Download as pdf or txt
Download as pdf or txt
You are on page 1of 37

Leptin: Master Regulator of Biological Functions

that Affects Breathing


Estelle B. Gauda,*1 Silvia Conde,2 Mirian Bassi,3 Daniel B. Zoccal,3 Debora Simoes Almeida
Colombari,3 Eduardo Colombari,3 and Nikola Despotovic4

ABSTRACT
Obesity is a global epidemic in developed countries accounting for many of the metabolic and
cardiorespiratory morbidities that occur in adults. These morbidities include type 2 diabetes, sleep-
disordered breathing (SDB), obstructive sleep apnea, chronic intermittent hypoxia, and hyperten-
sion. Leptin, produced by adipocytes, is a master regulator of metabolism and of many other
biological functions including central and peripheral circuits that control breathing. By binding
to receptors on cells and neurons in the brainstem, hypothalamus, and carotid body, leptin links
energy and metabolism to breathing. In this comprehensive article, we review the central and
peripheral locations of leptin’s actions that affect cardiorespiratory responses during health and
disease, with a particular focus on obesity, SDB, and its effects during early development. Obesity-
induced hyperleptinemia is associated with centrally mediated hypoventilation with decrease CO2
sensitivity. On the other hand, hyperleptinemia augments peripheral chemoreflexes to hypoxia and
induces sympathoexcitation. Thus, “leptin resistance” in obesity is relative. We delineate the cir-
cuits responsible for these divergent effects, including signaling pathways. We review the unique
effects of leptin during development on organogenesis, feeding behavior, and cardiorespiratory
responses, and how undernutrition and overnutrition during critical periods of development can
lead to cardiorespiratory comorbidities in adulthood. We conclude with suggestions for future
directions to improve our understanding of leptin dysregulation and associated clinical diseases
and possible therapeutic targets. Lastly, we briefly discuss the yin and the yang, specifically the
contribution of relative adiponectin deficiency in adults with hyperleptinemia to the development
of metabolic and cardiovascular disease. © 2020 American Physiological Society. Compr Physiol
10:1047-1083, 2020.

Didactic Synopsis • Leptin acts on the carotid body to modify spontaneous


minute ventilation and hypoxic ventilatory responses.
Major teaching points • Leptin is involved in carotid body responses to chronic
Leptin is a master regulator of metabolism and of many conditions as obesity and chronic intermittent hypoxia.
other biological functions including breathing. Neuronal
• Hyperleptinemia contributes to increased carotid body
circuits that are primarily involved in feeding behavior and
sensitization that promotes altered breathing and aug-
metabolism are interconnected with those in the respira-
mented sympathetic activity.
tory network. Leptin also modifies the activity from cells
and neurons in the carotid body, which mediate changes in • During early development, leptin does not decrease feed-
ventilation and sympathetic activity in response to hypoxia. ing behavior or increase CO2 sensitivity.
Leptin deficiency or leptin resistance occurring at any time • Leptin is key to organogenesis during early development.
during the lifespan can lead to adverse cardiorespiratory
outcomes including abnormal feeding behavior, obesity,
*Correspondence to estelle.gauda@sickkids.ca
hypertension, metabolic dysregulation, intermittent hypoxia, 1 Division of Neonatology, Department of Pediatrics, The Hospital for
and obstructive sleep apnea. Sick Children, Toronto, Ontario, Canada
2 CEDOC, NOVA Medical School, Faculdade de Ciências Médicas,
• Breathing must match the metabolic demands of the body. Lisboa, Portugal
3 Department of Physiology and Pathology, School of Dentistry, São
• Leptin, a hormone produced by fat tissue, is able to
Paulo State University (UNESP), Araraquara, São Paulo, Brazil
stimulate breathing by acting on central POMC-MC4R 4 Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
pathway.
Published online, July 2020 (comprehensivephysiology.com)
• Leptin-signaling deficiency potentially contributes to DOI:10.1002/cphy.c190031
obesity-associated breathing disorders. Copyright © American Physiological Society.

Volume 10, July 2020 1047


Leptin in the Control of Breathing Comprehensive Physiology

• Overnutrition (hyperleptinemia) and undernutrition subsequent protein was nonfunctional. This nonfunctional
(hypoleptinemia) during fetal and early development lead protein downregulated its expression leading to increased
to leptin resistance in adulthood and cardiorespiratory levels of detectable mRNA. Today, we recognize the ob gene
comorbidities. product as a mutant form of the leptin (LEP) gene, which was
aptly leptos from the Greek root for “thin.” Predictably, the db
• Adiponectin, another adipokine produced by adipocytes,
gene product is a mutation in the leptin receptor (LEPR) gene.
counterbalances the adverse effects of leptin, but circulat-
Leptin is primarily produced in white adipose tissue, although
ing levels are low in individuals with obesity and sleep-
brown adipose tissue, placental syncytiotrophoblasts, mam-
disordered breathing.
mary glands, skeletal muscle, and ovaries are amongst a
handful of other tissues capable of synthesizing this hormone
(111, 244). Its serum concentration is under diurnal regula-
Introduction tion, increasing at night to suppress hunger and decreasing
throughout the day to stimulate metabolism (81). Interest-
Leptin is pleiotropic adipocyte-derived hormone that is
ingly, its serum concentration increases exponentially with
most notable for its role in energy homeostasis and appetite
adipose tissue mass (66).
suppression. However, it is also a master regulator of many
biological functions including organogenesis, metabolism,
learning and memory, reproduction, cardiovascular home- Leptin gene and protein product
ostasis, and immunity, to name a few. After describing
LEP is located on chromosome 7q32.1 in humans, which
the journey to its discovery, characterization, and cellular
encodes a 16 kDa peptide hormone consisting of 146 amino
and molecular signaling, this article will largely focus on
acids. Within the primary sequence is a 21 amino acid signal
the unique contribution of leptin to central and peripheral
peptide that is highly conserved between animal species
systems that control breathing in health and disease.
(172). While there are no common human variants of the
leptin gene, eight mutant forms have been discovered in
Eastern countries—all of which result in extreme infant
Overview of Leptin obesity and hyperphagia. Seven of the mutant forms impair
LEP expression, resulting in low serum leptin. Conversely,
Discovery of leptin the most recently discovered mutation (89, 274), in 2014,
In 1949 at the Jackson Laboratory, Dr. George Snell and identified a transversion that resulted in high serum levels of
his colleagues noticed that one of their mouse lines devel- a nonfunctioning protein (376).
oped extreme obesity. After a series of investigations, they
concluded that it was caused by a mutation in an autosomal
recessive gene for obesity (ob/ob) (161). Although they Leptin receptor
were unable to recognize the gene or identify its product, Leptin exists in both a free and bound form in the cir-
they were able to locate the gene to chromosome 6. In the culation; both forms are capable of binding to the leptin
1960s, another new strain of obese and diabetic mice (db/db) receptor (Ob-R). Ob-R is a member of the class I cytokine
was discovered at the Jackson Laboratory (158). Similar receptor family, which includes other receptors with the
to the ob/ob mouse, the db/db mouse had hyperphagia, gp130 subunit, such as the IL-2, 3, 4, 6, and 7 receptors
extreme obesity, diabetes, neuroendocrine dysfunction, and (133, 380). The six Ob-R isoforms—identified as Ob-Ra
infertility. to Ob-Rf, sequentially—are alternative splicing products
A seminal parabiosis experiment performed by Coleman of the LEPR gene. The isoforms are subdivided into three
(60) substantially moved the field forward. They surgically classes: long (Ob-Rb), short (Ob-Ra, Ob-Rc, Ob-Rd, and
joined the circulatory systems of ob/ob and db/db mice and Ob-Rf), and secretory (Ob-Re). All of the Ob-R isoforms
observed that the ob/ob mouse was no longer hyperphagic have a conserved extracellular ligand-binding domain on their
and had significant weight loss and improved glucose reg- N-terminus, which consists of 816 amino acids, 4 essential
ulation. They concluded that the circulating factor in the cysteine residues, a Trp-Ser-X-Trp-Ser (WSXWS) motif, and
db/db mouse was inducing the changes observed in the ob/ob a fibronectin III domain. Five of the isoforms—excluding
mouse. Thereafter, Dr. Jeffrey Friedman, a young resident the secretory Ob-Re isoform—have a 34 amino acid trans-
physician, directed his full attention to the discovery and membrane domain that anchors the receptor to the plasma
characterization of the product of the ob gene. In 1994, using membrane, as well as a variable length intracellular domain.
positional cloning techniques, he detected a 20-fold increase The intracellular domain of the short isoforms ranges from
in expression of a 4.5 kb RNA strand in the adipose tissue 32 to 40 amino acids, while Ob-Rb has 303 (133, 244, 380).
of C57B1/6J ob/ob mice (401). Further studies revealed that The short isoforms contain a box 1 motif that allows for
the increase in 4.5 kb RNA was the result of a snowball it to activate Janus kinase (JAK) tyrosine kinases, but the
effect: The C57B1/6J ob/ob mice had a nonsense mutation strength of the signal is much weaker than the long Ob-Rb
that transcribed a detectable RNA product, but for which the isoform, which contains three motifs for the binding of JAK

1048 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

tyrosine kinase and the signal transducer and activator of 1, box 2, and box 3 motifs. Box 1 and box 2 motifs are required
transcription (STAT) transcription factor. Thus, the Ob-Rb for the binding of JAK tyrosine kinase, while box 3 allows for
has been identified as the master regulator of intracellular the binding of the STAT transcription factor. Short isoforms
signaling from leptin; the short isoforms are associated of Ob-R only contain the box 1 motif and are consequently
with leptin internalization and proteolysis, and the secretory unable to activate all JAK kinases (133). On the intracellular
Ob-Re isoform is a serum carrier protein that transfers leptin domain of the Ob-R receptors, there are four intracellular
to the membrane proteins (111, 133). As it relates to obesity, tyrosine residues (Tyr974, Tyr985, Tyr1077, and Tyr1139)
the Ob-Rb isoform is the most important because it predom- that are involved in the activation of the JAK/STAT and other
inates in the hypothalamus—where the central control of intracellular signal transduction pathways described below.
hunger takes place (266). Its role in feeding behavior, along Upon ligand binding to Ob-Rb, JAK2 binds to box 1 and
with the concept that Ob-Rb is the only isoform capable of box 2 motifs and is subsequently autophosphorylated. This
fully activating the JAK/STAT signal transduction pathway, phosphorylated JAK2 then activates Tyr985 and Tyr1138,
helps to explain why modifications to the Ob-Rb receptor are allowing for the binding of STAT proteins on the box 3 motif.
responsible for the development of early-onset obesity. STAT proteins—most notably STAT3—are then activated
through subsequent tyrosine phosphorylations by JAK2. The
activated STAT3 protein dissociates from the receptor and
Leptin signaling forms a homo- or heterodimer that is capable of translocating
As depicted in Figure 1 (111), leptin signaling primarily to the nucleus, where it exerts its influence on the gene
occurs through the JAK/STAT pathway following ligand expression of suppressor of cytokine signaling 3 (SOCS3)
binding to Ob-Rb. Only Ob-Rb is capable of fully activating and protein tyrosine phosphatase 1B (PTP1B), among other
this transduction pathway because it contains all three of box proteins (111, 133, 244). SOCS-3 and PTP1B are notable

L Leptin

Ob-Rb
Membrane

IRS 1/2
B P Cytosol
o JAK2
x PI3K
1
ERK
P
974 P
Grb-2
SHP-2
P SOCS3 SOCS3
985
AgRP

POMC
P STAT-5 +
1077
STAT-3
P P P −
1138 ST
1/3 AT STAT-3 Nucleus
/5

Figure 1 Intracellular signaling pathways activated when leptin binds to the long form of the
leptin receptor (Ob-Rb) in cells in the arcuate nucleus that express POMC and AgRP. Janus kinase
(JAK2) associates with the receptor via the box1 motif. The long isoform leptin (L) receptor (Ob-Rb) contains four
important tyrosine residues (Tyr974 , Tyr985 , Tyr1077 , and Tyr1138 ). These phosphorylated tyrosine residues pro-
vide docking sites for signaling proteins. Tyr1138 recruits the transcription factor STAT3, which is subsequently
phosphorylated by JAK2, dimerizes, and translocates to the nucleus, where it induces SOCS3 and POMC (pro-
opiomelanocortin) expression, while repressing AgRP (agouti-related peptide). SOCS proteins inhibit signaling by
binding to phosphorylated JAK proteins or interacting directly with tyrosine-phosphorylated receptors. The ability
of SOCS3 to inhibit leptin-stimulated phosphorylation of JAK2 and ERK provides a negative feedback mechanism
on the leptin signaling system. Grb-2, growth factor receptor binding-2; STAT3, signal transducer and activator of
transcription; SOCS, suppressor of cytokine signaling. Adapted, with permission, from Fruhbeck G, 2006 (111).

