Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

C H A P T E R

1
The Need to Study, Mimic, and Target Stem
Cell Niches
Ajaykumar Vishwakarma1,2, Jeroen Rouwkema1,2,3,
Peter Anthony Jones1,2,4, Jeffrey M. Karp1,2,4
1
Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, United States; 2Harvard-MIT Division of
Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, United States; 3University of
Twente, Enschede, The Netherlands; 4Harvard Stem Cell Institute, Cambridge, MA, United States

O U T L I N E

1. Introduction 3 3.3 Physical Cues and Matrix Mechanics 9


1.1 The Stem Cell Niche in Health and Disease 3 3.4 Oxygen and Metabolism 10
1.2 Components of Stem Cell Niche 4
3.5 Immune Cells, Inflammation, and
2. Biology of the Stem Cell Niche 5 Immunomodulation 10
2.1 Behavior of Stem Cells: Hierarchical Versus
Stochastic Model 5 4. Mimicking the Stem Cell Niche: Bioengineering
2.2 Embryonic and Adult Stem Cell Niches 6 Tools and Techniques 10
3. Biochemical and Biophysical Regulation of Stem 5. Bioengineering Specialized Artificial Stem Cell
Cell Behavior 7 Niches for Clinical Therapies 12
3.1 Extracellular Matrix and Biochemical Cues 7
3.2 Soluble Growth Factors and Ligands 9 References 12

1. INTRODUCTION
is composed of extracellular matrix (ECM) components
1.1 The Stem Cell Niche in Health and Disease for attachment/anchorage, diffusible biomolecules for
As opposed to single-celled organisms, cells in cell signaling, cell surface ligands for signal transduc-
complex multicellular organisms are associated with a tion, and essential cellecell interactions.
tissue-specific physiological environment. Different cell Studies of cell populations during embryonic devel-
types differ in morphology and function; yet, they are opment have led to the identification of stem cells that
genetically identical. This variation, caused by possess the capacity to produce a full organism from a
differential gene expression, is controlled by intrinsic fertilized egg.1 Stem cells are functionally defined as
mechanisms and by extrinsic signals from the local envi- undifferentiated embryonic or adult cells, which can
ronment, thereby controlling distinct cellular behavior, self-renew and generate differentiated cell types with
or “phenotype.” The local physiological microenviron- varying degrees of potency. The fundamental replicative
ment supporting the cell and driving extrinsic cues feature of stem cells, along with their generation of
from outside the cell is known as the “cell niche,” which differentiated progeny, accounts for the origin of the

Biology and Engineering of Stem Cell Niches


http://dx.doi.org/10.1016/B978-0-12-802734-9.00001-9 3 Copyright © 2017 Elsevier Inc. All rights reserved.
4 1. THE NEED TO STUDY, MIMIC, AND TARGET STEM CELL NICHES

word “stemness.” However, whether stem cells need a only dependent on factors promoting stemness but is
special environment that controls stem cell renewal, also a result of factors inhibiting differentiation path-
maintenance, and survival, and what is the nature of ways. Hence, in homeostasis, the underlying relation-
such microenvironment are pertinent questions many ship between stem cell and niche accommodates
researchers continue to explore. With growing evidence, nuances and involves various elements influencing the
there is a growing consensus that in vivo function and the stem cell functional parameters: replicative capacity
fate of stem and progenitor cells are regulated by the and potency. However, when tissue is injured or
interplay of various extrinsic signals of tissue-specific mi- diseased, the niche actively engages stem cells; guides
croenvironments, often referred to as “stem cell niches.” their proliferation, migration, and differentiation; and
The concept of a stem cell niche was first proposed by regulates their participation in tissue regeneration and
Schofield in the late 1970s as a physiologically restricted repair. Therefore, the niche should be regarded as a dy-
microenvironment that supports stem cells.2 The initial namic participant controlling stem cell number, fate, and
concept of anatomically distinct sites that regulate behavior in the health and disease of the tissue and the
hematopoietic stem cell (HSC) activity and self- organism.
renewal was later extended to acknowledge the discov-
ery of stem cells and their niches in multiple tissues.3
Stem cells are often linked with asymmetrical cell divi-
sion, and the niche maintains a stable number of stem
1.2 Components of Stem Cell Niche
cells during homeostasis, and removal of the niche in- The stem cell niche is a complex, heterotypic, and
duces differentiation. Extrinsic signals interact and inte- dynamic structure, which includes supporting ECM,
grate to ensure that one cell remains in the niche, while neighboring niche cells, secreted soluble signaling
another escapes it by receiving a differentiation signal. It factors (such as growth factors and cytokines), physical
is now clear that in high-turnover systems, such as in the parameters (such as shear stress, tissue stiffness, and topog-
gut and blood, the behavior of stem cells is not uni- raphy), and environmental signals (metabolites, hypoxia,
formly quiescent, and the various niche components inflammation, etc.) (Fig. 1.1).7,8 Stem cell niches are highly
may govern their relative proliferative activity.4e6 innervated and densely vascularized, thus are directly or
Also, it is emerging that stem cell performance is not indirectly influenced by vascular and neural inputs.

NICHE

Stem cell
Self-
renewal

Progenitor
cell
Differentiation

FIGURE 1.1 Components of stem cell niche. Adapted from Lane SW, Williams DA, Watt FM. Modulating the stem cell niche for tissue regeneration.
Nat Biotechnol 2014;32(8):795e803.

