the adult brain of a teleostean model
associated with neurogenesis. Aging Cell 13:965–974
for aging research: Nothobranchius furzeri. Ann Anat
Baumgart M, Priebe S, Groth M, Hartmann N, Menzel U,
196:183–191
Pandolfini L, Koch P, Felder M, Ristow M, Englert C,
D’Angelo L, De Girolamo P, Lucini C, Terzibasi ET,
Guthke R, Platzer M, Cellerino A (2016) Longitudinal
Baumgart M, Castaldo L, Cellerino A (2014b) Brain-
RNA-Seq analysis of vertebrate aging identifies mito-
derived neurotrophic factor: mRNA expression and
chondrial complex I as a small-molecule-sensitive
protein distribution in the brain of the teleost
modifier of lifespan. Cell Syst 2(2):122–132
Nothobranchius furzeri. J Comp Neurol 522
Benito-Gutiérrez E, Garcia-Fernàndez J, Comella JX
(5):1004–1030
(2006) Origin and evolution of the Trk family of
D’Angelo L, Lossi L, Merighi A, de Girolamo P (2016a)
neurotrophic receptors. Mol Cell Neurosci 31
Anatomical features for the adequate choice of experi-
(2):179–192
mental animal models in biomedicine: I. Fishes Ann
Broglio C, Gomez A, Duran E et al (2005) Hallmarks of a
Anat 205:75–84
common forebrain vertebrate plan: specialized pallial
D’Angelo L, Avallone L, Cellerino A, de Girolamo P,
areas for spatial, temporal and emotional memory in
Paolucci M, Varricchio E, Lucini C (2016b)
actinopterygian fish. Brain Res Bull 66(4–6):277–281
Neurotrophin-4 in the brain of adult Nothobranchius
Brosamle C, Halpern ME (2009) Nogo-Nogo receptor
furzeri. Ann Anat 207:47–54
signalling in PNS axon outgrowth and pathfinding.
Davis RE, Northcutt RG (eds) (1983) Fish neurobiology,
Mol Cell Neurosci 40:401–409
vol II. The University of Michigan Press, Ann Arbor
Cacialli P, Gueguen MM, Coumailleau P, D’Angelo L,
De Felice E, Porreca I, Alleva E, De Girolamo P,
Kah O, Lucini C, Pellegrini E (2016) BDNF expres-
Ambrosino C, Ciriaco E, Germanà A, Sordino P
sion in larval and adult zebrafish brain: distribution and
(2014) Localization of BDNF expression in the devel-
cell identification. PLoS One 11(6):e0158057
oping brain of zebrafish. J Anat 224(5):564–574
Cacialli P, D’Angelo L, Kah O, Coumailleau P, Gueguen
de Girolamo P, Leggieri A, Palladino A, Lucini C,
MM, Pellegrini E, Lucini C (2018) Neuronal expres-
Attanasio C, D’Angelo L (2020) Cholinergic system
sion of brain derived neurotrophic factor in the injured
and NGF receptors: insights from the brain of the
304 P. de Girolamo and L. D’Angelo

short-lived fish Nothobranchius furzeri. Brain Sci 10 Grupp L, Wolburg H, Mack AF (2010) Astroglial
(6):394 structures in the zebrafish brain. J Comp Neurol
Dethleffsen K, Heinrich G, Lauth M, Knapik EW, Meyer 518:4277–4287
M (2003) Insert-containing neurotrophins in teleost Hall ZJ, Tropepe V (2018) Visual experience facilitates
fish and their relationship to nerve growth factor. Mol BDNF-dependent adaptive recruitment of new neurons
Cell Neurosci 24(2):380–394 in the postembryonic optic tectum. J Neurosci 38
Detrich HW, Yergeau DA (2004) Comparative genomics (8):2000–2014
in erythropoietic gene discovery: synergisms between Hallböök F (1999) Evolution of the vertebrate
the Antarctic icefishes and the zebrafish. Methods Cell neurotrophin and Trk receptor gene families. Curr
Biol 77:475–503 Opin Neurobiol 9:616–621
Fernandez AA, Bowser PR (2010) Selection for a domi- Hallböök F, Wilson K, Thorndyke M, Olinski RP (2006)
nant oncogene and large male size as a risk factor for Formation and evolution of the chordate neurotrophin
melanoma in the Xiphophorus animal model. Mol Ecol and trk receptor genes. Brain Behav Evol 68:133–144
19(15):3114–3123 Han HW, Chou CM, Chu CY, Cheng CH, Yang CH, Hung
Finger TE (1983) Organization of the teleost cerebellum. CC, Hwang PP, Lee SJ, Liao YF, Huang CJ (2014)