Volume 10, July 2020 1049


Leptin in the Control of Breathing Comprehensive Physiology

for their ability to act as negative feedback modulators in the (MAPK) cascade, phosphoinositide 3-kinase (PI3K) cAMP
JAK/STAT pathway, by inhibiting the activity of JAK2 and pathway, and via 5′ -AMP-activated protein kinase (AMPK)
dephosphorylating the JAK2 protein, respectively (111). (111). Each of these pathways has a role in nonhypothalamic
In the hypothalamus, the Ob-Rb receptor is located on tissues, several of which are used in cells and neurons in the
two distinct neuronal populations: the proopiomelanocortin brainstem and peripheral chemoreceptors that are involved in
(POMC) and the neuropeptide Y/agouti-related protein respiration.
(NPY/AgRP) neurons. POMC neurons are anorexigenic;
as such, their activation through Ob-Rb ligand binding
increases POMC expression, which eventually leads to the
loss of adiposity. Contrarily, NPY/AgRP neurons—which are Leptin and Central Control of Breathing
orexigenic—are inhibited by Ob-Rb activity, leading to a loss
of appetite. It is through this JAK/STAT signaling pathway Overview of breathing control and chemoreception
for POMC, NPY, and AgRP that we as humans are able to Breathing is an automatic act generated by a complex net-
regulate our fat stores (374). In the case of obesity, the Ob-R work located in the hindbrain (the pons and medulla). This
often becomes resistant to leptin, so although leptin levels respiratory network, schematically depicted in Figure 2 (120),
are high in serum—signaling for weight loss—appetite is not rhythmically coordinates the contraction and relaxation of
suppressed. Physiological leptin resistance occurs during the thoracic and abdominal muscles to produce pressure gradients
neonatal, prepubertal, and pregnant stages of life facilitating that move the air in and out of the lungs, as well as regulate
rapid weight gain (134, 171, 173). muscles that control the upper airway patency and then the
Leptin signaling can also occur through several other path- resistance to airflow (77). The respiratory rhythm, inspiratory
ways including through the mitogen-activated protein kinase and expiratory phase transition, and muscular pattern control

Orexinergic neurons
SARs Peripheral
Inspiratory neurons Laryngeal
RARs arterial
chemoreflex
Expiratory neurons C fiber chemoreceptors

CO2/H+ chemosensors

Pontine
respiratory
group Nucleus of solitary tract
Forebrain (phase switching)
(hypothalamus) Dorsal respiratory group
inspiratory neurons

Ventral respiratory column

Facial Rostral (rhythm generating) Caudal (pattern forming)


nucleus PBC
BötzC Ventral respiratory group VRG
pacemaker neurons
expiratory inspiratory and
neurons Rostral VRG Caudal VRG
expiratory
RTN/pFRG
CO2/H+

Bulbospinal
neurons

Bulbospinal neurons synapse with respiratory motoneurons


(phrenic, intercostals, upper airway, abdominals)

Figure 2 The medullary respiratory network with pulmonary and pontine feedbacks. General
schematic diagram representing the respiratory network with two interacting feedback. This schematic shows the
interactions between different populations of respiratory neurons within major brain areas involved in the control
of breathing, such as the CO2 -sensitive cells in the hypothalamus, Raphe and RTN/pFRG, the pons, the ventral
respiratory group (BötzC, PBC and VRG), the NTS and the peripheral inputs and efferents. Abbreviations: BötzC,
Bötzinger complex; PBC, pre-Bötzinger complex; RTN/pFRG, retrotrapezoid nucleus/parafacial respiratory group;
SAR, slowly adapting receptors; RAR, rapidly adapting receptors. Adapted, with permission, from Gauda E and Martin
R, 2018 (120).

1050 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

are determined by the intrinsic membrane properties of the send signals to the respiratory neuronal networks in the
respiratory premotor and motor neurons located in the brain- brain (197).
stem and spinal cord, as well as by the reciprocal excitatory Respiratory movements emerge from the synchronous
and inhibitory synaptic interactions of pontomedullary res- interactions of respiratory neuronal groups located in the
piratory nuclei. Moreover, the functioning of the respiratory brainstem, with distinct phenotypic and biophysical features
central pattern and rhythm generator is continuously modified (307, 308, 405). The respiratory groups reside in a long
by neuromodulators, such as peptides, amines, and hormones, column of cells, extending from the caudal medulla along the
which set either the excitability of respiratory neurons or the ventrolateral medulla to the dorsolateral pons as well as to the
synaptic strength between neuronal groups. nucleus of the solitary tract (NTS) (35, 242, 251). Within this
An important characteristic of the mammalian respiratory network, there is a group of inspiratory neurons, located in
system is its ability to adjust breathing rhythm and pattern the ventral surface of medulla, which are able to intrinsically
according to changes in internal and external environments. generate fictive inspiratory bursts when isolated in vitro
Ventilation increases and decreases proportionally to swings (346). These neurons composed the so-called pre-Bötzinger
in carbon dioxide production and oxygen consumption complex (pre-BötC). Because the inhibition of the pre-BötC
caused by changes in metabolic rate. The respiratory system neurons in vivo stops breathing, this region is suggested to be
is also capable of compensating for disturbances that affect the primary inspiratory oscillator (361). In addition to their
the mechanics of breathing, such as the airway narrowing intrinsic properties, there is evidence indicating that breathing
that occurs in asthmatic patients. Breathing also undergoes generation under resting conditions also requires interactions
appropriate adjustments when the mechanical advantage with other respiratory neuronal groups, especially from the
of the respiratory muscles is altered by postural changes Bötzinger complex (BötC) (216)—region located in the
or by movement. This flexibility of breathing arises, to a rostral ventral respiratory column that contains inhibitory
great degree, from sensors distributed throughout the body expiratory neurons. Connections between pre-BötC/BötC
that send feedback and feedforward signals to the brainstem with pontine and other medullary regions (Raphe and NTS)
respiratory network. Among these sensors, there are chemore- are necessary for proper respiratory motor pattern formation
ceptors that detect changes in levels of oxygen and acidity and phase control (67, 345), generating eupneic breathing
in the arterial blood and in the brain; and mechanoreceptors (Figure 2) (120).
that monitor the expansion of the lung, the size of the airway, A second respiratory oscillator has also been identified in
the force of respiratory muscle contraction, and the extent of the rostral ventrolateral medulla (RVLM), which is condition-
muscle shortening (197). ally recruited to control expiratory motor activity (167). This
Although the diaphragm is the major inspiratory pumping oscillator, presented in the retrotrapezoid nucleus/parafacial
muscle, its action is assisted and augmented by a complex respiratory group (RTN/pFRG) and composed of expiratory
assembly of respiratory muscles. Intercostal muscles insert- neurons, is responsible for providing excitatory drive to
ing on the ribs, the abdominal muscles, and muscles such as promote abdominal expiratory contractions, hence increas-
the scalene and sternocleidomastoid that attach both to the ing minute ventilation, during situations of high metabolic
ribs and to the cervical spine at the base of the skull also demand, such as physical exercise, or during hypoxia
play an important role supporting the changes in pulmonary (reduced PO2 ) and hypercapnia (enhanced PCO2 ) (1, 2, 405).
volume, then the generation of inspiratory and expiratory Experiments are still being done to determine how this
flows. Moreover, laryngeal muscles and muscles of the oral conditional expiratory oscillator interacts with other res-
and nasal pharynx adjust the upper airway resistance to gas piratory compartments within the brainstem to coordinate
movement during inspiration and expiration. The fact that inspiratory and expiratory outputs during the occurrence
these muscles are engaged in other physiological functions, of active expiration. More recently, the “triple oscillator
such as speaking, suckling, chewing and swallowing, and network” theory has been proposed with the identification
postural maintenance, highlights the complexity of their of a group of excitatory neurons rostral to the pre-BötC and
control by the central nervous system. The nonrespiratory dorsomedial (DM) to the nucleus ambiguus, post-inspiratory
actions of the respiratory muscles indicate that the function- complex (PiCo), which putatively is responsible for the
ing of brainstem respiratory network (responsible for the rhythmical control of post-inspiratory activity (10). This third
automatic generation of breathing) is significantly influenced respiratory oscillator might be responsible for generating
by higher brain centers and even controlled voluntarily to post-inspiratory behaviors, such as vocalization, swallowing,
a substantial degree. An outstanding example of voluntary and breath holding (11).
control is the ability to suspend breathing by holding one’s Disturbances in blood gases and arterial pH are rapidly
breath. Inputs from higher brain centers to the brainstem detected by the body, causing effects on breathing. In con-
respiratory network promote breathing adjustments that not scious animals, minor changes in brain interstitial fluid (ISF)
only support behavioral-related respiratory changes but also pH are sufficient to generate reflex response on ventilation
satisfied metabolic demands accompanied by minimal use (101, 252, 264)—the reduction of ISF pH from 7.30 to 7.25 is
of energy. This flexibility in breathing patterns in large part associated with a twofold increase in alveolar ventilation. The
arises from sensors distributed throughout the body that traditional concept of the function of central chemoreception

Volume 10, July 2020 1051


Leptin in the Control of Breathing Comprehensive Physiology

is that it, along with peripheral chemoreception at the carotid baseline ventilation and the ventilatory response to CO2 in
body (CB), (i) regulates arterial PCO2 within normal limits leptin-deficient (ob/ob) mice (26). These results indicate that
in response to primary changes in CO2 , and (ii) regulates leptin facilitates the processing of ventilatory response to
blood pH in response to acid-base disturbances (142, 252). hypercapnia, possibly by acting on either chemosensitive or
Eight putative groups of neurons that exhibit capacity to respiratory neurons of the RVLM, including the neurons from
intrinsically detect changes in H+ levels have been identified the RTN and the rostral ventral respiratory column. Inter-
(i) the RTN/pFRG (198, 199), (ii) the rostral medullary raphe estingly, leptin produced negligible acute effects on basal
(MR) (raphe magnus) (152, 254), (iii) the caudal MR (152, activity and CO2 /H+ response of chemosensitive neurons
153) (raphe obscurus) indirectly via the RTN (78), (iv) the of the RTN (27), when administrated to brainstem slices in
caudal NTS (253), (v) the region just dorsal to the caudal vitro. One explanation for this finding is that leptin actions
ventral medullary surface (70), (vi) the pre-BötC (317), on ventral chemosensitive region of the medulla require a
(vii) the fastigial nucleus of the cerebellum (218), and (viii) time to develop and may involve the synthesis of intracellular
the orexin-containing neurons of the hypothalamus (252). proteins that control cellular activity and excitability. Alterna-
Mild focal acidosis at many, but not all, these brainstem sites tively, the effects of leptin may be indirect and involve other
can stimulate ventilation. types of cells. These possibilities still await experimental
The hypothalamus is a key center for autonomic and neu- verification to be confirmed.
roendocrine control but also influences other neurovegetative Findings from several studies in animals show that leptin
mechanisms including respiration. Different hypothalamic acts centrally to alter ventilation (12). In anesthetized rats, lep-
nuclei, such as the paraventricular nucleus (PVN), perifor- tin administration in the NTS—the primary site of peripheral
nical area (PFA), dorsomedial hypothalamus (DMH), and cardiorespiratory afferents in the brainstem—promotes an
lateral and posterior hypothalamus, are able to generate increase of respiratory motor output and ventilatory response
respiratory responses when engaged. These hypothalamic to CO2 (58, 163). Leptin also attenuates the cardiovagal
nuclei are interconnected with respiratory nuclei located component of the baroreceptor reflex (16) and potentiates
in the midbrain, pons, medulla and spinal cord (113). For sympathoexcitatory responses evoked by the activation of
instance, the activation of DMH neurons causes a vigorous the chemoreflex (58). Moreover, systemic administration of
stimulation of respiratory activity, causing hyperventilation leptin enhances c-fos expression in leptin receptor-expressing
(224). Moreover, projections from the PFA neurons to the neurons of the caudal aspect of the NTS (region that receives
NTS, to the dorsolateral pons, including the Kölliker-Fuse predominantly afferent inputs from peripheral chemorecep-
nucleus and the lateral and medial parabrachial nuclei and to tor) (93, 135), supporting the hypotheses that leptin activates
the respiratory motor neurons in the spinal cord have been NTS neurons involved with regulation of respiration.
identified (49, 114, 186, 248, 267, 279, 318, 319, 400). The In addition to the potential effects of leptin in the medulla,
PFA also receives afferent fibers from other hypothalamic leptin may also act on hypothalamic neurons and modify
areas such as the arcuate (ARC) nucleus (127) which is a crit- breathing. Chronic administration (7 consecutive days) of
ical center for modulation of the metabolic, cardiovascular, leptin in the lateral ventricle (LV) reduced daily food intake
and respiratory functions. and body weight while it enhanced baseline ventilation in rats
(Figure 3) (30). This leptin effect on basal ventilation was
principally secondary to an increase in tidal volume whereas
The role of leptin on the central mechanisms that respiratory frequency did not change, suggesting that leptin
control breathing acts on specific targets in the respiratory network rather than
The output from the respiratory network is constantly producing a generalized respiratory stimulation. Leptin given
adjusted by the actions of neuromodulators, which changes centrally also enhanced the ventilatory response to hyper-
the excitability of respiratory neurons (modifying the conduc- capnia again by increasing tidal volume but frequency was
tance of ion channels or the functional voltage range of the also increased when leptin was given centrally. In agreement
membrane) or the synaptic properties (86). The neuromod- with these observations, LepR/Nestin-cre mice (transgenic
ulators, mostly amines and peptides, can be endogenously model with deletion of leptin receptors in the central ner-
released, or be present in challenge situations (hypoxia vous system) exhibit a diminished ventilatory response to
or hypercapnia for example). Leptin, a peptide hormone hypercapnia (Figure 3); thereby reinforcing the notion that
produced by adipose tissue, acts in brain centers to control leptin acts centrally to modulate breathing and control CO2
critical physiological functions related to metabolism and homeostasis (30).
cardiorespiratory regulation. The importance of leptin for
respiratory control is evident from studies showing that leptin-
deficient mice (ob/ob) exhibit impaired ventilatory responses Leptin and control of breathing in newborns
to hypercapnia (increased PCO2 in the arterial blood), which Similar to adults and children, leptin levels are proportional
can be corrected by exogenous leptin administered to the to fat mass in the fetus and newborn, but it is also produced
central nervous system. For example, microinjections of by the placenta (371). Although leptin levels are lowest in
leptin for three consecutive days into the RVLM increased the smallest and youngest premature infants, it is higher

1052 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

(a) 60
Control Daily i.c.v. microinjections

Food intake (g/day)


45
αβ

30

15 α

0
(A) C1 C2 C3 t1 t2 t3 t4 t5 t6 Control-PBS LEP
SHU SHU + Lep
2400 1500 α
α
(mL/(min kg))

(mL/(min kg))
1600 1000 β
β

ΔVE
VE

αβ β
800 α
500

0
0
(B) Room air 7% CO2

Wild-type LepR/Nestin-cre
9000 9000 MC4R–/– LepR/POMC-cre
(mL/(min kg))

(mL/(min kg))

6000 6000
VE

ΔVE

θγ
θγ
3000 θ 3000
θ
θ θ
0 0
(C) Room air 7% CO2

Figure 3 The effect of central leptin administration on food intake and breathing. (A) Daily
food intake during control period (C1–C3) and during lateral ventricle (LV) treatment (t1–t6) with PBS (control),
(LEP, 5 μg/day), MC3/4R antagonist SHU-9119 (SHU, 0.6 nmol/day), or SHU-9119 + leptin for 7 days in rats.
(B) Basal ventilation and ventilatory response to 7% CO2 after LV treatments in rats. (C) Ventilatory responses in
mice: wild-type, leptin receptor deletion in the entire brain (LepR/Nestin-cre), leptin receptor deletion in POMC
neurons (LepR/POMC-cre) and mice with MC4R deficiency (MC4R−/− ). Adapted, with permission, from Bassi
M, et al., 2015 (30).