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


2. BIOLOGY OF THE STEM CELL NICHE 5
In addition to matrix and cell signaling elements differentiation potential and can differentiate into any
mentioned above, niche cells form functional units and all cells in an organism, plus the extraembryonic or
within the stem cell niche. These are neighboring tis- placental cells. Pluripotent stem cells can differentiate
sue-specific stem or somatic cell populations that into any cell within the three germ layers (endoderm,
interact with resident stem cells to regulate cell fate. mesoderm, and ectoderm). Embryonic stem cells (ESCs)
For example, mesenchymal stromal/stem cells in the are pluripotent and can divide and differentiate into cells
HSC niche or parenchymal hepatocytes in liver. In addi- of various types found in the body. Multipotent stem cells
tion to stem cell themselves, niche cells provide a source are progenitor cells that can differentiate into numerous
of physical and biochemical signals within the niche cell types but within a similar “family” or lineage. Lastly,
microenvironment by building extracellular matrix unipotent stem cells, the most restricted precursor, can
and producing cell surface or soluble signaling factors. only result in one cell fate. Unlike ESCs, stem cells from
Importantly therefore, stem cell microenvironments adult tissues are multipotent or unipotent.
are highly dynamic and display temporal variations. During development and in the healthy body, stem
Such variations in direct cellecell contacts and ECM cells can divide to produce new cells. This is a carefully
components, as well as their interaction with regulatory controlled process that allows the body to grow and to
molecules secreted by stem or niche cells and the spatial replace lost or damaged cells during adult life. For the
organization of niche components, ultimately enable the body to maintain homeostasis, stem cells proliferate
regulation of stem cells to render tissue homeostasis and before differentiating into a specific lineage, such that
regeneration.9 the generation of differentiated cells and the mainte-
nance of stem/progenitor pools are balanced. Two
2. BIOLOGY OF THE STEM CELL NICHE distinct models have been proposed to explain the line-
age choices of stem cells (Fig. 1.2). The hierarchical
model suggests a discrete arrangement of cells consist-
2.1 Behavior of Stem Cells: Hierarchical
ing of slow-cycling stem cells that can self-renew exten-
Versus Stochastic Model sively, which also give rise to short-lived transit
Understanding developmental biology is an important amplifying progenitor cells that then further differen-
approach to fully comprehend the structure and function tiate into committed nondividing cells. The stochastic
of the human body developed from a single totipotent model suggests that each stem cell chooses at random
stem cell, the zygote. The potency of a given cell to differ- between self-renewal and differentiation. In this model,
entiate into many specialized cells is defined by the de- each individual clone will vary in size.
gree of its plasticity and versatility at various stages. Recent lineage tracing studies have supported the
Totipotent stem cells are those with the greatest findings of the hierarchical model of stem cell behavior,

Hierarchical Model Stochastic Model


FIGURE 1.2 Hierarchial versus stochastic model for
behavior of stem cells.

Stem cell
Self
renewal

Transit
amplifying
cells

Differentiated cells

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


6 1. THE NEED TO STUDY, MIMIC, AND TARGET STEM CELL NICHES

yet also support that progenitors can behave in a sto- molecular signaling emanating from them. They discuss
chastic manner.10,11 Different types of stem cells at recent findings on how the niche network maintains
various body sites with distinct niches may work in different states of HSCs by providing multiple signaling
different ways, and it is possible that both these theories inputs from different cellular sources during homeosta-
are correct. In addition, based on recent discoveries, it sis and mobilization/homing.
has been proposed that the stem cell niche has a bicom- Compared with other somatic stem cell microenviron-
partmental organization12: one compartment that en- ments described since the late 1970’s, the microenviron-
gages in immediate, rapid new growth and one that ment of pluripotent cells had not been characterized
contributes later to long-term growth. In this way, until relatively recently. This is largely because of the tran-
stem cells might work cooperatively with their progeny sient nature of pluripotent cells in vivo and the character-
to sustain tissue regeneration. Although the precise ization of their regulation being restricted to in vitro
mechanisms mediating these phenotypic changes may methods. Also, pluripotent cells seem to be mutually
be complex, these lineage tracing studies demonstrate interacting with their neighboring cells, which are
that the niche is a critical component for the stem cells. dynamically changing in a short period of time relative
to somatic stem cells. Chapter 3 describes the microenvi-
ronment in early embryogenesis, which provides robust
information about cellular interactions required for the
2.2 Embryonic and Adult Stem Cell Niches acquisition of a pluripotent state. It also describes the
The first example of a stem cell niche was demon- transcriptional and functional heterogeneities observed
strated by Kimble and White in 1981, who showed that within established ESC cultures and their emerging sig-
the mesenchymal distal tip cell provides the essential nificance for self-renewal and maintenance of differentia-
microenvironment for the maintenance of the germ line tion potentials of ESCs.
stem cell in Caenorhabditis elegans.13 Later, Drosophila mela- Mesenchymal stem cells (MSCs) are multipotent stem
nogaster became the best characterized system to analyze cells found in stromal or connective tissue and are distinct
the relationship between stem cells and their niche from stem cells found in parenchymal or “functional” tis-
through studies in Drosophila germline stem cells.14 These sue. The MSC niche will not be discussed in this edition of
studies have provided an extensive understanding of the the book, since our understanding of what constitutes an
molecular basis of stem cell niche regulation. Meanwhile, MSC niche at different tissue sites is limited due to few
several niches of mammalian adult tissue stem cells were in vivo studies and heterogenous population variation of
discovered, for example, the HSC niche in bone marrow, MSCs in vitro. Further chapters in this section will discuss
hair follicle stem cells at the follicle bulge, and skin inter- stem cell niches in specific organs or organ systems.
follicular epidermis stem cells located in clusters at the In the adult mammalian brain, the two major stem
tops of rete ridges in the basal layer,15e17 among others. cell niches that support neurogenesis are the
This book covers in-depth many of the studied examples ventricularesubventricular zone that lies along the
of embryonic and adult stem cell niches, along with mo- lateral walls of the lateral ventricles and the dentate gy-
lecular mechanisms, in Section 1. rus subgranular zone found within the hippocampus.
Schofield’s 1978 mammalian stem cell niche hypothe- Chapter 6 focuses on the development of the brain’s
sis was based on the observation that colony-forming neuronal stem cell niches and further reviews key char-
stem cells were less capable of replacing blood cells acteristics and molecular regulators, including molecu-
compared with bone marrow cells when injected lar pathways that help support the mammalian brain’s
in vivo into irradiated animals.2 Since his discovery, two neurogenic stem cell niches.
the field of bone marrow HSCs has exploded. Schofield’s Stem cell niches play a critical role in cardiac homeo-
study postulated that HSCs reside in specific loci of bone stasis and myocardial repair after injury by replication
marrow and receive support from multiple cellular com- of a self-sustained pool of preexisting immature
ponents of their microenvironment. Since then, there myocytes. In the myocardium, cardiac progenitor cells
have been several studies that aimed at recognizing are clustered in interstitial microdomains with an archi-
such components and determining interactive signaling tectural organization distinct from the surrounding
mechanisms of the HSC niche.18e20 Chapters 4 and 5 nonestem cell tissue. The structural and molecular
comprehensively review how microenvironmental properties of this niche condition the response of the
cues from the bone marrow and intrinsic signals from tissue to physiologic and pathologic cues by creating a
the HSC dictate its fate to remain quiescent, become favorable environment for the interaction of stem cells
active, differentiate, migrate, or participate in regenera- with the surrounding cells and the interstitial space.
tion. These two chapters detail advances in genetically Chapter 7 emphasizes the relevance of cardiac progeni-
modified mouse models and high-resolution, real-time tor cells and their niches to cardiomyogenesis. Also, the
imaging to identify HSC niche components and the chapter discusses alternative stem celleindependent