In: Davis RE, Northcutt RG (eds) Fish neurobiology, The Nogo-C2/Nogo receptor complex regulates the
vol I. The University of Michigan Press, Ann Arbor, pp morphogenesis of zebrafish lateral line primordium
261–284 through modulating the expression of dkk1b, a Wnt
Folgueira M, Bayley P, Navratilova P, Becker TS, Wilson signal inhibitor. PLoS One 9(1):e86345
SW, Clarke JD (2015) Morphogenesis underlying the Harel I, Valenzano DR, Brunet A (2016) Efficient genome
development of the everted teleost telencephalon. Neu- engineering approaches for the short-lived African tur-
ral Dev 10:22 quoise killifish. Nat Protoc 11:2010–2028
Force A, Lynch M, Pickett FB, Amores A, Yan YL, Hashimoto M, Heinrich G (1997) Brain-derived
Postlethwait J (1999) Preservation of duplicate genes neurotrophic factor gene expression in the developing
by complementary, degenerative mutations. Genetics zebrafish. Int J Dev Neurosci 15(8):983–997
151:1531–1545 Heinrich G, Lum T (2000) Fish neurotrophins and Trk
Fredriksson R, Schioth HB (2005) The repertoire of G- receptors. Int J Dev Neurosci 18(1):1–27
protein-coupled receptors in fully sequenced genomes. Heinrich G, Pagtakhan CJ (2004) Both 50 and 30 flanks
Mol Pharmacol 67:1414–1425 regulate zebrafish brain-derived neurotrophic factor
Ganz J, Brand M. 2016. Adult neurogenesis in fish. Cold gene expression. BMC Neurosci 5:19
Spring Harb Perspect Biol. 8(7). Pii: a019018 Herbomel P, Thisse B, Thisse C (2001) Zebrafish early
Ganz J, Kaslin J, Freudenreich D, Machate A, Geffarth M, macrophages colonize cephalic mesenchyme and
Brand M (2012) Subdivisions of the adult zebrafish developing brain, retina, and epidermis through a
subpallium by molecular marker analysis. J Comp M-CSF receptor-dependent invasive process. Dev
Neurol 520(3):633–655 Biol 238:274–288
Ganz J, Kroehne V, Freudenreich D, Machate A, Holzschuh J, Ryu S, Aberger F, Driever W (2001) Dopa-
Geffarth M, Braasch I, Kaslin J, Brand M (2015) mine transporter expression distinguishes dopaminer-
Subdivisions of the adult zebrafish pallium based on gic neurons from other catecholaminergic neurons in
molecular marker analysis. F1000Res 3:308 the developing zebrafish embryo. Mech Dev
Gatta C, Altamura G, Avallone L, Castaldo L, 101:237–243
Corteggio A, D’Angelo L, de Girolamo P, Lucini C Howe K, Clark MD, Torroja CF, Torrance J, Berthelot C,
(2016) Neurotrophins and their Trk-receptors in the Muffato M, Collins JE, Humphray S, McLaren K,
cerebellum of zebrafish. J Morphol 277(6):725–736 Matthews L, McLaren S, Sealy I, Caccamo M,
Gerlai R (2016) Learning and memory in zebrafish (Danio Churcher C, Scott C, Barrett JC, Koch R, Rauch GJ,
rerio). Methods Cell Biol 134:551–586 White S, Chow W, Kilian B, Quintais LT, Guerra-
Getz AM, Janes TA, Visser F, Zaidi W, Syed NI (2020) Assunção JA, Zhou Y, Gu Y, Yen J, Vogel JH,
Neurotrophic factors and target-specific retrograde sig- Eyre T, Redmond S, Banerjee R, Chi J, Fu B,
naling interactions define the specificity of classical Langley E, Maguire SF, Laird GK, Lloyd D,
and neuropeptide cotransmitter release at identified Kenyon E, Donaldson S, Sehra H, Almeida-King J,
Lymnaea synapses. Sci Rep 10(1):13526 Loveland J, Trevanion S, Jones M, Quail M, Willey D,
Glenn TD, Talbot WS (2013) Signals regulating Hunt A, Burton J, Sims S, McLay K, Plumb B, Davis J,
myelination in peripheral nerves and the Schwann Clee C, Oliver K, Clark R, Riddle C, Elliot D,
cell response to injury. Curr Opin Neurobiol Threadgold G, Harden G, Ware D, Begum S,
23:1041–1048 Mortimore B, Kerry G, Heath P, Phillimore B,
Götz R, Köster R, Winkler C, Raulf F, Lottspeich F, Tracey A, Corby N, Dunn M, Johnson C, Wood J,
Schartl M, Thoenen H (1994) Neurotrophin-6 is a Clark S, Pelan S, Griffiths G, Smith M, Glithero R,
new member of the nerve growth factor family. Nature Howden P, Barker N, Lloyd C, Stevens C, Harley J,