during the first several days of life than afterward. Apnea and Knowing that leptin increases CO2 chemosensitivity in adult
periodic breathing are inversely related to gestational age, models, and CO2 sensitivity is highest during the first several
and essentially all infants born less than 28 weeks gestation days in newborn rats, then falls, before increasing again at
will need therapeutic intervention to stabilize breathing 10 days to stabilize to adult levels by postnatal day 21 (355),
and decrease the frequency prolonged apnea and associated Gauda and colleagues designed an experiment using SD rats
bradycardia and hypoxemia, for review see Ref. 80. Of note, to determine the effect of leptin on the ventilatory response to
apnea of prematurity, intermittent hypoxia (IH), and periodic hypercapnia (CO2 5%/O2 40% balanced in N2 ) in freely mov-
breathing increases in frequency after 48 h and peaks 2 to ing rats, during the first 3 to 4 weeks of postnatal development
4 weeks after birth (25, 80, 235). The increased frequency of (2–4, 7–9, and 19–21 days of age, n = 10 per age group). They
periodic breathing parallels the increased responsiveness of found that the ventilatory response to CO2 increased with
the CB (80, 91, 123). Premature infants also have reduced postnatal development; however, the animals between 19 and
sensitivity to CO2 at birth which increases within the first 21 postnatal days had the greatest change in ventilation when
several weeks of postnatal development (75, 108, 309). exposed to hypercapnia. Exogenous leptin (3 or 6 mg/kg)
Chemosensitivity to CO2 and hypoxia in Sprague Dawley given intraperitoneally augmented the ventilatory response to
(SD) rats matures within the first 3 to 4 weeks after birth, and CO2 only in the oldest animals (unpublished data).
SD rats are commonly used as a translational model to study In another series of experiments in unanesthetized and
maturation of mechanisms that control breathing (75, 121). unrestrained SD rats, at postnatal day 18 to 21, Gauda

Volume 10, July 2020 1053


Leptin in the Control of Breathing Comprehensive Physiology

and colleagues determined whether exposure to high CO2 (A)


Aq
(7% CO2 balanced in 40% O2 and N2 ) for 1 h would modify
CG
the level of protein expression of pSTAT (measured with
C3
western blot) in tissue homogenates of the NTS, LC, and
RTN, areas that are known to contain CO2 sensitive cells.
mlf
They found that pSTAT protein expression after 60 min of
exposure was upregulated in the NTS and LC but not in CLi
xscp
the RTN (unpublished data). Thus, these preliminary data
suggested that CO2 sensitivity in newborn rat pups prior to (B) Aq IC
3 weeks of life is not affected by leptin. Of note, 4 to 6 weeks
CG
after birth is the age when neurons in the arcuate nucleus
rDR
of the hypothalamus (ARH) developed the mature adult scp
response to leptin (23), discussed below. It would be of inter-
est to determine if similar cellular mechanisms that mature mlf
the ARH neurons are also occurring in CO2 chemosensitive rMR
areas in the brainstem during development.
(C) 4V
LC
Relative leptin deficiency in newborn infants and risk CG
of SIDS: a translational perspective mlf
cDR
Ob-R mRNA
Increased ventilatory and arousal responses to CO2 is an
important and essential defense response during airway SERT mRNA cMR
obstruction or rebreathing. Premature infants and those
who are small for gestational age (SGA) have a two- to SERT/Ob-R mRNA
threefold increased risk of dying of sudden infant death
syndrome (SIDS) (18, 91). SIDS occurs during sleep and Figure 4 Anatomical localization of serotonin (SERT) and
frequently after infants are fed. Infants who are formula Ob-R mRNA expression in the rostral brainstem of the mon-
fed have increased risk of SIDS in contrast to those who key. Plates represent sections containing five regions of Raphe nuclei
included in the analysis: CLi (A), rostral DR (rDR; B), rostral MR (rMR; B),
are breastfed. In fact, exclusive breastfeeding reduces the caudal DR (cDR; C), and caudal MR (cMR; C). Symbols represent the
odds of SIDS to 0.27 (95% CI 0.44–0.69) (148). Breastmilk locations of cells containing SERT mRNA (gray triangles), Ob-R mRNA
is a source of leptin, and plasma leptin levels do increase (empty inverted triangles), or both SERT/Ob-R mRNAs (black circles).
Aq, cerebral aqueduct; CG, central gray; C3, oculomotor nucleus; mlf,
postprandial in infants (57). Savino et al. (326) measured medial longitudinal fasciculus; xscp, decussation of superior cerebellar
serum leptin levels in a cohort of term infants between 2 peduncle; IC, inferior colliculus; scp, superior cerebellar peduncle; 4V,
and 3 months of age who were exclusively breastfed (n = 25) fourth ventricle; LC, locus coeruleus. Adapted, with permission, from
Finn PD, et al., 2001 (102).
or formula fed (n = 26). Leptin serum levels were higher
in breastfed (7.1 ± 10.4 ng/mL) than in formula-fed infants
(3.7 ± 3.87 ng/mL) infants (P < 0.05). One could speculate mice, the findings could be extrapolated to models of sudden
that increased leptin levels in breastfed infants may con- death when reduced arousal responses are hypothesized to
fer some protection from SIDS by increasing ventilatory be contributory (45). The developmental trajectory of CO2
responses to CO2 when the airway becomes compromised chemosensitivity of these dorsal raphe neurons mediating
or rebreathing occurs during sleep. Smith et al. (326, 344), arousal in animals or humans during sleep is not known.
recently published, that in adult mice, dorsal raphe serotonin Moreover, it is not known whether leptin augments the
neurons mediate CO2 -induced arousal from sleep without arousal response to CO2 during early postnatal develop-
increasing ventilation. Finn et al. used situ hybridization to ment and whether the insensitivity of these neurons to CO2
map neurons in the rostral brainstem that expressed mRNA contributes to the pathophysiology of SIDS.
for the serotonin transporter and Ob-Rs in monkeys (102);
neurons in the caudal dorsal raphe and rostral median raphe
had the highest level of co-expression of mRNA for Ob- Central effects of leptin on cardiorespiratory
Rs and serotonin transporter as shown in Figure 4 (102). coupling
Dorsal raphe neurons in ob/ob mice and lean littermates Cardiorespiratory disorders are commonly associated with
also co-express immunoreactivity for Ob-Rs and serotonin obesity and metabolic dysfunction (Figure 5) (107). The
transporter. Of interest, the leptin-deficient mice had less obstructive sleep apnea (OSA) syndrome is the most com-
neurons in the dorsal raphe that co-expressed immunoreac- mon breathing disease related to obesity (71, 212, 214,
tivity for Ob-Rs and serotonin transporter (61). While the 265). The pathological mechanisms causing hypertension,
mouse study designed by Collin et al. (61) was intended to baroreflex impairment, and hypoxia-induced activation of
test hypotheses related to symptoms of depression in ob/ob chemoreflex sensors, which increase both the sympathetic

1054 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

Obesity
Type 2 diabetes

Mechanical load
Narrowed airway
Increased fat
Weight-dependent
mass

Sympathetic activity
Oxidative stress
Inflammation
Lipolysis Sleep
Intermittent hypoxia
Sleep fragmentation apnea

Sympathetic
activity Decreased
Leptin resistance chemosensitivity
Insulin resistance
Hyperglycemia Physiology-dependent
Sympathetic activity
inflammation

Figure 5 Schematic diagram depicting the interrelationship between weight-


dependent and physiological-dependent mechanisms on metabolic and cardiorespi-
ratory responses in obesity. While weight-dependent mechanisms are a function of the physical
increase in body mass or fat mass (e.g., increased mechanical load, narrowed airway), physiology-
dependent mechanisms are physiological changes coincident with obesity or diabetes which go on to
influence chemosensitivity and sleep apnea either directly or via action on sympathetic activity, inflam-
mation, or other mechanisms. Adapted, with permission, from Framnes SN and Arble DM, 2018 (107).

tone and peripheral vascular resistance (215). Emerging proximity, it has been suggested that the respiratory neurons
hypotheses postulate that pathophysiological components of of the ventral medulla establish synaptic connections with
obesity, including altered glycemic control, insulin action, the presympathetic neurons of the RVLM and generate the
and leptin signaling, contribute to the development of OSA respiratory oscillation in the sympathetic activity (147, 223,
(Figure 5 and Table 1) (107). It is possible that obese 231, 402, 406). Other brain areas are also critical to the
physiology leads to greater reductions in pharyngeal dilator circuitry regulating the coupling of respiratory and sympa-
muscle tone promoting airway obstruction during sleep or thetic activities. The neurons of the NTS receive afferents
causing central sleep apnea via decreased chemosensitivity from baroreceptors and chemoreceptors, for this reason, this
(Figure 5) (107). nucleus is considered an important integrative center to medi-
In addition to leptin’s effects on appetite and metabolic ate sympathetic and chemorespiratory response (404, 405).
functions, growing evidence suggests that leptin is able to Based on these pathophysiological aspects of the mecha-
mediate autonomic sympathetic activity (219, 295) and also nisms of respiratory-sympathetic coupling, a previous study
central breathing control (26, 29, 53, 164, 214). In obese evaluated the possibility that changes in the strength of
subjects, the excess leptin is associated with the percent- interactions between respiratory and sympathetic neurons
age of adipose tissue, suggesting that obese people have a may contribute to increasing baseline sympathetic activity in
selective leptin resistance, inhibiting its action on appetite chronic intermittent hypoxia (CIH) (Figure 6B) (239).
modulation (66). Systemic administration of leptin has been shown to
Sympathetic oscillations coupled to the respiratory activity increase c-fos expression in neurons of the cNTS (93, 95) and
have been proposed as an important homeostatic mechanism enhance phrenic burst amplitude (53). Moreover, microin-
optimizing tissue perfusion and blood gas uptake/delivery, jections of leptin in the NTS increase renal sympathetic
especially under conditions of blood gas challenges (hypoxia activity (219) and ventilation (163). Therefore, leptin can
and hypercapnia). The ventral surface of the medulla oblon- act directly on NTS cells. There is a positive correlation
gata has been implicated as the main site of synaptic between obesity and high levels of leptin in OSA patients that
interactions between respiratory and sympathetic neurons develop hypertension, which is less pronounced in nonobese
(Figure 6A) (239, 404). The ventral medulla houses the OSA patients (146, 272, 328). Therefore, it is possible that
pre-sympathetic neurons of the RVLM—the major source of CIH, peripheral chemoreflex activation, and leptin effects
excitatory inputs to the preganglionic sympathetic neurons may be correlated. This hypothesis comes from experi-
of the spinal cord that maintain baseline arterial pressure ments that evaluated the effects of leptin or l-glutamate
in adequate levels (138, 311). Due to their anatomical microinjections into the cNTS on sympathetic response

Volume 10, July 2020 1055


Leptin in the Control of Breathing Comprehensive Physiology

Table 1 Summary of Presented Evidence that Obstructive Sleep


Apnea and Its Components are Associated with Decreased Glycemic
Leptin Effects on Melanocortin/POMC
Control, Insulin Resistance, Increased Leptin, and Decreased Neurons—Linking Energy to Breathing
Chemosensitivity
The involvement of the melanocortin/POMC neurons
Model Results in mediating leptin’s action on cardiorespiratory
responses
Obstructive sleep apnea ↓ Hypoxic ventilatory response Melanocortin receptors (MC3/4R) mediate most of the
(human)
↓ Hypercapnic ventilatory response metabolic and cardiovascular actions of leptin. In the CNS,
↓ Glycemic control leptin depolarizes POMC neurons in the arcuate nucleus
of the hypothalamus (ARC) inducing the release of α-
↑ Insulin resistance
melanocyte-stimulating hormone (α-MSH) which, in turn,
↑ Leptin activates the MC3/4Rs located in several hypothalamic nuclei
Type 2 diabetes + obstructive ↓ Glycemic control as well as in other regions of the brain including the brainstem
sleep apnea (68, 144, 243) (Figure 8). Deletion of Ob-Rs specifically in
↑ Apnea-hypopnea index
POMC neurons abolished the ability of leptin to raise blood
↑ Central sleep apnea
pressure and to reduce fasting glucose and insulin levels
↑ Insulin resistance (84). Moreover, pharmacological blockade of MC3/4R com-
Sleep fragmentation ↓ Glycemic control pletely prevented the anorexic and antidiabetic effects of
↑ Insulin resistance leptin (72).
Previous experiments in anesthetized rodents showed that
↑ Leptin
leptin activates neurons in brain sites essential for breathing,
Intermittent hypoxia ↓ Glycemic control including the NTS, RVLM, and possibly the RTN, key cen-
↑ Insulin resistance tral chemosensitive areas (27). The melanocortin pathway
↑ Leptin has been identified as a putative downstream mechanism
(30). To better understand how leptin modulates the central
↓ Chemosensitivity
breathing function, Yao et al. (391) investigated the effect of
Obesity + intermittent hypoxia ↑ Insulin resistance leptin on medulla versus hypothalamus in ob/ob mice. Leptin
↓ Hypoxic ventilatory response improved the upper airway obstruction during sleep when
↓ Hypercapnic ventilatory response administered into the lateral cerebral ventricle. However,
leptin administration into the fourth ventricle had no effect
Reprinted, with permission, from Framnes SN and Arble DM, 2018 on airway obstruction during sleep, suggesting that leptin
(107). is acting on the hypothalamus instead of the brainstem to
mediate airway flow (391). It appears that upper airway
motoneurons were synaptically coupled to neurons of the
during the peripheral chemoreflex activation (58). Thus leptin DMH that were immunopositive for phosphorylated STAT3
enhances the sympatho-excitatory responses to peripheral whereas the phrenic motoneurons were synaptically coupled
chemoreflex activation through its actions on cNTS neurons to the immunopositive STAT3 neurons of the NTS. Perhaps,
involved with the processing of peripheral chemoreceptor the POMC neurons of the hypothalamus are mediating
inputs. these effects of leptin; however, this hypothesis needs to be
A recent study reinforces the hypothesis that leptin can tested.
be involved in cardiorespiratory changes related to obesity. The melanocortin/POMC neurons was previously reported
Rats fed for 12 weeks with high-fat diet (HFD) had elevated as a mediator of leptin’s effects on ventilation. In accord with
mean arterial pressure (MAP), impaired reflex bradycardia, a study, chronic (6 days) intracerebroventricular microinjec-
increased respiratory frequency, and accentuated abdom- tion of MC3/4R antagonist reduced the ventilatory response
inal motor activity during activation of the chemoreflex to hypercapnia and abolished the ability of leptin to increase
with hypercapnia (Figure 7 and Table 2) (350). Once the baseline ventilation in rats (28). In addition, the ventilatory
cardiorespiratory coupling seems to be coordinated due to response to CO2 was diminished in mice with targeted dele-
the activity of dorsal and ventral nuclei of medulla, espe- tion of POMC neurons (LepR/POMC-cre transgenic mice)
cially NTS and RVLM, it is possible that rats exposed to (28). This attenuation of chemorespiratory response is similar
HFD have altered neuronal sympathetic-respiratory activ- to the response found in mice that had a targeted deletion of
ity due to high circulating leptin levels, which certainly Ob-Rs in the brain (LepR/Nestin-cre transgenic mice). Taken
contribute to cardiorespiratory changes in HFD rats. Nev- together, this information reveals that the absence of leptin
ertheless, the mechanisms by which leptin optimized the signaling pathways as well as POMC cells in the brain impair
chemoreflex-induced respiratory response and modify the the chemosensitive response. According to this data, it was
respiratory-sympathetic coupling are still to be determined. demonstrated that obese agouti yellow mice who lack MC4R,