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


3. BIOCHEMICAL AND BIOPHYSICAL REGULATION OF STEM CELL BEHAVIOR 7
mechanisms that may account for the global replenish- satellite cell activation, proliferation, and differentiation.
ment of lost cardiomyocytes. The chapter describes the cross talk between satellite
The adult intestinal epithelium represents one of cells and their niche along with regulation of satellite
the most suitable models to study tissue stem cells cell functions, namely commitment and self-renewal,
in vitro. It is a rapidly self-renewing tissue, and the in resting, injured, and pathologic muscle.
design of its basic unit, the cryptevillus, is highly Cancer stem cells (CSCs) reside in a microenviron-
stereotypical. Since the discovery of Lgr5þ stem cells ment that comprises various other cell types such as
at the base of intestinal crypts, it has become possible tumor-associated endothelial cells, mesenchymal cells,
to unveil the molecular mechanisms that control the and immune cells. In addition to these nonmalignant
homeostasis of these stem cells, including the extrinsic cells, ECM, metabolites, endocrine signals, waste
signaling cues from the crypt niche. Based on these products, and other secreted factors contribute to the
insights, in vitro propagation of murine and human proliferation, survival, and dissemination of tumor
intestinal stem cells in the form of ever-expanding cells.21 It is clear from recent reports that the niche plays
organoids has also become possible.20a Chapter 8 an important role in triggering stem cellelike programs
discusses such recent developments in the field of in subpopulations of cancer cells and at each different
the intestinal stem cell niche. Additionally, the chapter stage of tumorigenesis such as tumor initiation, progres-
focuses on a three-dimensional (3D) organoid culture sion, metastasis, and drug resistance. Chapter 11
system, originally established for murine small intes- reviews the current knowledge on the CSC niche and
tine that has now been adapted for other types of its contribution to tumorigenesis.
tissue stem cells by adding additional small molecules Understanding stem celleniche interactions requires
and/or growth factors. Organoids offer numerous the elucidation of a complex microenvironment with
possibilities for the study of basic research questions possibly hundreds of biochemical and biophysical cues
on tissue development and maintenance, stem cell acting in concert, perhaps synergistically, to control
characteristics, the detailed description of endoge- stem cell fate. The chapters in the following section focus
nous niche factors, gene function, etc. Furthermore, on key niche factors that regulate stem cell behavior.
in vitro organoids have the potential to contribute to
drug development and hold promise for regenerative
medicine, through the possibility to transplant these 3. BIOCHEMICAL AND BIOPHYSICAL
organoids back into their original organs. REGULATION OF STEM CELL BEHAVIOR
Skin is a primary protective barrier and is being
constantly renewed. As such, skin stem cells play a 3.1 Extracellular Matrix and Biochemical Cues
key role in maintaining epithelial homeostasis, similar
to the intestinal epithelium. Likewise, Lgr5þ cells in ECM is a natural substrate manufactured by the cells
hair follicles has been show to comprise an actively themselves providing multiple biochemical and biophys-
proliferating and multipotent stem cell population.20b ical cues to stem cells residing on or in it, thus maintaining
Chapter 9 describes the location and cellular hierarchy stem cell pools. It is a mixture of long biopolymers con-
of each epithelial stem cell of the skin within the sisting of proteins (e.g., collagen, fibronectin, vitronectin,
epidermis, hair follicle, sebaceous gland, and sweat elastin, and laminin) and glycosaminoglycans (e.g., hepa-
gland and highlights the intrinsic regulators, which rin, chondroitin sulfate, keratin sulfate, and hyaluronic
maintain their stemness. The chapter reviews the acid). ECM was once considered an inert support struc-
extrinsic regulators of epithelial stem cell function and ture, but research has revealed it to be a signaling core,
sheds light on recent findings that introduce new actors with a critical role in the niche for developing and
in the epithelial stem cell niche in the skin. Also, it dis- maturing stem cells. Matrix provides the necessary chem-
cusses how the niche components in the skin may vary ical and mechanical signals to modulate cellular processes
depending on the body location. such as cell renewal, morphogenesis, differentiation,
In the skeletal muscle, a specialized population repair, and homeostasis. Stem cells bind to matrix adhe-
known as satellite stem cells contribute to its regenera- sion ligands by integrins and other receptors localized
tive capacity. Owing to their ability to generate both in their plasma membrane. Integrins are heterodimeric
stem cells and committed myogenic progenitors, satel- proteins consisting of a form and b form, and the combi-
lite stem cells allow self-renewal of the satellite cell nation of their subtypes, such as a1b1, specifies the type of
reservoir and provide myogenic progenitor cells to ECM proteins they bind (collagen or laminin in case of
repair the muscle tissue. Chapter 10 highlights the indis- a1b1, for instance) and provides intracellular signals,
pensable role of the satellite cell niche in the regulation leading to cellular responses.22 In addition to the effect
of the stem cell functions. This niche maintains the mus- of cellematrix adhesion on directing the behavior of cells,
cle stem cell in a quiescent state; however, in response to cells are also able to degrade and remodel matrix by
muscle injury, the niche actively generates signals for providing enzymes into their surroundings, such as