372(6503):266–269 Holt K, Panagiotidis G, Lovell J, Beasley H,
Henderson C, Gordon D, Auger K, Wright D,
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 305

Collins J, Raisen C, Dyer L, Leung K, Robertson L, Lanave C, Colangelo AM, Saccone C, Alberghina L
Ambridge K, Leongamornlert D, McGuire S, (2007) Molecular evolution of the neurotrophin family
Gilderthorp R, Griffiths C, Manthravadi D, Nichol S, members and their Trk receptors. Gene 394:1–12
Barker G, Whitehead S, Kay M, Brown J, Murnane C, Lee S, Park JY, Lee WH, Kim H, Park HC, Mori K, Suk K
Gray E, Humphries M, Sycamore N, Barker D, (2009) Lipocalin-2 is an autocrine mediator of reactive
Saunders D, Wallis J, Babbage A, Hammond S, astrocytosis. J Neurosci 29:234–249
Mashreghi-Mohammadi M, Barr L, Martin S, Leggieri A, Attanasio C, Palladino A, Cellerino A,
Wray P, Ellington A, Matthews N, Ellwood M, Lucini C, Paolucci M, Terzibasi Tozzini E, de
Woodmansey R, Clark G, Cooper J, Tromans A, Girolamo P, D’Angelo L (2019) Identification and
Grafham D, Skuce C, Pandian R, Andrews R, expression of Neurotrophin-6 in the brain of
Harrison E, Kimberley A, Garnett J, Fosker N, Nothobranchius furzeri: one more piece in
Hall R, Garner P, Kelly D, Bird C, Palmer S, Neurotrophin research. J Clin Med 8(5):Pii: E595
Gehring I, Berger A, Dooley CM, Ersan-Ürün Z, Lindsey BW, Hall ZJ, Heuzé A, Joly JS, Tropepe V,
Eser C, Geiger H, Geisler M, Karotki L, Kirn A, Kaslin J (2018) The role of neuro-epithelial-like and
Konantz J, Konantz M, Oberländer M, Rudolph- radial-glial stem and progenitor cells in development,
Geiger S, Teucke M, Lanz C, Raddatz G, plasticity, and repair. Prog Neurobiol 170:99–114
Osoegawa K, Zhu B, Rapp A, Widaa S, Langford C, Lucini C, D’Angelo L, Cacialli P, Palladino A, de
Yang F, Schuster SC, Carter NP, Harrow J, Ning Z, Girolamo P (2018) BDNF, brain, and regeneration:
Herrero J, Searle SM, Enright A, Geisler R, Plasterk insights from zebrafish. Int J Mol Sci 19(10):Pii:
RH, Lee C, Westerfield M, de Jong PJ, Zon LI, E3155
Postlethwait JH, Nüsslein-Volhard C, Hubbard TJ, Lundin LG, Larhammar D, Hallböök F (2003) Numerous
Roest Crollius H, Rogers J, Stemple DL (2013) The groups of chromosomal regional paralogies strongly
zebrafish reference genome sequence and its relation- indicate two genome doublings at the root of the
ship to the human genome. Nature 496(7446):498–503 vertebrates. J Struct Funct Genom 3:53–63
Hu CK, Brunet A (2018) The African turquoise killifish: a Lyons DA, Talbot WS (2014) Glial cell development and
research organism to study vertebrate aging and dia- function in zebrafish. Cold Spring Harb Perspect Biol 7
pause. Aging Cell 17:e12757 (2):a020586
Hu Q, Gao F, Tian W, Ruteshouser EC, Wang Y, Lazar A, Lyons DA, Guy AT, Clarke JD (2003) Monitoring neural
Stewart J, Strong LC, Behringer RR, Huff V (2011) pro-genitor fate through multiple rounds of division in
Wt1 ablation and Igf2 upregulation in mice result in an intact vertebrate brain. Development
Wilms tumors with elevated ERK1/2 phosphorylation. 130:3427–3436
J Clin Invest 121:174–183 Mahler J, Driever W (2007) Expression of the zebrafish
Imperatore R, D’Angelo L, Safari O, Motlagh HA, intermediate neurofilament nestin in the developing
Piscitelli F, de Girolamo P, Cristino L, Varricchio E, nervous system and in neural proliferation zones at
di Marzo V, Paolucci M (2018) Overlapping distribu- post-embryonic stages. BMC Dev Biol 7:89
tion of orexin and endocannabinoid receptors and their Mann A, Bhatia S (2019 Sep 16) Zebrafish: A powerful
functional interaction in the brain of adult zebrafish. model for understanding the functional relevance of
Front Neuroanat 12:62 noncoding region mutations in human genetic diseases.