1056 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

(A) Respiratory-sympathetic coupling (B) Control CIH


(active l/passive E) (active l/active E)
Larger Traube-Hering waves

Peripheral afferent
information PP
(e.g., pulmonary receptors,
baroreceptors, chemoreceptors)

Brainstem respiratory network Upper airways


PN
Expiratory Inspiratory Spinal cord
Motor nuclei late-E

Respiratory muscles
Sympathetic
tSN
Rostral ventrolateral medulla

late-E
Spinal cord Blood vessels no late-E
IML Heart
AbN

l E l E
2s 2s

Figure 6 (A) Schematic model of interactions between respiratory network and sympathetic nervous system. The
brain stem respiratory nuclei generate the coordinated inspiratory and expiratory motor activities responsible to control upper airway
resistance and respiratory movements. It has been suggested that the respiratory neurons interact with presympathetic neurons of rostral
ventrolateral medulla (RVLM), generating the respiratory oscillations in the sympathetic activity. In addition to these central mechanisms,
the peripheral afferent inputs, like those from pulmonary stretch receptors, arterial baroreceptors, and peripheral chemoreceptors, may
interact with respiratory and sympathetic neurons and also contribute to respiratory-sympathetic coupling. (B) Illustration shows the pattern
of raw and integrated (∫ ) activities of thoracic sympathetic (tSN), abdominal (AbN), and phrenic nerves (PN) as well as the magnitude
of perfusion pressure (PP) of control and chronic intermittent hypoxia (CIH) rats. Note that in control rats the amplitude of AbN activity is
low, indicating that the respiratory pattern is composed of active inspiration (active I) and passive expiration (passive E). On the other
hand, in CIH rats the AbN exhibits an addition burst during the late part of expiration (late-E), indicating that not only inspiration but also
expiration are active in CIH rats (active I/active E). In addition, as a consequence of the active expiratory pattern, the tSN exhibits a
correlated peak of discharge during late-E. EXP indicates expiratory neurons; INSP, inspiratory neurons; SYMP, presympathetic neurons;
IML, intermediolateral column; PMN, phrenic motor neurons; AMN, abdominal motor neurons. Adapted, with permission, from Moraes
DJ, et al., 2012 (239).

200 90

*
breaths/min

150
breaths/min

60
fR

ΔfR

100
30

50
0
Basal 7% CO2 SD HFD
300 450
Δ DIA amplitude (%)

Δ ABD amplitude (%)

200 #
# 300 *#
100
150

0
0
SD HFD
SD HFD

Figure 7 Effect of HFD on ventilation in rats. Rats fed with high-fat diet (HFD) during
12 weeks have high variation of the respiratory frequency (fR ) and abdominal expiratory
motor activity (ABD) during hypercapnia (10% of CO2 ) compared to standard diet (SD)
fed rats. The activity of the diaphragm muscle (DIA) was similar among the groups (SD
n = 14 and HFD n = 13). * Different from SD and # different from basal condition, P < 0.05.
Adapted, with permission, from Speretta GF, et al., 2018 (350)

Volume 10, July 2020 1057


Leptin in the Control of Breathing Comprehensive Physiology

Table 2 Body Weight, Adiposity and Metabolic Parameters in Rats rats, whereas food restriction to prevent excess weight gain
Fed with Standard Chow Diet or High-fat Diet for 12 Weeks in MC4R deficient rats may attenuate the elevation in MAP
caused by CIH (85). The authors also found that MC4R
SD HFD deficiency was associated with impaired ventilatory response
to hypercapnia independently of obesity. More studies are
Final body weight (g) 437 ± 11 507 ± 12* needed to be understand the involvement of MC4R signaling
Epididymal adipose tissue 0.41 ± 0.08 0.82 ± 0.12* in mediating the blood pressure response to CIH, more studies
(g/100 g b. wt.) are needed.
Retroperitoneal adipose tissue 0.55 ± 0.10 1.77 ± 0.17*
The MC3/4Rs also are present on PVN, DMH and perifor-
(g/100 g b. wt.) nical (PeF) nuclei of hypothalamus (179) and these regions
are modulating the metabolism, cardiovascular, and breath-
Mesenteric adipose tissue 0.58 ± 0.06 1.24 ± 0.15*
(g/100 g b. wt.) ing function (106). The activation of PVN and DMH nuclei
promotes activation of phrenic nerve which, in turn, enhances
Total cholesterol (rag/dL) 62 ± 4.5 84 ± 5.3*
pulmonary ventilation (106, 210, 394). Indeed, the disinhibi-
HDL (mg/dL) 26 ± 3.1 27 ± 3.9 tion of PeF nuclei promotes an increase in MAP, phrenic nerve
TGL (mg/dL) 37 ± 1.0 58 ± 4.9* discharge, and firing rate of the chemosensitive RTN/pFRG
Glucose (mg/dL) 90 ± 7.6 112 ± 4.8* neurons of isoflurane-anesthetized rats (106, 200). However,
how leptin affects breathing via activation of the melanocortin
Kitt (%/min) 3.7 ± 0.1 1.7 ± 0.1*
system in DMH or PeF nuclei needs further investigation.
Leptin (ng/mL) 1.2 ± 0.5 6.4 ± 0.7*
VO2 (mL/(kg min)) 17.01 ± 1.63 14.88 ± 1.40
Leptin’s unique effects during early development
VCO2 (mL/(kg min)) 13.02 ± 0.79 11.36 ± 1.05
Leptin is a master regulator of feeding behavior and energy
RQ (VCO2 /VO2 ) 0.78 ± 0.02 0.76 ± 0.01
balance in children and adults via binding to Ob-Rs on
All values are presented as means ± SEM. Non-paired Student t-test neurons in the ARC in the hypothalamus. However, during
“*” different from SD group; P < 0.05 (n = 4–10/group). SD, standard early development, leptin does not influence feeding behav-
chow diet; HFD, high-fat diet; HDL, high-density lipoprotein; TGL, triacyl- ior or metabolism. Relevant to feeding behavior, the ARC
glycerol; RQ, respiratory quotient; Kitt (%/min), percentage of plasma contains two populations of neurons that express Ob-Rs:
glucose concentration decline per minute; b. wt., body weight. Total
cholesterol, HDL, TGL and glucose were measured in serum and leptin
(i) orexigenic neurons, which co-express NPY and AgRP,
in plasma. and when activated, promote feeding; and (ii) anorexigenic
Reprinted, with permission from Elsevier, from Speretta GF, et al., 2018 neurons, which express melanocortin peptides derived from
(350). POMC, and when activated, suppress feeding behavior.
In nonobese adult models, the summation of the effect of
leptin is to induce satiety and to increase energy expenditure.
exhibited attenuated ventilatory response to CO2 and have Thus, leptin decreases the activity of orexigenic neurons
a normal ventilatory response to hypoxia, suggesting that (NPY/AgRP) and activates anorexigenic neurons (POMC).
the melanocortin system appears to mediate the ventilatory In contrast, in rodents, during the first weeks of postnatal
response to hypercapnia more than hypoxia (287). development leptin binding to orexigenic neurons actually
Considering the effects of leptin on sympato-respiratory depolarizes neurons and has little effect on the membrane
coupling and the possible involvement of the melanocortin potential of POMC neurons (23).
system as a mediator of these actions, a recent study was The anorectic effect of leptin, in rodents, occurs after wean-
designed to determine the role of CNS MC4R on CIH-induced ing between 4 and 6 weeks of postnatal life, at which time
hypertension and ventilatory responses to hypercapnia (85). the membrane electrical properties of the NPY and POMC
Experimentally induced CIH during sleep in animals is neurons, as well as their axonal projections, are fully devel-
commonly used as a model of OSA that occurs in humans. oped (42, 137). Baquero et al. (23) concluded, from a series
Similar to humans, experimental animals exposed to CIH of well-designed experiments, that acquisition of functional
have augmented chemoreceptor reflexes (175, 289, 290), ATP-sensitive potassium channels in NPY/AgRP expressing
sympathoexcitation, reduced cardiac parasympathetic activ- neurons leads to hyperpolarization versus depolarization
ity, decreased baroreflex sensitivity, and hypertension (204, prior to 4 weeks of postnatal life in response to leptin binding
249, 403). do Carmo et al. (85) demonstrated that obese to Ob-Rs on these neurons. Barquero and colleagues also
MC4R knockout rats exposed to 7 to 18 days of CIH had reported, that prior to 4 to 6 weeks of postnatal age in rodents,
reduced food intake with no change in plasma glucose, leptin, leptin functions more like a trophic factor, depolarizing NPY
or insulin concentrations. Although, lean pair-weight-MC4R neurons to shape the neuronal circuit that forms the intricate
deficient rats exhibited attenuated arterial pressure responses network that regulates feeding behavior and metabolism. In
to CIH. Thus, endogenous CNS MC4R does not appear to the absence of leptin, this neuronal circuit does not properly
play an essential role in elevating MAP during CIH in obese develop as illustrated in the figure created by Bouret and

1058 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

Hypothalamic projections to brainstem


Fat cells
Leptin CNS action
Leptin
Brainstem
LC
POMC NTS
Hypothalamus DMV M

N
C3

PB
/4
R
KF
NPY/AgRP POMC NA
7N BötC Pre-BötC VRG
Arcuate nucleus
RVLM
α-MSH RTN/pFRG

4R
3/
C
M

PVN
Heart and
Food intake Diaphragm
blood vessels
RSNA AP Abdominal
Ventilation

Obese
Increase in food intake
Increase in body weight
Reduced HCVR
LepR/POMC-cre Leptin resistance

Figure 8 Schematic diagram depicting of main central nervous system (CNS) sites of
leptin action to modulate ventilation including proopiomelanocortin (POMC) neurons.
Upper panel: Leptin activates leptin receptors (LRs) in the ARC nucleus causing inhibition of neuropep-
tide Y/agouti-related peptide (NPY/AgRP) and depolarization of POMC neurons leading to the release
of alpha melanocyte-stimulating hormone (α-MSH) which, in turn, activates the melanocortin 3 and 4
receptors (MC3/4R) mainly in the arcuate nucleus (ARC) as well as in the others nuclei located in brain-
stem such as nucleus of the solitary tract (NTS) and rostral ventrolateral medulla (RVLM). The evoked
responses induced by leptin in the CNS are a reduction in the food intake, increase in renal sympa-
thetic activity (RSNA), increase in arterial pressure and in the ventilation. Lower panel: LepR/POMC-cre
mice are obese and have reduced hypercapnic ventilatory response (HCVR). Abbreviations: AP, arte-
rial pressure; BötC, Bötzinger complex; DMV, dorsal motor nucleus of the vagus; 7N, facial nucleus;
KF, Kölliker Fuse; LC, locus coeruleus; NA, nucleus ambiguous; PBN, parabrachial nucleus; PVN, par-
aventricular nucleus of the hypothalamus; pre-BötC, pre-Bötzinger complex; RTN/pFRG, retrotrapezoid
nucleus/parafacial respiratory group; and VRG, ventral respiratory group. Modified, with permission,
from Bassi M, et al., 2015 (28).