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


8 1. THE NEED TO STUDY, MIMIC, AND TARGET STEM CELL NICHES

metalloproteinases.23 Thus, the communication between the different niches are indicated. Looking closely at
cells and matrix is bidirectional and dynamic. the matrix, it is possible to identify the major structural
Furthermore, matrix is able to provide biochemical cues and chemical entities as patterns and combinations of
to cells effectively, because it sequesters various bio- functional groups, present in proteins, proteoglycans,
macromolecules, such as growth factors, through specific and peptide sequences. To date, many studies have
and nonspecific bindings, and makes residing cells more examined the relationship between stem cells and
accessible to these molecules.24 Cells recognize and matrix in vitro using biomaterial scaffolds that mimic
respond to secreted signaling factors through multiple the natural ECM.26 In a particularly interesting
pathways: for instance, cells express receptors to their ag- approach, researchers applied printing techniques using
onists in their microenvironment and initiate intracellular automatic pipettes or robots to generate a library of arti-
signal transduction once binding between a receptor and ficial niches in a single experiment.27 Another group
an agonist is established. created microarrays of artificial niche components
The chemical composition of the ECM can vary comprising ECM proteins for probing single stem cell
widely among tissues. Fig. 1.3 shows four different fates in high throughput.28 These studies clearly demon-
stem cell niches, together with their cellular and ECM strated a robust proof-of-concept that adding known
components. ECM molecules playing major roles in ECM molecules or functional groups to synthetic

FIGURE 1.3 Extracellular matrix composition of four different stem cell niches. Adapted from Gattazzo F, Urciuolo A, Bonaldo P. Extracellular
matrix: a dynamic microenvironment for stem cell niche. Biochim Biophys Acta (BBA) 2014;1840:2506e19.

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


3. BIOCHEMICAL AND BIOPHYSICAL REGULATION OF STEM CELL BEHAVIOR 9
constructs enables the development of controlled envi- control of the spatiotemporal presentation of soluble
ronments regulating stem cell behavior. It is critical to signaling molecules and insoluble, matrix-bound cues
further understand such relationships to fully appre- is essential for effective stem cell bioengineering
ciate the effect of chemical functional groups in the schemes and critically depends on the development of
stem cell niche on cell behavior. The small molecular advanced, multi-biofunctional biomaterials. Chapter
patterns found throughout the body can have a 14 summarizes biomaterials-based approaches for the
significant influence on stem cell behavior when found spatial and temporal control of factor and ligand presen-
in high numbers within a niche. Studying each unit in tation. The chapter focuses on the presentation of
isolation helps to delineate their individual effects on growth factors from versatile 3D polymeric matrix plat-
the stem cells. Chapter 13 discusses some of the chemi- forms for the biomimetic presentation of multiple mo-
cal functional groups that have been extensively investi- lecular effectors and for the adjustment of physical
gated to understand their role in modulating stem cell constraints.
behavior. In addition to matrix in the extracellular sur-
rounding, niches may consist of cells, either a single 3.3 Physical Cues and Matrix Mechanics
type or a range of interacting multiple cell types. Celle
cell communication can be mediated by interactions It is now well understood that not only the biochem-
via direct contact between cells carrying distinct func- ical properties of the ECM but also the biophysical
tional group, such as cadherin-based adherens junctions properties of the microenvironmentdstiffness and elas-
in epithelial cells and endothelial cells, affecting their ticity, electrostatic charges, wettability, and structural
behavior. The prospect of replicating the complex ECM informationdregulate cellular functions. One of the
chemistry using engineered natural or synthetic biophysical properties that has been intensively studied
biomaterial substrates would be highly valuable, not is the stiffness of the microenvironment. A variety of
only to understand complex synergies guiding stem cells recognize the stiffness of their surroundings and
cell behavior but also to develop novel translational respond to this. For example, MSCs become quiescent
technologies, such as regenerative scaffolds. on polyacrylamide gels with a stiffness of approximately
200 Pa, which is comparable with the stiffness of bone
marrow.29 Myocytes and cardiomyocytes cultured
3.2 Soluble Growth Factors and Ligands ex vivo exhibit striation, when they contact polyacryl-
Niches contain secreted or cell surface factors that can amide gels of which the stiffness mimics the stiffness
be secreted by stem cells themselves or by other cells of skeletal muscle or heart tissue, respectively.30,31 The
that reside in the niche. Cellular communication lineage of MSC differentiation is also affected by the
employs different sets of secreted soluble hormones, stiffness of the microenvironment: MSCs are able to
growth factors, and cytokines to regulate cellular differentiate into adipocytes most effectively on approx-
functions, including proliferation, differentiation, or imately 200-Pa polyacrylamide gels, which is the
migration. A few key biomolecules that control stem stiffness of adipose tissues,29 whereas osteogenic differ-
cell renewal, maintenance, or survival include Notch, entiation becomes more effective as the stiffness of
Wnt, fibroblast growth factor, epidermal growth factor, polyacrylamide gels increases.32,33 Thus, mimicking
transforming growth factor-b, stem cell factor, and the stiffness of the microenvironment, represented by
chemokine families. the stiffness of tissues in many cases, may be a prerequi-
A vast majority of the currently applied methods for site for stem cells to reenact in vivo cellular functions.
ex vivo stem cell expansion, maintenance, and differen- Along with stiffness, diverse substrate patterns gener-
tiation rely on the addition of soluble microenviron- ated using innovative fabrication processes have also
mental components, namely cytokines and growth been shown to alter stem cell behavior by applying
factors. The action of signaling molecules is highly mechanical constraints on cells. They can be regulated
interconnected and activation steps downstream of by careful design of the microscale and nanoscale
receptor binding can depend on the mode of factor features of the substrate geometry. Chapter 15 focuses
presentation. Hence, it is important to consider their on the recent advances in exploitation of mechanical
pleiotropic actions in the design of engineered stem stimulations to differentiate stem cells. It also discusses
cell microenvironments. Contrary to the conditions of several mechanisms that underlie the stem cell’s
standard in vitro cell culture, growth factors do not response to mechanical stimuli, including changes in
randomly diffuse in vivo but are spatially and tempo- the cell cytoskeleton, nuclear alterations affecting gene
rally organized by the neighboring cells and the ECM expression, and cell adhesion site reconfigurations.
within the niche. Also, many ligands that influence Each of these biophysical elements mediates mechanical
stem cell fate decisions in vivo are tethered to the cell forces, and together guide cell behavior, organization,
membrane or reversibly bound to the ECM. Thus, the and differentiation.