Jaaro H, Beck G, Conticello SG, Fainzilber M (2001) Biomedicines 7(3):Pii: E71
Evolving better brains: a need for neurotrophins? Martin SC, Marazzi G, Sandell JH, Heinrich G (1995) Five
Trends Neurosci 24:79–85 Trk receptors in the zebrafish. Dev Biol 169
Jeffery WR (2009) Chapter 8. Evolution and development (2):745–758
in the cavefish Astyanax. Curr Top Dev Biol Matsui H, Kenmochi N, Namikawa K (2019) Age- and
86:191–221 α-Synuclein-dependent degeneration of dopamine and
Jessen KR, Mirsky R (2005) The origin and development noradrenaline neurons in the annual killifish
of glial cells in peripheral nerves. Nat Rev Neurosci Nothobranchius furzeri. Cell Rep 26(7):1727–1733.e6
6:671–682 Mazzolini J, Le Clerc S, Morisse G, Coulonges C, Kuil
Kaslin J, Ganz J, Brand M (2008) Proliferation, LE, van Ham TJ, Zagury JF, Sieger D (2019) Gene
neurogenesis and regeneration in the non-mammalian expression profiling reveals a conserved microglia sig-
vertebrate brain. Philos Trans R Soc Lond Ser B Biol nature in larval zebrafish. Glia 68(2):298–315
Sci 363:101–122 McKeown KA, Moreno R, Hall VL, Ribera AB, Downes
Kotrschal K, Van Staaden MJ, Huber R (1998) Fish GB (2012) Disruption of Eaat2b, a glutamate trans-
brains: evolution and environmental relationships. porter, results in abnormal motor behaviors in devel-
Fish Biol Fish 8:373–408 oping zebrafish. Dev Biol 362:162–171
Lai KO, Fu WY, Ip FC, Ip NY (1998) Cloning and Meyer A, Schartl M (1999) Gene and genome duplications
expression of a novel neurotrophin, NT-7, from carp. in vertebrates: the one-to-four (to-eight in fish) rule
Mol Cell Neurosci 11(1–2):64–76 and the evolution of novel gene functions. Curr Opin
Cell Biol 11:699–704
306 P. de Girolamo and L. D’Angelo

Mueller T (2011) The conserved bauplan of the teleostean Postlethwait JH, Yan Y-L, Gates M, Horne S, Amores A,
telencephalon. Brain Behav Evol 78(4):259–260 Brownlie A, Donovan A, Egan E, Force A, Gong Z,
März M, Chapouton P, Diotel N, Vaillant C, Hesl B, Goutel C, Fritz A, Kelsh R, Knapik E, Liao E, Paw B,
Takamiya M, Lam CS, Kah O, Bally-Cuif L, Strähle Ransom D, Singer A, Thomson M, Abduljabbar TS,
U (2010) Heterogeneity in progenitor cell subtypes in Yelick P, Beier D, Joly J-S, Larhammar D, Talbot WS
the ventricular zone of the zebrafish adult telencepha- et al (1998) Vertebrate genome evolution and the
lon. Glia 58:870–888 zebrafish gene map. Nat Genet 18:345–349
Neve LD, Savage AA, Koke JR, García DM (2012) Raphael AR, Talbot WS (2011) New insights into signal-
Activating transcription factor 3 and reactive astrocytes ing during myelination in zebrafish. Curr Top Dev Biol