Simerly (42) which schematically depicts the ontogeny of the growth and organ development that occurs in the fetus and
ARC projections in wild type and ob/ob mice (lack the leptin newborn; thus, in the fetus and newborn, leptin does not
gene) (Figure 9) (42). Although of normal weight at birth, lead to satiety, or other metabolic effects even though leptin
ob/ob mice exhibit hyperphagia, severe obesity, metabolic levels are higher in rodents during the first and second week
features of type II diabetes, hypogonadism, infertility, as well of postnatal life, being 2–3 times adult levels. Moreover,
as OSA (391). Thus, the absence of leptin during fetal and intraperitoneal leptin administration (1 mg/kg body weight)
early postnatal development can have profound effects on the to lean and ob/ob mice pups from 7 to 10 days of age did
development and function of the ACR of the hypothalamus, not affect milk intake, oxygen consumption, body weight, or
and other organ systems (19–21). weight of epididymal fat pad (230); intracerebroventricular
Leptin is detected in the human fetus as early as 18 weeks injection of (1 mcg) leptin also did not affect feeding behavior
gestation (168). Leptin is also produced by the placenta, and and metabolism until the mice were 3 to 4 weeks postnatal
placental leptin contributes to fetal and maternal levels, while age (230).
maternal leptin does not appear to contribute to fetal leptin
levels (17, 335). A leptin surge occurs postnatally in many
mammalian species (42), peaking between postnatal days 7 Early life exposure to undernutrition/intrauterine
and 10 in rodents (79), and increasing exponentially from growth restriction and leptin signaling
34 to 40 weeks of gestation in humans (168). This surge is It is highly likely that leptin provides trophic support for the
directly proportional to fat mass, which rapidly increases in development of the neuronal network that regulates feeding
the human fetus after 34 weeks of gestation. It is important and energy expenditure during early development. Of course,
that energy intake meets the metabolic demand for the rapid biochemical, neurophysiological, and tract tracing studies

Volume 10, July 2020 1059


Leptin in the Control of Breathing Comprehensive Physiology

P6 P12

WT Lepob/Lepob WT Lepob/Lepob
AVPV AVPV

MPN MPN

PVH PVH

ARH ARH
LHA LHA

DMH DMH

P16 Adult
WT Lepob/Lepob WT Lepob/Lepob
AVPV AVPV

MPN MPN

PVH PVH

ARH ARH
LHA LHA

DMH DMH

Figure 9 Schematic diagram showing the effect of leptin deficiency on the ontogeny
of projections of the ARH through the hypothalamus during development. Left panel
(wild-type WT) and right panel leptin deficient (Lepob /Lepob ) on postnatal day (P) P6, P12, P16,
and adults (P60). Leptin deficiency permanently disrupts the formation of projections from the arcu-
ate nucleus to each major target nucleus. The relative size of each pathway is roughly proportional to
the thickness of the lines associated with it. AVPV, anteroventral periventricular nucleus; MPN, medial
preoptic nucleus; DMH, dorsomedial hypothalamus; PVH, periventricular hypothalamic nuclei; and
LHA, lateral hypothalamic area. Leptin deficient mice are slow to develop and have a reduced number
of projections from the ARH to other key areas of the hypothalamus that regulate feeding behavior.
Adapted, with permission, from Bouret SG and Simerly RB, 2004 (42).

cannot be done in human infants to delineate exactly how Undernutrition resulting in leptin deficiency in
leptin “programs” feeding behavior during early development premature and small for gestational age infants to
and across the life span. However, several “natural experi- SGAs infants: translational perspective
ments” exist in humans from which the potential mechanism
of leptin effects during early development can be gleaned. Leptin deficiency may also occur when infants are born pre-
Spontaneous mutations in the genes that encode leptin and maturely and/or SGA, birth weight less than 10th percentile
Ob-Rs may occur, and the phenotypes of patients with these for age. Although leptin can be detected in human fetal cord
gene mutations have been described in case reports (89, 237). blood as early as 18 weeks gestation, circulating leptin levels
These patients present with hyperphagia starting within days remain quite low (median 0.61 ng/mL; range 0.41–6.5 ng/mL)
of birth leading to severe early-onset obesity with a rapid until 34 weeks gestation at which time leptin levels markedly
and dramatic increase in body weight within months of birth, increase (median 3.5 ng/mL; range 0.7–39 ng/mL) by term
hypogonadotropic hypogonadism, and increased risk of infec- gestation (37–40 weeks) (168). In human infants, similar to
tion. Those patients who were homozygous for the mutation adults, the rise in leptin levels is proportional to the increase
in the leptin gene had low levels (<1 ng/mL) of circulating in fat mass which rapidly increases from 34 weeks to term
leptin (89, 237). Daily administration of recombinant leptin gestation (Figure 10), and sexual dimorphism occurs early
to these patients improves many of the metabolic, feeding in gestation with females having higher leptin levels than
and endocrine problems (274). Mutations in human genes term male infants after controlling for fat mass (168). Infants
coding for Ob-Rs also lead to severe early-onset obesity, who are SGA, particularly those who have not grown well
with a rapid increase in body weight by 4 months of age, and in utero and have intrauterine growth restriction (IUGR)
associated comorbidities throughout life (237). However, in have less fat mass, and thus lower levels of leptin than their
those patients with nonfunctioning Ob-Rs, exogenous leptin appropriate for gestational age (AGA) counterparts. Male
is not an effective treatment. infants who are both premature and IUGR have the lowest

1060 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

45 remain quite low for many weeks when many of the organs
are developing, specifically the brain, kidney, pancreas, and
40
cardiovascular system. Obesity and metabolic comorbidities
35 observed in ob/ob mice are similar what is observed in
models and infants who are exposed to undernutrition or
30
overnutrition during fetal development; thus comorbidities
Leptin (ng/mL)

25 (MetS, hypertension, and cardiovascular disease) that occurs


in adulthood in former IUGR infants could be explained by
20 insufficient leptin levels during organogenesis.
15 Leptin is expressed by the mammary gland and is in fat
droplets of freshly expressed breastmilk (184, 347); it is in
10 lower concentrations in breastmilk from mothers who deliver
5 prematurely and is even lower in the breastmilk from mothers
of IUGR infants (90, 258). Resto et al. (302) did not detect
0 leptin in infant formula or human milk that had either been
15 20 25 30 35 40 45 sterilized or pasteurized; thus, donor milk is not a significant
Gestational age (week) source of leptin. Of note, leptin was detected in commercially
available bovine formulas prepared in Spain (188), but this
Figure 10 Serum leptin concentrations in cord blood has not been a consistent finding reported by others using
(ng/mL) according to gestational age (weeks) in fetuses
and newborns. Blue symbol represents values obtained from fetuses
infant formulas from other manufactures. Thus, SGA and
and newborns with normal growth; red circles, represents values from IUGR infants, especially those who are born less than 34
fetuses and newborns with IUGR. Modified, with permission, from weeks of gestation and who already have undetectable leptin
Jaquet D, et al., 1998 (168).
levels at a key time during organ development, do not receive
leptin from human milk or commercially available premature
leptin levels (<1 ng/mL) of all other infant groups, and the formulas or human milk fortifiers. Ob-Rs are present in the
levels can remain low for many months after birth (96, 371) stomach of newborn animals and human adults (51, 348).
and reviewed in Ref. 122. Hellgren et al. (150) longitudinally Exogenously administered leptin to nursing dams increases
followed leptin levels in infants born less than 28 weeks milk leptin levels and circulating leptin in nursing rat pups
gestation and reported that leptin levels were consistently low (323). Human recombinant leptin is available and FDA
until infants reached a weight of 1900 g. These low leptin approved to be used in patients with congenital leptin defi-
levels in IUGR infants are similar to levels in patients with ciency. The other populations who may benefit from leptin
mutations in the leptin gene (89, 237). supplementation are extremely premature infants and infants
Postnatal weight gain is always less than in utero weight who are IUGR. Leptin supplementation for at-risk infants
gain, and thus, leptin levels remain quite low at a time should be considered (354). Studies to determine efficacy and
when trophic influence of leptin on organ development is safety of leptin supplementation to reduce the risk of adverse
quite important (44). For example, infants born at 28 weeks, metabolic and cardiovascular outcomes in this population
4 weeks later, at postmenstrual age of 32 weeks, median lep- should be considered.
tin levels were 0.01 ng/mL (min–max of 0.00–2.40 ng/mL)
which did not differ from the levels at birth. On the other
hand, infants born at 32 weeks of gestation had median leptin Early Life Exposure to Overnutrition
levels of 0.63 ng/mL (0.00–6.48 ng/mL) (150). Similar to
infants who have congenital leptin deficiency, infants who HFD/obesity during pregnancy modifies leptin
are born IUGR have increased risk of developing metabolic POMC signaling pathways in offspring
syndrome (MetS) with hyperphagia, obesity, insulin resis- Similar to undernutrition, overnutrition during fetal or early
tance, hypertension, and cardiovascular disease reviewed in postnatal life can also cause lifelong changes to metabolic
Ref. 174. While genetic mutations in the leptin gene and systems often leading to obesity in adulthood (261, 310).
the Ob-R are rare, being born prematurely and IUGR is These metabolic changes also increase the susceptibility of
not, occurring in 10% to 15% of all pregnancies, American developing obesity-associated comorbidities such as diabetes,
College of Obstetrics and Gynecology. As clinical care of cardiovascular disease, and suboptimal mental health (36).
high-risk pregnancies continues to improve more infants Studies using animal models of maternal undernutrition, and
with IUGR will be born, and with the rapid advances in overnutrition show that changes in nutritional environment
neonatology, these infants will survive with low leptin levels may increase susceptibility to obesity in adult offspring
at a critical period of organ development. Moreover, the by compromising the proper development of hypothalamic
limit of viability continues to decrease, such that infants born pathways regulating appetite (41, 42). Early overnutrition
between 22 and 23 weeks gestation are surviving and growing increased body weight, susceptibility to obesity, hyper-
into childhood. Leptin levels are undetectable at birth and leptinemia, and leptin resistance later in life (54, 125).

Volume 10, July 2020 1061


Leptin in the Control of Breathing Comprehensive Physiology

Leptin regulates energy balance and exhibits neurotrophic


Fetal/neonatal Excessive adiposity Pregnancy
effects during critical developmental periods (178, 321). hyperleptinaemia (overnutrition) and lactation
How HFD and high circulating leptin levels alter metabolic
function appear to be, in part, associated with a reduced abil- Leptin
resistance and
ity of POMC neurons to respond to leptin, which is critical for impaired neurotrophic Selective leptin resistance
the regulation of POMC and other hypothalamic regulatory action
in the hypothalamus
neuronal projections (125, 143). One mechanism by which Metabolic SNS Offspring
leptin activates POMC neurons is through phosphorylation of action activity phenotype
STAT3 and downstream signaling. Leptin induced phospho- Altered leptin
signalling in
rylation of STAT3 is attenuated in models of early overnutri- Hyperphagia
hypothalamus Hypertension
and obesity
tion (125). POMC neurons of diet-induce-obesity (DIO) mice in adulthood

are resistant to activation of STAT3 phosphorylation by lep-


Figure 11 Overnutrition and excessive adiposity during pregnancy
tin. Indeed, these DIO animals had overexpression of both and lactation alter the fetal programming leading to obesity and hyper-
leptin receptors selectively in hypothalamic POMC neurons tension. The altered leptin signaling in the hypothalamus promotes a
and the SOCS3 mRNA in the arcuate (ARC) nucleus of DIO selective leptin resistance in which the anorexic effects of leptin are lost,
whilst the pressor effect of leptin is enhanced. Adapted, with permission,
mice (117). from Taylor PD, et al., 2014 (364).
Hypothalamic systems that regulate appetite may be per-
manently modified during early development in response to
suboptimal nutritional states. Adult offspring of rodents fed exposure to high leptin levels in the immediate postnatal
an obesogenic diet prior to and throughout pregnancy and period, from increased fetal adiposity, leads to selective
lactation have hyperphagia and an increased adiposity (178). leptin responsiveness, hypertension, and altered myocar-
Juvenile (Day 30) offspring of obese dams were resistant dial function. The increase in low: high-frequency ratio of
to the inhibitory effects of leptin on food intake and body heart rate variability (marker of sympathetic tone) in 30-day
weight, at a time when serum leptin levels were normal; the leptin-treated animals is indicative of heightened sympathetic
leptin resistance was still present in adulthood (Day 90) when efferent tone (320). Experiments using transgenic technology
the animals were obese and hyperleptinemic (178). Offspring to restore MC4R in the PVN of Mc4rKO mice showed that
of obese dams have amplified and prolonged neonatal lep- these mice develop higher blood pressure than Mc4rKO or
tin surge, which is accompanied by elevated leptin mRNA wild-type (WT) mice, suggesting that neonatal hyperleptine-
expression in their abdominal white adipose tissue. At postna- mia due to maternal obesity induces persistent changes in
tal day 30, before the onset of hyperphagia, in these animals, the central melanocortin system, thereby contributing to
serum leptin levels are normal, but leptin-induced appetite hypertension in offspring (322).
suppression and phosphorylation of STAT3 in the ARC are Respiratory function can also be altered in offspring of
attenuated (41, 178). The level of AgRP immunoreactivity dams who have been feed a dyslipidaemic (DLP) diet during
in the PVN, which is derived from neurons in the ARC, is pregnancy and lactation. At postnatal day (P)30, the offspring
developmentally dependent on leptin and is also diminished of DLP rats had increased circulating levels of triglycerides
in these animals (178). and cholesterol levels. At P90, DLP offspring exhibited
Roberts et al. (310) have characterized the synaptic devel- higher MAP and augmented cardiorespiratory chemoreflex
opment of POMC neurons in the ARC, their sensitivity to response to hypercapnia and hypoxia (140). Taken together,
leptin, and the impact of early overnutrition on the develop- these data suggest that maternal dyslipidemia alters cardiores-
ment of these neurons. Accordingly, leptin’s overall effects on piratory reflex in offspring and may be a predisposing factor
POMC neurons are excitatory during late development and for hypertension and disordered breathing in adulthood.
into adulthood (256). In early development, chronic postna-
tal overnutrition (CPO) promotes an increase in the ampli-
tude of postsynaptic inhibitory currents of POMC neurons, Leptin and Peripheral Chemoreceptors
while during late development, CPO decreases the amplitude
of these currents and the excitatory effect of leptin on these Peripheral chemoreceptors
currents. Taken together, these studies suggest that postna- The peripheral chemoreflexes contribute significantly to the
tal overfeeding alters the postsynaptic development of POMC cardiorespiratory responses to hypoxia, hypercapnia, and
neurons and induces long-lasting leptin resistance in ARH- acidosis by altering respiratory amplitude and frequency, and
POMC neurons. by activating the sympathetic nervous system. The peripheral
Moreover, maternal diet-induced obesity is associated chemoreceptors include the aortic bodies, and in particular,
with cardiovascular dysfunction in male offspring during the CBs that modulate respiration and sympathetic output.
adulthood (37, 320, 364) (Figure 11). These offspring have The CBs, the most vascularized organs in the body, are
cardiac hypertrophy and impaired systolic and diastolic func- located bilaterally in the bifurcation of the carotid artery
tion. Interestingly, these cardiac changes occur independently and sense arterial PO2 (PaO2 ), arterial PCO2 (PaCO2 ), pH,
of differences in the bodyweight of the offspring (37). The and temperature. They are responsible for the greatest part