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


10 1. THE NEED TO STUDY, MIMIC, AND TARGET STEM CELL NICHES

Interfacial wettability and its effect on cellular microenvironment is best characterized in the bone
behaviors were first proposed in a study by Lampin marrow. The HSC niche is laden with immune cells
in 1997, which established a correlation between implicated in complex cell, ECM, and cytokine interac-
matrix wettability and cell adhesion and migration.34 tions, which is vital for the development of the lympho-
Over the years, many wettability-mediated matrices hematopoietic system.37e39 Researchers are also now
have been fabricated to modulate a wide range of able to reveal specific immune regulatory elements
cellular functions from cell adhesion and prolifera- involved in regulating marrow function in concert
tion35 to cell pattern.36 The matrix wettability effect with stromal cells. For example, macrophages have
on stem cell behaviors can be extremely dependent been implicated in HSC mobilization through the pro-
on the cell type. To better understand and evaluate duction of granulocyte-colony stimulating factor,40
the interfacial wettability stem cells need according whereas neutrophils are seen to indirectly influence
to their types, Chapter 16 discusses how interfacial the MSC niche through macrophages.41 Furthermore,
wettability affects stem cell adhesion, proliferation, inflammation in response to injury causes a transient in-
and differentiation. crease of immune cells in tissues to protect against path-
Stem cells, like several other cell types, are sub- ogens and promote tissue healing. The transient stem
jected to fluid flow in the body. In particular, stem cells celleimmune niche interactions mediate endogenous
in vivo experience shear and chemotransport from tissue repair and regenerative mechanisms.42 The func-
fluid flow within mechanically active tissues and tion of immune cells can be modulated to promote
while migrating from niches to homing targets in the stem cell function in cases of continuous tissue injury
body. Chapter 17 introduces this concept describing and scarring. Application of immunomodulation re-
fluid flow devices used for cell stimulation. It further mains an interesting aspect of tissue regeneration strate-
discusses fluid flow mechanical stimulus applied to gies.43 Designing and controlling for this is a current
stem cells as a regulator of proliferation and quies- challenge, and greater appreciation is needed between
cence, differentiation into various lineages (osteo- the interactions of the immune system, the cells
genic, cardiovascular, neural, etc.), migration, and involved in tissue healing, and biomaterials.
tissue remodeling, with a brief discussion of mechan-
ically active pathways.
4. MIMICKING THE STEM CELL NICHE:
BIOENGINEERING TOOLS AND
3.4 Oxygen and Metabolism TECHNIQUES
Of the many metabolic factors influencing cell
The stem cell niche is a complex, dynamic microenvi-
behavior, oxygen tension in the cellular microenviron-
ronment, which is best characterized in an in vivo
ment plays a pivotal role, serving as both metabolic sub-
model. However, it is this complexity that makes
strate and a signaling molecule regulating stem cell fate.
understanding the specific function of individual niche
In vivo, low oxygen tension or hypoxia is a common
components difficult. At the same time, decoding the
feature of stem cell niche shared among different types
effect of specific niche components at the single-cell level
of stem cells and linked to their plasticity. Chapter 18
is challenging. Studying stem cells clonally, as individ-
summarizes the physiological relevance of hypoxia in
ual cellular entities able to self-renew and differentiate,
regulating stem cell metabolism and biological proper-
is emerging as a major focus to understand their origin
ties, including self-renewal, multipotency in differentia-
and key features. Population-based analyses of stem
tion, ischemic resistance, cellular senescence, and
cell behavior often fall short in defining mechanisms
paracrine secretion. It also discusses hypoxia precondi-
that may be unique to these specialized cells. Similarly,
tioning as a therapeutic strategy to enhance efficacy
niche interactions at an individual cell level can reveal
during stem cell transplantation under the context of
significant information on a cell’s microenvironment
disease treatment, including stroke, ischemic heart
and behavior at specific locations when compared with
injury, and kidney injury.
studies performed with cell aggregates. Although the
implementation of clonal assays for the analysis of
stem cells and its niche interaction is experimentally
3.5 Immune Cells, Inflammation, and
challenging, it is crucial for understanding the complex
Immunomodulation mechanisms that govern stemness.
Immunological cells provide dynamic biochemical Although traditional two-dimensional (2D) culture
regulation of the stem cell niche during homeostasis. systems provide a simple means to study stem cell niche
The presence of innate and adaptive immune cells to interactions, they suffer from inherent limitations to
help maintain the integrity of the stem cell replicate the complexity of the native niche and in