following optic nerve injury in zebrafish. Comp 97:1–19
Biochem Physiol C Toxicol Pharmacol 155:213–218 Reichard M, Polačik M (2019). Nothobranchius furzeri, an
Nieuwenhuys R (2009) The structural organization of the ‘instant’ fish from an ephemeral habitat. Elife 8:Pii:
forebrain: a commentary on the papers presented at the e41548
20th annual Karger workshop ‘Forebrain evolution in Reichwald K, Petzold A, Koch P, Downie BR,
fishes’. Brain Behav Evol 74(1):77–85 Hartmann N, Pietsch S, Baumgart M, Chalopin D,
Nieuwenhuys R, Meek J (1990) The telencephalon of Felder M, Bens M, Sahm A, Szafranski K,
Actinopterygian fishes. In: Jones EG, Peters A (eds) Taudien S, Groth M, Arisi I, Weise A, Bhatt SS,
Comparative structure and evolution of the cerebral Sharma V, Kraus JM, Schmid F, Priebe S, Liehr T,
cortex. Plenum Press, New York, pp 31–73 Görlach M, Than ME, Hiller M, Kestler HA, Volff JN,
Nilsson AS, Fainzilber M, Falck P, Ibáñez CF (1998) Schartl M, Cellerino A, Englert C, Platzer M (2015)
Neurotrophin-7: a novel member of the neurotrophin Insights into sex chromosome evolution and aging
family from the zebrafish. FEBS Lett 424(3):285–290 from the genome of a short-lived fish. Cell 163
Nittoli V, Sepe RM, Coppola U, D’Agostino Y, De (6):1527–1538
Felice E, Palladino A, Vassalli QA, Locascio A, Rodriguez F, Lopez JC, Vargas JP et al (2002) Conserva-
Ristoratore F, Spagnuolo A, D’Aniello S, Sordino P tion of spatial memory function in the pallial forebrain
(2018) A comprehensive analysis of neurotrophins and of reptiles and ray-finned fishes. J Neurosci 22
neurotrophin tyrosine kinase receptors expression dur- (7):2894–2903
ing development of zebrafish. J Comp Neurol 526 Sahu MP, Pazos-Boubeta Y, Pajanoja C, Rozov S,
(6):1057–1072 Panula P, Castrén E (2019) Neurotrophin receptor
Northmore DPM, Williams B, Vanegas H (1983) The Ntrk2b function in the maintenance of dopamine and
teleostean torus longitudinalis: responses related to serotonin neurons in zebrafish. Sci Rep 9(1):2036
eye movements, visuotopic mapping and functional Saleem S, Kannan RR (2018) Zebrafish: an emerging real-
relations with the optic tectum. J Comp Physiol A time model system to study Alzheimer’s disease and
150:39–50 neurospecific drug discovery. Cell Death Discov 4:45
Pan YA, Choy M, Prober DA, Schier AF (2012) Robo2 Sato Y, Yano H, Shimizu Y, Tanaka H, Ohshima T (2017)
determines subtype-specific axonal projections of tri- Optic nerve input-dependent regulation of neural stem
geminal sensory neurons. Development 139 cell proliferation in the optic tectum of adult zebrafish.