1062 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

of the hyperventilatory response during hypoxia and con- activity. The CB also contains glial-like type II cells that enve-
tribute to the hyperventilation that accompanies respiratory lope type I cells. Although originally thought to only play a
or metabolic acidosis (130). In response to hypoxia, hyper- supportive role (130), new data from several studies show that
capnia, and acidosis, type 1 CB cells—the chemoreceptor type II cells have many important functions in the CB. They
unit of the CB—release neurotransmitters/neuromodulators contribute to neurogenesis in vivo in response to prolonged
that bind to the carotid sinus nerve, thereby modulating its hypoxia, contribute to paracrine signaling, and to purinergic
firing rate (130). CSN activity is integrated into the NTS catabolic pathways by integrating the sensory output of the
in the brainstem (Figure 2) (120) output to neurons that CB during chemotransduction (259, 270, 284, 285).
innervate the muscles of respiration (130), and to the RVLM Besides its substantial role in the control of ventilation,
that output to the sympathetic nervous system that regulates it has been proposed that the CB is also a metabolic sen-
blood pressure (220). sor; implicated in the control of energy homeostasis (65,
Despite the growing knowledge as to how several stimuli 182, 183) and more recently in the regulation of peripheral
activate the CB, no consensus has been achieved as to how insulin sensitivity and glucose homeostasis (63, 65, 306,
the CB senses the classical CB stimulus, low arterial O2 , 315). Data from previous work performed, in cats (6–8) and
and its molecular identity and how O2 sensing translates into dogs (182, 183), and more recently in humans (381), sup-
increased chemosensory activity and to a systemic cardiores- ported a role for the CB in the counter-regulatory responses
piratory response (297). However, it is generally accepted to hypoglycemia. However, from the conclusions based
that the stimulus-secretion coupling in type I cells during on the results of the experiments published by Pardal and
CB activation by hypoxia triggers a series of molecular Lopez-Barneo in 2002 (269), the hypothesis that the CB has
and cellular responses that are schematically depicted in glucose-sensing properties was born. Nevertheless, while
Figure 12, and outlined here: (i) O2 -sensing at an O2 -sensor, many agree that the CB is involved in the counter-regulatory
(ii) activation of coupling mechanisms with K+ channels, responses to hypoglycemia, the hypothesis that the CB
(iii) change in kinetics of these K+ channels resulting in a is a glucose sensor is often refuted; recently reviewed by
decrease in their opening probability, (iv) cell depolarization, Conde et al. (64); thus, the sensitivity of the CB to hypo-
(v) activation of voltage-operated channels, (vi) Ca2+ entry glycemia remains a hot topic in the field and with no clear
and increase in intracellular free Ca2+ , (vii) activation of consensus.
exocytosis and neurotransmitter release (129, 130). More recently, results from Conde and co-workers (306)
The type I cells in the CB contain catecholamines showed that other metabolic stimuli, such as insulin activate
(dopamine and norepinephrine), serotonin, ACh, GABA, CB chemoreceptors: They report that (i) insulin receptors are
neuropeptides (substance P and enkephalins), adenosine, and present and undergo phosphorylation in response to insulin
ATP; for a review see Refs 62, 185. All these neurotransmit- in the whole CB, (ii) insulin increases the levels of [Ca2+ ]i
ters/neuromodulators, as well as agonists and antagonists of in type I cells and induces neurosecretion of dopamine and
their receptors, are capable of modifying carotid sinus nerve ATP from the CB, and (iii) insulin administered to adult

ANG ll Leptin Insulin Respiratory


PO2 responses
K+ Channel
PCO2

CSN Cardiovascular
activit responses
y
[Ca2+]

Renal responses
Carotid body
Carotid body Type I cell

Neuroendocrine
responses

Figure 12 Schematic representation of some stimuli that activate carotid body (CB)
chemoreceptors and the responses elicited by the carotid body. Decreased arterial oxygen
pressure (PO2 ), increased carbon dioxide arterial pressure (PCO2 ), angiotensin II (ANG II), leptin, and
insulin are examples of stimuli that activate the CB. In general, these stimuli originate type 1 CB cell depo-
larization, increase in intracellular Ca2+ and the release of neurotransmitters that act on the CB sensitive
nerve, the carotid sinus nerve (CSN), to increase its activity aiming its integration at the central nervous
system level to produce respiratory, cardiovascular, renal, and endocrine responses.

Volume 10, July 2020 1063


Leptin in the Control of Breathing Comprehensive Physiology

rodents increases ventilation in a dose-dependent manner leptin replacement in ob/ob mice, respiratory rate decreased
during an euglycemic clamp; an effect that was abolished between 11% and 13%, whereas respiratory rate was unaf-
with bilateral denervation of the CSN (306). Also, the same fected in vehicle-treated ob/ob mice; and (iii) hyperoxia
authors propose that the CBs are involved in the genesis decreased tidal volume by 13%, with this component of
of peripheral insulin resistance and deregulation of glucose the hypoventilatory response to hyperoxia being absent
metabolism; in animals who are fed a hypercaloric diet, in +/+ or vehicle-treated mice. Taken together these data
the CB is overactivated, and metabolism and hemodynamic supported a direct or indirect effect of leptin on CB function
responses are dysregulated; these altered effects, induced by in the hypoventilatory response in normoxic to hyperoxic
hypercaloric diet, were prevented or reversed after chronic transitions (136). Seven years after Groeben’s publication
denervation of the CSN (306, 313, 315). From these data, in 2004 (136), Porzionato and co-workers (288) reported
Conde et al. (65) proposed that hyperinsulinemia, induced the presence of leptin and leptin receptor isoforms in type
by hypercaloric diets, is one of the key drivers for CB over- I, but not in type II, chemoreceptor cells of both rat and
activation. However, obese animals with insulin resistance human CB; approximately 40% of human CB type I cells
have a greater increase in spontaneous ventilation, ischemic were positive for leptin immunoreactivity with more than
hypoxia-induced hyperventilation, CB weight, and tyrosine 50% being immunopositive for all leptin receptor isoforms
hydroxylase expression within the CB than lean animals of which 30% being reactive for the isoform Ob-Rb. Cor-
with hyperinsulinemia and insulin resistance (306), sug- roborating the findings reported by Porzionato et al. (288),
gesting the existence of an obesity-related factor, such as Messenger et al. (228) found that rat CB cells co-expressed
leptin, may contribute to CB stimulation when metabolism is immunoreactivity for Ob-Rb and tyrosine hydroxylase
dysregulated. (marker for type I cells), indicating that this receptor is
In addition to obesity, OSA is another chronic disorder present in type I CB cells. More recently, in 2019, Caballero-
associated with increased CB activity and altered hypoxic Eraso et al. (46) reported that Ob-Rb immunoreactivity is
ventilatory responses (131, 250, 275, 303); patients with present in approximately 74% of mice CB type I cells. Taken
OSA are often obese (22, 39, 387). Among the several risk together, these results promoted the idea that the ventilatory
factors that can contribute to the development of OSA, effects of leptin are, at least in part, mediated by the CB
obesity plays an important role in its pathogenesis. Approx- chemoreceptors.
imately 40% of obese individuals also present with OSA, Further supporting the role of leptin in modulating the
and approximately 70% of individuals with OSA are obese contribution of the CB to ventilation, leptin given to Wis-
(74, 375). More specifically, visceral obesity is common in tar rats either intravenously (262, 305) or in the carotid
subjects with OSA and is positively correlated with apnea artery close to the CB, dose-dependently increased baseline
index (341); circulating leptin is increased in OSA patients ventilation and ventilation in response to occlusion of the
(165, 375), and correlate with OSA severity (165). Thus, it common carotid artery (including ischemic in the CB) (305).
can be postulated that part of the increased CB activity and Similarly, in C57BL/6J mice, subcutaneous infusion of lep-
altered hypoxic ventilatory responses in patients with OSA tin (120 μg/day) increased minute ventilation and hypoxic
and obesity might involve a direct effect of leptin on CB ventilatory response that was abolished by CB denervation
activity. (46). These data, thereby, confirmed the contribution of
CB to leptin effects on basal ventilation and in response to
acute hypoxia. Lastly, Ribeiro et al. (304) tested the acute
The role of leptin in acute peripheral chemoreceptors effect of leptin on basal ventilation in rats submitted to
responses to hypoxia and hypercapnia CSN denervation and showed that the CB contributed with
Indubitable evidence supports the role of leptin in the central approximately 30% to baseline ventilation. On the other
control of breathing with classically experiments published hand, chronic administration of leptin for 7 days (60 μg/day)
by O’Donnell et al. in 1999 showing leptin replacement in to adult rats did not change resting respiratory parame-
(ob/ob) leptin-deficient mice (260, 363) reversed baseline ters but did increase hypoxic ventilatory response (399),
hypoxemia and hypercapnia. However, it was only in this suggesting that acute and chronic effects of leptin might
century that leptin’s action on peripheral chemoreceptors be mediated by different signaling pathways and thereby
was first explored (136). In this seminal study, Groeben and modifying the contribution of the CB to the response. Apart
co-workers (136) assessed CB function, as the % decrease from contributing to basal ventilation and to responses
in respiratory parameters (respiratory rate and tidal vol- to acute hypoxia, leptin seems to be involved also in the
ume) in normoxic to hyperoxic transitions (50, 76, 189), responses to acute hypercapnia, as (ob/ob) mice showed
in wild type (+/+) and leptin-deficient (ob/ob) mice with significant reductions in hypercapnic ventilatory sensitivity
and without leptin supplementation. They suggested, for relative to +/+ homozygotes mainly due to an attenuated
the first time, that leptin may be contributing to peripheral tidal volume (362).
control of breathing. They found that (i) hyperoxia decreased Apart from the apparent consensus on the contribution
respiratory rate by 10% from baseline in +/+ mice, but of the CB to the effects of leptin on spontaneous baseline
had no effect on respiratory rate in ob/ob mice; (ii) after ventilation, role of CB in mediating leptin effects during acute

1064 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

hypoxic and hypercapnic responses are far from being con- The role of leptin in sensitization of peripheral
clusive. For example, Ribeiro et al. (305), using a superfused arterial chemoreceptors in chronic conditions
ex vivo preparation of the rat CB, found that leptin increased
basal CSN electrical activity in rats without affecting the While the role of leptin on the peripheral chemoreceptor
hypoxic response whereas Caballero-Eraso et al. (46), using responses to hypoxia and hypercapnia is inconsistent (46,
adult mice in vivo preparation, found that leptin infused 262, 291, 305, 342), a consistent observation is that condi-
intravenously augmented both baseline activity of the CSN tions leading to hyperleptinemia, such as OSA and obesity,
and activity in response to acute hypoxic exposure. Leptin- leptin signaling in the CB augments spontaneous ventilation
induced increase in hypoxic chemosensitivity of the mice and hypoxic ventilator responses (165, 280, 325).
CSN, using an in vivo preparation, had also been reported
by Shirahata et al. (342). In these mouse experiments, leptin Chronic intermittent hypoxia
(25–100 ng/mL) perfused for 5 min did not change baseline
CSN activity but did increase CSN activity in response to CIH, characterized by cyclic hypoxic episodes of short
hypoxia, an effect that was blocked by the nonspecific tran- duration followed by normoxia, occurs in patients with OSA.
sient receptor potential (Trp) channel blockers SKF96365 Several well-designed experiments in animals implicate the
and 2-APB (342). overactivation of the CB as the genesis of OSA-mediated
The different effects of leptin on the CB responses to hypertension, due to increased activity of the sympathetic
hypoxia between the ex vivo versus in vivo preparations sug- nervous system (104, 275, 276, 303). Moreover, patients with
gests that the potentiation of CSN activity in acute hypoxia OSA have increased peripheral CB drive (for a review see
promoted by leptin can be due to other factors than the direct Ref. 65). The increased CB drive with heightened sympa-
action of leptin within the cells of the CB or its signaling thetic activity is evidenced by the increase in tidal volume
pathways within the organ, as the action on specific type of at baseline (207) and augmented ventilatory and cardiovas-
cells within the CB or in the efferent pathway that innervates cular reflex responses induced by acute hypoxia (119, 250,
the CB, which is absent in the ex vivo preparation. Species 349) observed in these patients. Among the several factors
differences might also explain the diverse effects of the leptin that have been proposed to be responsible for the increased
on the CB hypoxic responses; in the CB from mice, leptin sympathetic nerve activity in cardiovascular and metabolic
potentiated CSN responses to acute hypoxia (46, 342) but not abnormalities are hyperinsulinemia (191–193, 300) and
in the CB from rats (305). hyperleptinemia (149, 294). Indeed, CIH in rats increased
Leptin directly affects molecular and cellular response circulating leptin levels (227, 228, 262) concomitantly with
of the type I cell. Acute application of leptin (200 ng/mL) increased sympathetic tone, increased minute ventilation, and
to isolated type I cells from the rat, increased intracellu- increased hypoxic ventilatory responses (262).
lar calcium (291) and BKCa currents, but failed to alter In addition to the increased leptin levels induced by CIH,
the resting membrane potential or modify anoxic/acidic CIH also promotes visceral adipose tissue dysfunction in
depolarizations (291), suggesting that leptin receptors are both lean and obese animals (245, 314). Therefore, it can be
functional on type I cells but that their acute activation does hypothesized that CIH induces upregulation of leptin pro-
not alter chemosensory properties. On the other hand, also duction by adipose tissue; elevated circulating leptin levels
using isolated type I cells from the rat, Ribeiro et al. (305) induces CB sensitization thereby augmenting sympathetic
reported that leptin (40 ng/mL) was unable to increase Fura-2 tone and hypoxic ventilatory responses. In support of this
ratio in rat CB isolated type I cells (i.e., intracellular Ca2+ hypothesis, adult rats exposed to 8 h of CIH have altered lep-
levels) both at resting and in response to acute hypoxia. tin protein expression and leptin signaling pathways within
These differences in calcium responses between the two the CB (227, 228). Indeed, CIH increased leptin, ERK1/2,
studies may be explained by the fivefold increase in leptin and Fra-1/2 immunoreactivity within CB type I cells (227,
concentration in the superfusate used in experiments reported 228), an effect attributed to activation of the leptin signal-
by Pye et al. (291) in comparison to that used in experiments ing cascade. Similarly, immunoreactivity for downstream
reported by Ribeiro et al. (305). Consistent with lack of signaling molecules (ERK1/2 and Fra-1/2) induced by CIH
leptin-induced changes in intracellular response reported by is also observed in CB type I cells (227, 228). Moreover,
Ribeiro et al. (305), leptin also did not induce secretion of 8 h of CIH also increased protein expression of leptin, the
catecholamines from the whole rat CB (262), but did induce short form of the leptin receptor (Ob-R100 kDa) and SOCS3
adenosine release (305), an excitatory mediator involved in while protein levels for the long form of leptin receptors
the responses of CB to hypoxia. Additionally, it should be (Ob-Rb) protein were decreased (228), suggesting that leptin
considered that acute leptin administration (291, 305, 342) produced via autocrine/paracrine mechanisms or leptin from
might have different effects on the whole CB in vivo via other sources increased excitation of CB cells and modulates
activation of different leptin signaling pathways, and there- CB chemosensitivity in CIH conditions.
fore produce different effects on spontaneous ventilation and The downstream effects mediated by CIH on leptin pro-
hypoxic ventilation responses in situations associated with duction and activation of leptin signaling pathways within the
chronic exposure to high levels of leptin. CB are likely to differ between the experimental paradigms