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


4. MIMICKING THE STEM CELL NICHE: BIOENGINEERING TOOLS AND TECHNIQUES 11
providing greater insights into in vivo cellecell and crystalline ceramic and amorphous glass compounds.
cellematrix interactions. This has motivated the need Chapter 20 overviews the development of the most
to develop 3D platforms for stem cell culture and for commonly used bioactive ceramics for tissue repair
engineering artificial stem cell niches. The 3D culture and regeneration. It highlights understanding in the
technology opens new avenues for studying relationship between their structure and biocompati-
fundamental questions regarding stem cells and their bility toward designing next-generation bioceramic ma-
niches. Interestingly, it has also allowed several terials for stem cell niche applications.
advanced applications in personalized medicine,44,45 Stem cells can be regulated or controlled by
such as tissue-engineered constructs, biomanufacturing manipulating their microenvironment, and therefore,
approaches, and platforms for drug discovery and controlling the interactions between biomaterials and
toxicity testing. To recapitulate the in vivo niche envi- stem cells is a critical factor for exploring the complete
ronment and further examine the roles of the extrinsic potential of biomaterials. There are a variety of methods
cues in controlling the behavior of a stem cell, artificial and technologies available to fabricate and modify
niches with tunable physical, biochemical, and cellular biomaterials according to the choice of the cell or tissue
parameters have been prepared.9 Bioengineering tools properties that needs to be regenerated. For example,
along with novel fabrication techniques provide useful surface functionalization or modification is one of the
ways to tune niche properties of these stem cells. Bioen- approaches that can create biomimetic microenviron-
gineering tools include synthesizing novel natural, syn- ments, which are able to control stem cell fate and func-
thetic, or hybrid biomaterials and micro- or tions. Chapter 21 highlights the different processes
nanofabrication of niche components in 2D or 3D. involved in modifying the surface of a biomaterial by
Together with the application of sophisticated bioengi- attaching molecules or substances via physical or
neering techniques and analysis methods, chemical methods, or both. It focuses on the biological
manufacturing synthetic stem cell niches will allow us relevance of surface-functionalized biomaterials in the
to (1) culture stem cells under defined conditions, context of stem cell research.
thereby improving reproducibility; (2) facilitate mecha- Fabricating biomaterial scaffolds in 3D with control-
nistic studies to reveal specific roles of various niche lable topographies and stiffnesses and conjugating
cues in regulation of stem cell fate; and (3) develop novel signaling factors to scaffolds to regulate stem cell fates
strategies to engineer stem cell fates in vivo for tissue are critical to simulating in vivo conditions.9 Hydrogels,
regeneration.46 which are hydrated polymer networks, share many key
A range of synthetic and natural polymers is physical properties with native tissues and can be
currently used to fabricate scaffolds. Broadly, bioactive designed to include elements to control cell fate and
materials can be classified into natural and synthetic bio- function. Chapter 22 comprehensively describes
materials. Some of the commonly used natural materials hydrogel design criteria, including source material,
used to mimic ECM include biopolymers like collagen, degradation, topography, adhesion, and growth factor
hyaluronic acid, chondroitin sulfate, fibronectin, alginate, presentation, in the context of 3D stem cell culture. To
chitosan, and silk fibroin. Synthetic polymers, such as complement, Chapter 23 explicitly discusses the
poly(ethylene glycol), poly(lactic acid), poly(glycolic structural features that control the mechanical proper-
acid), and copolymer poly(lactic-glycolic acid) are some ties within hydrogel networks of different types. It also
of the most commonly employed synthetically engineered discusses how gels with different mechanical properties
scaffolds. Chapter 19 highlights recent progress in can be used clinically to control cell differentiation and
polymer design and development for 3D stem cell culture, function.
comparing the advantages of both natural- and synthetic- Recent advancement in fabrication technologies like
based precursors. Additionally, special attention is given nano- and microfabrication has provided opportunities
to smart synthetic polymer systems that exhibit to design biomaterials with intricate topographical
responsiveness to environmental stimuli (e.g., electricity, structures. In addition to functional tissue formation,
temperature, enzyme, light, heat, pH). some of the in vitro applications include (but are not
Although various natural and synthetic bioactive limited to) identifying suitable ECM candidates as
polymers have been engineered to improve the environ- substrates for stem cell culture through micropatterning
mental conditions of a cell, artificially, the choice of the of ECM in 2D; high-throughput ECM microarrays; and
material depends on the selected application, the cell synthesizing novel biomaterials. Chapter 24 reviews
type used, and the tissue type. For example, bioceramics the principal techniques used to generate micro- and
have been considered as one of the most suitable mate- nanotopographical features in substrates suitable for
rials for the repair and reconstruction of diseased or stem cell culture and niche engineering. It describes
damaged parts of the skeletal system. Ceramic materials use of micro- and nanotopographic patterning as a
are inorganic, nonmetallic solids, which include means to probe the stem cell niche, and ultimately to