(3):591–600 Dev Neurobiol 77(4):474–482
Parichy DM (2015) Advancing biology through a deeper Schartl M (2014) Beyond the zebrafish: diverse fish spe-
understanding of zebrafish ecology and evolution. elife cies for modeling human disease. Dis Model Mech 7
25:4 (2):181–192
Petzold A, Reichwald K, Groth M, Taudien S, Sieger D, Peri F (2013) Animal models for studying
Hartmann N, Priebe S, Shagin D, Englert C, Platzer microglia: the first, the popular, and the new. Glia
M (2013) The transcript catalogue of the short-lived 61:3–9
fish Nothobranchius furzeri provides insights into age- Sieger D, Moritz C, Ziegenhals T, Prykhozhij S, Peri F
dependent changes of mRNA levels. BMC Genomics (2012) Long range Ca2+ waves transmit brain-damage
14:185 signals to microglia. Dev Cell 22:1138–1148
Philippe C, Hautekiet P, Grégoir AF, Thoré ESJ, Stewart AM, Braubach O, Spitsbergen J, Gerlai R, Kalueff
Pinceel T, Stoks R, Brendonck L, Boeck G (2018) AV (2014) Zebrafish models for translational neurosci-
Combined effects of cadmium exposure and tempera- ence research: from tank to bedside. Trends Neurosci
ture on the annual killifish (Nothobranchius furzeri). 37:264–278
Environ Toxicol Chem 37:2361–2371 Stewart AM, Ullmann JF, Norton WH, Parker MO,
Pinzón-Olejua A, Welte C, Abdesselem H, Málaga- Brennan CH, Gerlai R, Kalueff AV (2015) Molecular
Trillo E, Stuermer CA (2014) Essential roles of psychiatry of zebrafish. Mol Psychiatry 20(1):2–17
zebrafish rtn4/Nogo paralogues in embryonic develop- Svahn AJ, Graeber MB, Ellett F, Lieschke GJ, Rinkwitz S,
ment. Neural Dev 9:8 Bennett MR, Becker TS (2013) Development of
Popper AN, Fay RR (1993) Sound detection and ramified microglia from early macrophages in the
processing by fish: critical review and major research zebrafish optic tectum. Dev Neurobiol 73:60–71
questions. Brain, Behav. Evolution 41:14–38
20 Neurotrophins in the Brain of Teleost Fish: The State of the Art 307

Tay TL, Ronneberger O, Ryu S, Nitschke R, Driever W Wang W, Hu CK, Zeng A, Alegre D, Hu D, Gotting K,
(2011) Comprehensive catecholaminergic projectome Ortega Granillo A, Wang Y, Robb S, Schnittker R,
analysis reveals single-neuron integration of zebrafish Zhang S, Alegre D, Li H, Ross E, Zhang N,
ascending and descending dopaminergic systems. Nat Brunet A, Sánchez AA (2020a) Changes in
ComTmun 2:171 regeneration-responsive enhancers shape regenerative
Tettamanti G, Cattaneo AG, Gornati R, de Eguileor M, capacities in vertebrates. Science 369(6508):eaaz3090
Bernardini G, Binelli G (2010) Phylogenesis of Wang J, Li Y, Lai K, Zhong Q, Demin KA, Kalueff AV,
brain-derived neurotrophic factor (BDNF) in Song C (2020b) High-glucose/high-cholesterol diet in
vertebrates. Gene 450(1–2):85–93 zebrafish evokes diabetic and affective pathogenesis:
Tozzini ET, Baumgart M, Battistoni G, Cellerino A (2012) the role of peripheral and central inflammation,
Adult neurogenesis in the short-lived teleost microglia and apoptosis. Prog Neuro-
Nothobranchius furzeri: localization of neurogenic Psychopharmacol Biol Psychiatry 96:109752
niches, molecular characterization and effects of Wendler S, Hartmann N, Hoppe B, Englert C (2015)
aging. Aging Cell 11(2):241–251 Age-dependent decline in fin regenerative capacity in
Valenzano DR, Benayoun BA, Singh PP, Zhang E, Etter the short-lived fish Nothobranchius furzeri. Aging Cell
PD, Hu CK, Clément-Ziza M, Willemsen D, Cui R, 14:857–866
Harel I, Machado BE, Yee MC, Sharp SC, Bustamante Wullimann MF (1994) The teleostean torus longitudinalis:
CD, Beyer A, Johnson EA, Brunet A (2015) The a short review on its structure, histochemistry, connec-
African turquoise killifish genome provides insights tivity, possible function and phylogeny. Europ J
into evolution and genetic architecture of lifespan. Morphol 32:235–242
Cell 163:1539–1554 Wullimann MF, Mueller T (2004) Teleostean and mam-
van den Bos R, Mes W, Galligani P, Heil A, Zethof J, malian forebrains contrasted: evidence from genes to
Flik G, Gorissen M (2017) Further characterisation of behavior. J Comp Neurol 475:143–162
differences between TL and AB zebrafish (Danio Yamamoto K, Ruuskanen JO, Wullimann MF, Vernier P
rerio): gene expression, physiology and behaviour at (2010) Two tyrosine hydroxylase genes in vertebrates.
day 5 of the larval stage. PLoS One 12(4):e0175420 New dopaminergic territories revealed in the zebrafish
brain. Mol Cell Neurosci 43:394–402

You might also like