Volume 10, July 2020 1065


Leptin in the Control of Breathing Comprehensive Physiology

of CIH related to time, duration and level of hypoxia. For suggest an interaction/integration of different pathways
example, CIH for 7 and for 95 days decreased leptin protein within the CB, which in turn may influence chemoreceptor
levels, increased the long-form leptin receptor (Ob-Rb) activity.
expression without changing the levels of protein expression
of the short form leptin receptor (Ob-R100) (229) in the
Obesity
CB. These effects on the abovementioned proteins differed
from what was described after 8 h of CIH (227, 228). CIH Pathological weight gain contributes significantly to morbid-
for 8 h increased protein expression for leptin, Ob-R100 ity and mortality worldwide due to its comorbidities, which
kDa and SOCS3 while protein levels for Ob-Rb decreased are thought to be responsible for 1 in 5 deaths in the world
(228). (128, 386), Obesity comorbidities include cardiovascular
The upregulation of Ob-Rb isoform of the leptin recep- disease, type 2 diabetes (T2D), hypertension, and some
tor may be explained by a feed-forward mechanism that cancers (386) and play a key role in the development of OSA
produces CB overactivation contributing to sympathetic and obesity hypoventilation syndrome (OHS), two important
nervous system activation and increased hypoxic ventilatory breathing disorders. The pathophysiology underlying the
responses. In fact, protein expression for leptin receptors generation of obesity and obesity-related illnesses are not
within the CB is increased in animal models of obesity and completely understood, but it is generally believed that the
insulin resistance (305), conditions known to be associated sympathetic nervous system plays a role in the generation of
with hyperleptinemia. both obesity and obesity-related illness (191, 192, 365), and
The duration of CIH affects which leptin signaling path- several lines of evidence suggest a link between breathing
ways will be activated within the CB with pSTAT3 protein disorders and altered peripheral control of sympathetic activ-
levels being significantly decreased in the homogenates of ity (110, 299), supporting the hypothesis that CB insulin/and
the CB after 7 days of exposure to CIH, but not after 95 days, or leptin action is contributing to the CB response (63, 305,
whereas SOCS3 protein levels were elevated after 7 days and 306). Trayhurn and Wood in 2004 (366, 367) suggested
pERK1/2 was elevated after 95 days of CIH (229). Taken that obesity-related white adipose tissue hypoxia as the key
together these data support the notion that leptin signaling underlying mechanism triggering adipose tissue dysfunction.
in the CB is dependent on the intensity and chronicity of Since then, several studies have been undertaken both in in
the stimuli. The summation, however, of the effects of CIH, vitro and in animal models (99, 366, 367, 393), with only a
regardless of the signaling pathway involved, is that CIH very limited clinical investigation to prove this concept (132,
increases circulating leptin levels, which then directly acti- 154, 377). Hypoxia does develop within the adipose tissue as
vates CB type I cells leading to an increase in CSN activity the tissue mass expands and the reduction in interstitial PO2
(46, 305, 342); thus, the increased CB chemosensitivity in induces a pro-inflammatory response (115, 271, 366). The
models of CIH is mediated by leptin. secretion IL-6, TNFα adipokines are upregulated by hypoxia
Several lines of evidence suggest that leptin signaling in (115, 366). In addition to inducing a pro-inflammatory
the CB may also be regulated by the angiotensin system. response, local hypoxia promotes insulin resistance in fat
Angiotensin II (ANG II) promotes leptin production and cells (217, 301, 392), and stimulates the secretion of leptin,
secretion (177, 343), ANG II type 1 receptors (AT1Rs) are even with a normoxic systemic PaO2 (154, 366). Although
expressed within the CB, and ligand binding to ANG II leptin contributes to the control of breathing in obesity, as
receptors in the CB increases CSN activity (5); ANG II the administration of the hormone reverses the hypoxia and
also mediates CB overactivation in sympathetic mediated hypercapnia commonly encountered in animals with absence
diseases, such as chronic heart failure (HF) (203, 330). Both of the functional hormone (260, 363), the mechanisms by
captopril (an angiotensin-converting enzyme inhibitor) and which obesity affects hypoxic ventilatory responses and
losartan (blocks AT1Rs) are commonly used to treat hyper- the contribution of the peripheral chemoreceptors to these
tension and some types of congestive HF in humans. Using mechanisms were not clearly understood until very recently.
these drugs, Moreau et al. (241), investigated the effects of It was found that the effect of leptin in increasing spon-
ANG II blocking on leptin and leptin signaling in the rat taneous and ventilation in response to ischemic-hypoxia
CB. They reported that 3 days of treatment with captopril, induced by carotid artery occlusion and augmenting CSN
abolished the increase in plasma leptin levels induced by 8 h activity was blunted in animals submitted to 3 weeks of HFD
of CIH; whereas, losartan treatment had no effect on plasma (305). Associated with these effects, the CBs from these
leptin levels (241). Additionally, both captopril and losartan animals exhibited 35% overexpression of leptin receptors,
abolished the CIH-induced increased protein expression for suggesting a feed-forward mechanism aiming to increase CB
leptin, Ob-R100, and the CIH-induced changes in pSTAT3 activity or the saturation of Ob-R caused by diet-induced
and (pERK1/2) protein levels in the CB (241), suggesting hyperleptinaemia; a similar effect has been described for the
that ANG II can regulate leptin action in the CB as a result of CNS of animals exposed to HFD (316). Taken together the
CIH. These findings, taken together with the previous obser- authors postulated that although leptin might be involved in
vations that ANG II modifies CB chemosensitivity to increase triggering CB overactivation in initial stages of obesity and
sympathetic tone and hypoxic chemosensitivity (202, 330), dysmetabolism, resistance to leptin signaling and blunting of

1066 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

responses develops in animal models of MetS, which is in Hypercaloric diets,


agreement with the lack of increase in minute ventilation in obesity,
chronic intermittent hypoxia
rats exposed to hyperlipidic diets for 8 (399) and 16 weeks
(296). In parallel with the study of Ribeiro et al. (305), Yuan
and co-workers (399) showed that obese Zucker rats, that lack Adipose tissue dysfunction
the gene that codes for Ob-R, have decreased hypoxic ven-
tilatory responses, and that chronic administration of leptin Increased circulating leptin
to Zucker lean littermates did not alter baseline ventilation,
but produced a heightened hypoxic ventilatory response;
effects abolished by CSN denervation. Additionally, the Carotid body
expression of pSTAT3, as well as putative O2 -sensitive K+ overactivation
channels including TASK-1, TASK-3, and TASK-2 within
the CB was reduced in obese Zucker rats, effects also abol-
ished by CSN denervation (399). Moreover, chronic leptin Sympathetic
Spontaneous nervous system activity
administration increased the expression of pSTAT3 and ventilation
TASK channels in Zucker lean littermates, suggesting that Hypoxic ventilatory
response
leptin signaling in the CB via pSTAT3 and TASK channels
contributes to the increased hypoxic ventilatory responses in Hypertension Insulin resistance
obesity. Confirming the role of the CB in mediating leptin
effects on spontaneous ventilation and hypoxic ventilatory
Figure 13 Diagram representing the involvement of carotid
responses in obesity, Caballero-Eraso et al. (46) reported
body in leptin effects in the control of breathing and on
that obese db/db mice transfected with Ob-R in the CBs sympathetic overactivation that participates in genesis of
had increased minute ventilation during wakefulness and insulin resistance and hypertension. Hyperleptinemia produced
sleep and increased the hypoxic ventilator response. The by adipose tissue dysfunction induced by dysmetabolism, obesity
and/or by chronic intermittent hypoxia originates an increase in
authors speculated that leptin signaling in the CB could carotid body sensitization that is on the basis of increased spontaneous
maintain respiratory function in severe obesity by preventing ventilation, increased hypoxic ventilatory response, and augmented
the development of OHS. Moreover, leptin signaling in the sympathetic activity.
CB may contribute to maintaining oxygenation in hypoxic
conditions, such as during exposure to high altitude, and in obesity, for a review see Conde et al. (63); leptin’s action
patients with cardiorespiratory disease. Nevertheless, we can- within the CBs might have a role in promoting sympathetic
not forget that leptin significantly augments sympathetic tone overactivation in all these conditions. Recently, Ribeiro et al.
thereby contributing to the development of obesity-induced 2019 (304) tested the contribution of CB to acute leptin
cardiometabolic and respiratory comorbidities (32, 181, 215, effects on sympathetic activity and mean blood pressure in
373) (Figure 13). control and obese rats after CSN denervation. Preliminary
data showed that the CB contributes to the effect of leptin
Future directions: filling the gaps on leptin’s action on sympathetic activity, an effect that is decreased in obese
on peripheral chemoreceptors animals probably due to CB leptin resistance in obesity (304).
Additional research is needed to fully understand leptin’s
In the last decades, a growing body of evidence emerged action in the CB as well as its contribution to chemoreflex
linking leptin to the peripheral control of breathing. It is responses. Obesity, hypertension, metabolic dysregulation,
now generally accepted that leptin acts on the CB to modify and breathing disorders occur throughout the life span.
spontaneous minute ventilation and hypoxic ventilatory Essentially nothing is known about the effect of leptin on
responses in acute and chronic conditions such as obesity and peripheral control of breathing during development and
CIH (46, 305, 399). However, there remains a significant gap aging.
in knowledge regarding the specific mechanisms and signal
pathways by which leptin activates or modifies CB action
and CSN activity promoting alterations in ventilation and Leptin Dysregulation Mediating
the sympathetic nervous system both in acute and chronic
diseases. For example, a greater understanding of which Disordered Breathing and Therapeutic
leptin signaling pathways are activated in the CB in acute and Interventions
chronic diseases could potentially lead to new therapeutic
targets for the treatment of obesity-related respiratory dis- Effect of exercise training on leptin sensitivity
eases. Another point of interest is how the CB contributes to Previous studies have consistently shown that changes in
sympathetic overactivation in sympathetic mediated diseases, lifestyle such as hypocaloric diet and exercise training
such as MetS, T2D, hypertension associated with OSA and reduce obesity, high blood pressure, and diabetes (190,
essential hypertension, some of which co-occur with of 368). These nonpharmacological interventions also improve

Volume 10, July 2020 1067


Leptin in the Control of Breathing Comprehensive Physiology

chemorespiratory function in patients with MetS and decrease energy balance, sympathetic activity, blood pressure, and
the severity of OSA (370). A 10% reduction in body weight ventilation. In this context, the effect of physical exercise
is associated with a 30% reduction in apnea-hypopnea index on leptin concentrations has been studied. Several studies
(AHI) (264, 278). However, the mechanisms explaining the suggest that short-term exercises (<60 min) and exercises
effects of diet and exercise training in patients with MetS and that generated energy expenditure lower than 800 kcal do
comorbidities in OSA are unknown. not modify serum concentrations of leptin (40, 194, 383).
Aerobic (180, 283) and resistance (196, 351) training is Instead, long-term and high-intensity exercise lower lep-
recommended for the prevention and treatment of obesity- tin concentrations in trained males running on a treadmill
induced cardiovascular disorders. Exercise training (10 weeks burning 800 and 1500 kcal (97). These exercise conditions
of moderate-intensity resistance in a vertical ladder) prevented decreased plasma leptin concentrations after 48 h which was
the cardiovascular and inflammatory responses (increases in preceded by a decrease in insulin concentrations. These data
TNF-α and IL-1β) produced by HFD in sedentary rats (351). suggest that the decline in leptin concentration was likely
Interestingly, the anti-inflammatory markers (IL-10; ATII from disruption in metabolic homeostasis created by high-
and Mas receptors and others) were increased in the NTS intensity, long duration, energy expenditure and subsequent
after the resistance training compared to sedentary HFD rats excess post-oxygen consumption (after-burn). Data from
(351). These data demonstrated that moderate intensity of other studies show that leptin concentrations were signifi-
resistance exercise prevented obesity-induced cardiovascular cantly lower immediately after exercise (70% of Vo2 max,
disorders and reduced inflammatory responses in the NTS, energy expenditure > 800 kcal), 10 and approximately 20 to
an important region of the brain that mediate cardiovascular 48 h post exercise (257, 263).
and respiratory function, as explained in the section “Leptin The low Vo2 max is a good predictor of mortality in obese
and Central Control of Breathing”, page 4. subjects (127). High-intensity interval training (HIIT) is an
In addition, the combination of hypocaloric diet interesting alternative to improve the Vo2 max and to reduce
(500 kcal/day for 4 months) and exercise training (three body fat. HIIT can favorably impact the neuroendocrine
60-min sessions three times/week for 4 months) improved axis (i.e., leptin levels) and improves lipid metabolism in
sympathetic peripheral chemoreflex sensitivity and venti- overweight/obese and lean subjects (38, 47, 162). Six weeks
latory central chemoreflex hypersensitivity in patients with of isocaloric training, HIIT or moderate-intensity continu-
MetS and OSA (213). Body weight, waist circumference, ous training (MICT) in adult men with obesity, promoted
glucose levels, systolic/diastolic blood pressure, and AHI similar adaptation to Vo2 max and blood pressure values
were reduced after the hypocaloric diet and exercise ses- (124). Although both exercise protocols were able to increase
sions. Indeed, in the same study, muscle sympathetic nerve Vo2 max, neither MICT nor HIIT changed lipid profile,
activity (MSNA) at rest and in response to hypoxia and body composition or leptin concentrations. In this study,
hypercapnia after exercise was also reduced, suggesting that neither HIIT nor MICT was associated with a change in fat
the less MSNA in patients with MetS and OSA depends mass possibly accounting for no change in circulating leptin
on the reduction of OSA severity and the normalization of levels. However, in two additional studies from the same
chemoreflex (213). group, they found reductions in leptin concentrations after
Improvement of insulin sensitivity and decreased waist 5 (47) and 8 weeks of aerobic training without a reduction
circumference after exercise is associated with amelioration in fat mass in participants (162). In both studies, partici-
of OSA in some subjects. Sleep apnea in obese subjects pants were young, normal weight, physically active males,
causes CIH and sleep fragmentation which leads to and and the reduction in leptin levels seems to be related, as
exacerbate obesity and T2D by increasing sympathetic activ- outlined above, to high energy expenditure during exercise
ity, oxidative stress, inflammation, and lipolysis. Moreover, sessions.
obesity or diabetes can influence chemosensitivity and sleep The effects of HIIT and MICT exercise programs on
apnea either directly or via action on sympathetic activity, cardiorespiratory function have been investigated. The results
inflammation, or other mechanisms (151). Indeed, long-term obtained in obese people (adult men) show that anaerobic
improvements in insulin action due to continuous positive contribution was more pronounced after HIIT. Both exercises
airway pressure (CPAP) would support the hypothesis that improved the Vo2 max and decreased systolic blood pressure
OSA leads to or exacerbates insulin resistance and under- without changes in the visceral or subcutaneous fat mass.
mines the hypothesis that insulin resistance itself leads Moreover, 6 weeks of HIIT or MICT exercise improve
to OSA. In nonobese, HFD fed rats, metformin treatment cardiorespiratory function and blood pressure in obese men
increased insulin sensitivity and prevented the development (124). The after-mentioned findings are in accordance with
of sleep apnea independent of body weight (298). previous studies that showed that short-term aerobic training
Changes in circulating leptin levels is another potential was able to improve Vo2 max (31) and body fat even with-
mechanism by which exercise optimizes the cardiorespi- out reduction in weight (384). Thus, exercise, specifically
ratory function in OSA and MetS patients. As outlined HIIT, is an important therapeutic intervention to mini-
above, leptin is a hormone produced by fat tissue that acts mize the pathogenesis of obesity-induced cardiorespiratory
in the CNS mediating many biological functions including disorders.