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


12 1. THE NEED TO STUDY, MIMIC, AND TARGET STEM CELL NICHES

guide stem cell fate through mechanical cues that specialized artificial stem cell niches helps researchers to
govern processes such as adhesion, proliferation, and elucidate the mechanisms by which stem cells receive in-
differentiation. Self-assembly in particular is one of the formation from the multifactorial microenvironment.
most promising fields in building materials and struc- Promoting desirable stem cell phenotypes allows for
tures at the nanoscale. It takes a bottom-up approach the exploitation of their unique property of stemness
for fabrication, where small molecular components and their ability to home and differentiate, thus contrib-
interact with each other under specific conditions to uting to tissue repair. Additionally, in vitro engineered
spontaneously organize into more complex 2D or 3D niches can be used to screen for molecules that can regu-
structures.47,48 Chapter 25 provides a comprehensive late niche biology and thus avoid the need for exogenous
background on self-assembly nanofabrication tech- cell therapy.
niques and their applications in bioengineering stem The currently available therapeutic strategies for end-
cell niches. stage debilitating disease such as congestive heart or
The in vivo ECM in particular possesses a nanoscale liver failure are all invasive surgical approaches,
fibrous topography. Chapter 26 discusses the role of including implantable devices and, ultimately, organ
this topography in modulating stem cell fates in vitro transplantation. However, these surgical treatments are
and details nanofiber fabrication techniques to produce unable to completely restore damaged tissue. For
matrices that have a morphological resemblance to example, all current therapeutic procedures for heart
fibers naturally found in the ECM. Their similar charac- failure to date only modulate hemodynamics and none
teristics suggest that nanofibers could be used as a are available to regenerate heart tissue. Regenerative
supportive matrix for creating artificial niches for stem therapies based on stem cells hold promise as a
cells, upon which additional functionalities could be treatment to overcome this limitation, but to date have
incorporated to further modulate stem cell fates.49 achieved only modest outcomes in clinical trials. To
Stem cells are exposed to a multitude of biochemical enhance potential stem cell behavior and exploit its
gradients across a niche, subjecting them to different maximum functional effect, numerous studies have
signals in different locations in the niche, which shapes taken great effort to elucidate the tissue-specific stem
the way the cells divide. Hence, engineering a modu- cell and progenitor cell niches. Chapters 28e37 demon-
lated niche with spatial complexity would better reflect strate such current advances in the construction of
the in vivo situation. Microfluidics allows for the biomimetic niches to modulate cellematrix interactions
controlled delivery of signals to specific locations within or cellecell interactions for enhanced repair or regener-
the niche, thus modulating the properties of an artificial ation in different tissue types, namely cardiac, bone,
stem cell niche. It provides ways to manipulate single cartilage, skin, and liver. For vascular tissues and
stem cells to better understand behavior across a diverse organs, de novo blood vessel formation, also known as
population of stem cells. In addition, fabricating micro- vasculogenesis, and the subsequent expansion of the
fluidic channels within biomaterials has been shown to nascent vascular network via angiogenesis, constitutes
be a promising tool to mimic nutrient and gas transport a complex vascular niche essential to tissue repair.
for stem cell niche engineering.50 Chapter 27 reviews Coordinating blood vessel generation along with paren-
microfluidic concepts used to generate a wide variety chymal tissue development is critical so that an
of biomolecule gradients and presents the most adequate supply of nutrients and oxygen is maintained
successful applications of microfluidic deviceeinduced along with the removal of waste metabolites. Harness-
gradients for pluripotent stem cell patterning. ing niche biology and in vitro model systems to engineer
a regenerative therapeutic is the ultimate goal.

5. BIOENGINEERING SPECIALIZED
ARTIFICIAL STEM CELL NICHES FOR
CLINICAL THERAPIES References
1. Blau HM, Brazelton TR, Weimann JM. The evolving concept of a
Stem cells are promising cell source candidates for use stem cell: entity or function? Cell 2001;105:829e41.
2. Schofield R. The relationship between the spleen colony-forming
in regenerative medicine. With the recent discovery of cell and the haemopoietic stem cell. Blood Cells 1978;4:7e25.
induced pluripotent stem cells (iPSCs), emerging combi- 3. Morrison SJ, Spradling AC. Stem cells and niches: mechanisms that
nations of biomaterials and iPSCs are bringing promote stem cell maintenance throughout life. Cell 2008;132:
unprecedented opportunities for treating debilitating hu- 598e611.
man diseases.51 Potential knowledge from the biomate- 4. Carlone DL, Breault DT. Slowly cycling versus rapidly cycling
intestinal stem cells: distinct roles or redundancy. Cell Cycle 2011;
rials-mediated approaches for enhancing stem 10:723e4.
cellebased tissue repair is being applied for achieving 5. Glauche I, et al. Stem cell proliferation and quiescenceetwo sides
many therapeutic and nontherapeutic goals. Engineering of the same coin. PLoS Comput Biol 2009;5:e1000447.

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS


REFERENCES 13
6. Wilson A, et al. Hematopoietic stem cells reversibly switch from 29. Winer JP, Janmey PA, McCormick ME, Funaki M. Bone marrow-
dormancy to self-renewal during homeostasis and repair. Cell derived human mesenchymal stem cells become quiescent on
2008;135:1118e29. soft substrates but remain responsive to chemical or mechanical
7. Discher DE, Mooney DJ, Zandstra PW. Growth factors, matrices, stimuli. Tissue Eng Part A 2009;15:147e54.
and forces combine and control stem cells. Science 2009;324:1673e7. 30. Engler AJ, et al. Myotubes differentiate optimally on substrates
8. Lane SW, Williams DA, Watt FM. Modulating the stem cell niche with tissue-like stiffness: pathological implications for soft or stiff
for tissue regeneration. Nat Biotechnol 2014;32:795e803. microenvironments. J Cell Biol 2004;166:877e87.
9. Peerani R, Zandstra PW. Enabling stem cell therapies through 31. Chopra A, Tabdanov E, Patel H, Janmey PA, Kresh JY. Cardiac
synthetic stem cell-niche engineering. J Clin Invest 2010;120: myocyte remodeling mediated by N-cadherin-dependent
60e70. mechanosensing. Am J Physiol Heart Circ Physiol 2011;300:
10. Mascre G, et al. Distinct contribution of stem and progenitor cells H1252e66.
to epidermal maintenance. Nature 2012;489:257e62. 32. Engler AJ, Sen S, Sweeney HL, Discher DE. Matrix elasticity directs
11. Doupe DP, Klein AM, Simons BD, Jones PH. The ordered architec- stem cell lineage specification. Cell 2006;126:677e89.
ture of murine ear epidermis is maintained by progenitor cells with 33. Huebsch N, et al. Harnessing traction-mediated manipulation of
random fate. Dev Cell 2010;18:317e23. the cell/matrix interface to control stem-cell fate. Nat Mater 2010;
12. Greco V, Guo S. Compartmentalized organization: a common and 9:518e26.
required feature of stem cell niches? Development 2010;137:1586e94. 34. Lampin M, Warocquier-Clérout R, Legris C, Degrange M, Sigot-
13. Kimble JE, White JG. On the control of germ cell development in Luizard MF. Correlation between substratum roughness and
Caenorhabditis elegans. Dev Biol 1981;81:208e19. wettability, cell adhesion, and cell migration. J Biomed Mater Res
14. Xie T, Spradling AC. A niche maintaining germ line stem cells in 1997;36:99e108.
the Drosophila ovary. Science 2000;290:328e30. 35. Oliveira SM, Song W, Alves NM, Mano JF. Chemical modification
15. Driskell RR, Giangreco A, Jensen KB, Mulder KW, Watt FM. Sox2- of bioinspired superhydrophobic polystyrene surfaces to control
positive dermal papilla cells specify hair follicle type in mamma- cell attachment/proliferation. Soft Matter 2011;7:8932e41.
lian epidermis. Development 2009;136:2815e23. 36. Piret G, et al. Culture of mammalian cells on patterned superhy-
16. Hsu YC, Pasolli HA, Fuchs E. Dynamics between stem cells, niche, drophilic/superhydrophobic silicon nanowire arrays. Soft Matter
and progeny in the hair follicle. Cell 2011;144:92e105. 2011;7:8642e9.
17. Trumpp A, Essers M, Wilson A. Awakening dormant haemato- 37. Golde DW, Gasson JC. Hormones that stimulate the growth of
poietic stem cells. Nat Rev Immunol 2010;10:201e9. blood cells. Sci Am 1988;259:62e71.
18. Taichman RS. Blood and bone: two tissues whose fates are inter- 38. Gordon MR. Superpowers seek accord on visiting mental
twined to create the hematopoietic stem-cell niche. Blood 2005; hospitals. NY Times Web 1988;1:12.
105:2631e9. 39. Torok-Storb B. Cellular interactions. Blood 1988;72:373e85.
19. Yin T, Li L. The stem cell niches in bone. J Clin Invest 2006;116: 40. Chow A, et al. Bone marrow CD169þ macrophages promote the
1195e201. retention of hematopoietic stem and progenitor cells in the mesen-
20. Boulais PE, Frenette PS. Making sense of hematopoietic stem cell chymal stem cell niche. J Exp Med 2011;208:261e71.
niches. Blood 2015;125:2621e9. 41. Kohler A, et al. G-CSF-mediated thrombopoietin release triggers
20a. Yin X, Farin HF, van Es JH, Clevers H, Langer R, Karp JM. Niche- neutrophil motility and mobilization from bone marrow via induc-
independent high-purity cultures of Lgr5þ intestinal stem cells and tion of Cxcr2 ligands. Blood 2011;117:4349e57.
their progeny. Nat Methods 2014;11:106e12. 42. Kizil C, Kyritsis N, Brand M. Effects of inflammation on stem cells:
20b. Jaks V, Barker N, Kasper M, van Es JH, Snippert HJ, Clevers H, together they strive? EMBO Rep 2015;16:416e26.
Toftgård R. Lgr5 marks cycling, yet long-lived, hair follicle stem 43. Vishwakarma A, et al. Engineering immunomodulatory biomate-
cells. Nat Genet 2008;40:1291e9. rials to tune the inflammatory response. Trends Biotechnol 2016;34:
21. Kreso A, Dick JE. Evolution of the cancer stem cell model. Cell Stem 470e82.
Cell 2014;14:275e91. 44. Yin X, et al. Engineering stem cell organoids. Cell Stem Cell 2016;18:
22. Reichardt LF, Tomaselli KJ. Extracellular matrix molecules and 25e38.
their receptors: functions in neural development. Annu Rev 45. Griffith LG, Swartz MA. Capturing complex 3D tissue physiology
Neurosci 1991;14:531e70. in vitro. Nat Rev Mol Cell Biol 2006;7:211e24.
23. Lu P, Takai K, Weaver VM, Werb Z. Extracellular matrix degrada- 46. Song H, et al. Interrogating functional integration between injected
tion and remodeling in development and disease. Cold Spring Harb pluripotent stem cell-derived cells and surrogate cardiac tissue.
Perspect Biol 2011;3. Proc Natl Acad Sci USA 2010;107:3329e34.
24. Vishwakarma A, Sharpe P, Shi S, Ramalingam M. Stem cell biology 47. Ariga K, Hill JP, Ji Q. Layer-by-layer assembly as a versatile
and tissue engineering in dental sciences. Academic Press; 2014. bottom-up nanofabrication technique for exploratory research
25. Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic and realistic application. Phys Chem Chem Phys 2007;9:2319e40.
microenvironment for stem cell niche. Biochim Biophys Acta 2014; 48. Borges J, Mano JF. Molecular interactions driving the layer-by-
1840:2506e19. layer assembly of multilayers. Chem Rev 2014;114:8883e942.
26. Gattazzo F, Urciuolo A, Bonaldo P. Extracellular matrix: a dynamic 49. Lim SH, Mao HQ. Electrospun scaffolds for stem cell engineering.
microenvironment for stem cell niche. Biochim Biophys Acta 2014; Adv Drug Deliv Rev 2009;61:1084e96.
1840:2506e19. 50. Cuchiara MP, Allen AC, Chen TM, Miller JS, West JL. Multilayer
27. Flaim CJ, Chien S, Bhatia SN. An extracellular matrix microarray microfluidic PEGDA hydrogels. Biomaterials 2010;31:5491e7.
for probing cellular differentiation. Nat Methods 2005;2:119e25. 51. Tong Z, et al. Application of biomaterials to advance induced
28. Gobaa S, et al. Artificial niche microarrays for probing single stem pluripotent stem cell research and therapy. EMBO J 2015;34:
cell fate in high throughput. Nat Methods 2011;8:949e55. 987e1008.

I. BIOLOGY OF STEM CELL NICHES AND MOLECULAR MECHANISMS

You might also like