1068 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

Effect of bariatric surgery on leptin sensitivity and levels and, in some cases, glucose intolerance. Interestingly,
breathing the respiratory stimulant effect of leptin compensates for
increased mechanical loads on the diaphragm (363) and
OHS is a serious disorder characterized by daytime improving chemosensitivity (26, 29) in ob/ob mice. How-
hypercapnia, disordered breathing, and a reduction in chemo- ever, it is possible that a breakdown in leptin signaling,
sensitivity. Bariatric surgeries, including vertical sleeve potentially through obesity-induced leptin resistance directly
gastrectomy (VSG) and the laparoscopic adjustable gastric contributes to the pathogenesis of OHS independent of the
band (LAGB), (Figure 14) (126) contribute to OSA improve- physical, mechanical load of obesity (246). Alternatively, lep-
ment due to a decrease of body weight and uptake of glucose tin may indirectly improve chemosensitivity and disordered
metabolism (324). However, it was suggested that the VSG breathing through its effect on glucose metabolism (243).
surgery improved glucose and leptin levels through weight Preclinical studies have identified a link between glycemic
independent mechanism (13). For this reason, VSG was control and insulin action on chemosensitivity and disordered
considered more effective than LAGB technique (48, 159). breathing (107).
VSG, a bariatric surgical procedure resulting in weight In animal models, simulation of OSA using CIH has greatly
loss and weight independent improvements in glucose advanced the knowledge of how OSA may impact disease
metabolism, is associated with substantial improvement in states via cyclic drops in blood oxygen. In both rodents and
sleep-disordered breathing (SDB). However, it is hard to humans, previous studies demonstrated that CIH increased
dissociate the effects of obesity per se from metabolic and circulating levels of glucose and leads to gluconeogenesis
hormonal influences in obese people on the chemorespiratory (255, 286, 327, 382). Thus, in OSA patients, it is possible
function in OHS patients (14). that CIH alters the glucose metabolism (226).
Results from human studies performed in obese subjects Once T2D patients have autonomic dysfunction and asso-
indicate that fat mass imposes a physical, mechanical load on ciated cardiorespiratory disorders, autonomic dysfunction
respiratory muscles which in turn can lead to hypoventilation is a potential mechanism for glucose intolerance and OSA
(268). Indeed, obesity results in significantly elevated leptin (34). CIH increases tonic and reactive afferent chemoreceptor
activity from the CB which in turn increases plasma cate-
cholamine levels (166, 334), fasting hyperglycemia (340),
(A) Laparoscopic adjustable (B) Roux-en-Y gastric
gastric banding (LAGB) bypass (RYGB)
and hypertension (166). Peripheral chemoreceptors are more
sensitive to O2 and glucose then CO2 . However, any dis-
ruption of glucose can affect the sensitivity to O2 and CO2
Inflatable Distal jejunum
band impairing respiratory function (118). Addition of 2-deoxy-
d-glucose (2DG; a glucoprivic agent) in the drinking water
of rats can prevent phrenic long-term facilitation, a form of
Biliopancreatic respiratory motor plasticity, suggesting that alterations in
secretions
glucose sensing can directly alter breathing (209).
The reciprocal interaction between O2 and glucose sensing
and control of breathing is suggested from data obtained from
Proximal jejunum
human studies in patients with two types of bariatric surgery
(VSG and LAGB), with the intent to improve T2D and obe-
sity (Figure 14) (126). Twice as many patients had resolution
of OSA 1 year after VSG when compared to patients who had
(C) Sleeve gastrectomy (SG) (D) Biliopancreatic diversion LAGB (48, 159), suggesting that more than reducing body
with duodenal switch (BPD-DS) weight, the improvement in metabolic function ameliorate
the symptoms of OSA. Moreover, in OHS patients who had
bariatric surgery, arterial blood gases normalized acutely and
Duodenum
remained normal overtime (356, 357).
The role of leptin in modulating chemosensitivity and
disordered breathing following bariatric surgery is largely
unknown. The hypothesis that VSG alters chemosensitivity
Ileum and ventilatory drive through a leptin-dependent mech-
anism (103) has been tested in animals. VSG improves
Jejunum chemosensitivity and lowers leptin levels independent of
changes in body and fat mass in WT mice with diet-induced
obesity (103). On the other hand, leptin-deficient ob/ob
Figure 14 Graphic depiction of types of bariatric surgery. mice undergoing VSG failed to show an improvement in
Printed, with permission, from Gletsu-Miller N and Wright BN, 2013 hypercapnia ventilatory response (HCVR); however, HCVR
(126). did improve with leptin replacement in these animals.

Volume 10, July 2020 1069


Leptin in the Control of Breathing Comprehensive Physiology

Thus, VSG in ob/ob mice was associated with a reduction reduction in diurnal and nocturnal systolic and diastolic blood
in body mass index (BMI), fat mass, and improvements in pressure (238). Another recent meta-analysis systematically
glucose metabolism. Of note, leptin deficiency prevented analyzed six studies addressing the effect of CPAP on diur-
VSG-induced improvements in chemosensitivity, suggesting nal blood pressure in patients with OSA and drug-resistant
that VSG improves chemosensitivity and ventilatory drive hypertension, and found a favorable reduction after treatment
through a leptin-dependent mechanism (103). with CPAP (160). The meta-analysis by McEvoy et al. (225)
Besides the effects of bariatric surgery in improving lep- inclusive of 2717 eligible adults between 45 and 75 years
tin’s effect on food intake and body weight (353), it is also of age, who had moderate to severe OSA and coronary
possible that the VSG surgery improved the action of leptin or cerebrovascular disease, concluded that subjects who
on chemoreception. Modulating central leptin signaling received CPAP had reduced snoring and daytime sleepiness
specifically in the periaqueductal gray leads to disordered and improved health-related quality of life and mood when
breathing and reduces the HCVR (103). Thus, VSG not compared to subjects who received usual care without
only affects central control of feeding behaviors and glucose CPAP. However, both groups had similar rates of death
regulation, but it also affects ventilation and chemosensitivity. from cardiovascular causes, myocardial infarction, stroke, or
Substantial evidence from preclinical and clinical studies hospitalization for HF, unstable angina, or transient ischemic
suggests that bariatric surgeries can improve glucose reg- attack. Importantly, the effects of CPAP therapy on cardiovas-
ulation through weight-independent pathways; it is likely cular morbidity occurred only in patients who were adherent
that other pathophysiological states associated with obesity to treatment longer than 4 h per night (225).
may also improve. Taken together, these results reinforce the The AHI is associated with high plasma leptin levels.
evidence that metabolic surgery, as well as leptin, may be IH increases leptin production, and serum leptin levels are
employed as future treatments to optimize chemosensitive elevated in OSA patients, independent of obesity (165, 169,
function and respiratory disorders. 312). However, in some cases, OSA seems to be unrelated
to leptin plasma levels, as is the case of subjects with OSA
but with controlled body weight (24, 272, 328). Rodent
Effect of CPAP on leptin sensitivity, breathing, and studies (29, 260, 363) have delineated how leptin influences
energy balance breathing stability, findings that have been translated to our
Chronic OSA with recurrent episodes of apneas, CIH and understanding of leptin’s influence on the development of
sleep fragmentation over years alters metabolic balance and OSA in humans. Some studies implement hyperleptinemia
alters the function of many organ systems, including the and leptin dysregulation as contributing to OSA. Alterna-
brain and the cardiovascular system (43, 139). These adverse tively, OSA can cause leptin serum levels to rise as a result of
effects lead to a variety of clinical sequelae including daytime intermittent hypoxemia, sleep fragmentation, or heightened
sleepiness from nocturnal sleep fragmentation in 80% of the sympathetic activity. A drop in serum leptin levels observed
patients with OSA. As the disorder progresses, the sleepi- in patients with OSA after nocturnal CPAP supports this idea
ness becomes increasingly dangerous, causing impaired (24, 146, 165, 339).
performance at work and major work-related and road The relationship between CPAP therapy, changes in BMI
accidents (139, 170). Many patients can develop cognitive and leptin levels is not consistent across all studies. CPAP
and neurobehavioral dysfunction, inability to concentrate, therapy for 8 weeks decreases leptin levels without affecting
memory impairment and depression. If untreated, OSA is BMI in cohort of patients with OSA (146). CPAP use for
a major determinant of cardiovascular morbidity and mor- ≥4 h per night, reduced total cholesterol, low-density lipopro-
tality (43, 398). Insulin resistance, T2D, and altered serum tein, and leptin levels. Indeed, circulating leptin levels were
lipid profile occur frequently in patients with OSA further significantly correlated with insulin resistance. In patients
increasing the risk of cardiovascular morbidity (170, 337). Of with moderate-to-severe OSA, compliant CPAP usage may
importance, metabolic impairment can occur in patients with improve insulin secretion (69). Results of a meta-analysis
OSA independent of body weight. showed that CPAP could significantly reduce leptin levels in
CPAP, available since the beginning of the 1980s, is rec- obesity sleep apnea and hypoventilation syndrome (OSAHS)
ognized as most effective and is commonly used to treat patients without concomitant weight loss (55).
patients with OSA (358). Alternative options include weight On the other hand, Drummond et al. (88) reported that in
control (diet and exercise), mandibular advancement devices, response to CPAP therapy, mean leptin serum levels decreased
and a number of upper airway surgical approaches. However, to 11.0 μg/L, and 6 months later it was 11.4 μg/L, suggest-
nasal CPAP is highly effective in controlling symptoms, ing that CPAP has a small effect on leptin levels. Yosunkaya
improving quality of life and reducing the clinical sequelae et al. (397) found no correlation between serum leptin levels
of sleep apnea. A short period of CPAP treatment amelio- and AHI, oxygen saturation, or excessive daytime sleepiness.
rated nocturnal obstructive events, SaO2 desaturations, and CPAP treatment did not significantly change the BMI, waist
daytime sleepiness (273, 352). A large meta-analysis, which and neck circumference, or leptin levels in OSAS patients.
included 32 studies with a total of 1948 patients, showed that Thus, the authors concluded that CPAP treatment does not
CPAP treatment is associated with a modest, but significant significantly affect serum leptin levels (397).

1070 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

While weight loss improves OSA, it remains unclear why affects hypothalamic neuropeptides involved in body weight
CPAP is often associated with weight gain (338). CPAP homeostasis (332). It also induced significant weight loss and
likely affects eating behavior and/or metabolism involved reduced adipose tissue mass in both lean and diet-induced
in regulating energy balance. As outlined above, CPAP can obese animals. On the other hand, the energy intake was
change the hormonal profile by reducing abnormally high less affected in lean animals during the 32-week diet (331).
circulating levels of leptin and ghrelin in patients with OSA Weight reduction without changes in energy intake may be
which reduces excessive food intake. CPAP may alter energy explained by the thermogenetic effects of leptin (378), an
metabolism by reducing the metabolic rate in wakefulness open question for future experiments.
and sleep, although studies supporting this observation are Fliedner et al. (105) administered intranasal leptin
limited and inconsistent. Moreover, CPAP use in patients (0.2 mg/kg) to Wister rats. This dose would achieve a tissue
with OSA is not consistently associated with altered physical concentration of 50 ng/g within the hypothalamus; 120 times
activity (336). Thus, combining CPAP with other weight loss the physiological level (105). Perhaps, this concentration is
approaches should be used to encourage optimal outcomes necessary to activate hypothalamic neurons in obese subjects
in OSA patients (338). The results from a recent study sup- with leptin resistance. However, for intranasal leptin to be
port this recommendation. When CPAP was combined with considered a therapeutic option for the treatment of obesity,
exercise training in patients with OSA, AHI did not change more preclinical studies are required to better understand the
significantly in the control group, decreased moderately in the long-term effects that may be undesirable.
exercise group, and decreased significantly in the CPAP and The ARC is a major target for leptin in the hypothalamus
exercise CPAP groups. Both exercise and CPAP improved (246) and in this nucleus, leptin receptors are present on two
subjective excessive daytime sleepiness and quality of life different populations of neurons, one of which synthesizes
in HF patients. However, compared with the control group, NPY and AgRP and a second population of neurons that
changes in the quality of life were significant only in the express POMC and CART immunoreactivity (333). The
exercise groups. Furthermore, peak oxygen consumption,
orexigenic NPY/AgRP neurons are inhibited, whereas the
muscle strength, and endurance improved only in the exercise
anorexigenic POMC/CART neurons are activated (3, 246).
groups compared with the CPAP group. Interestingly, there
Conversely, POMC gene expression in the ARC is decreased
was a trend in improved sexual function when exercise was
in leptin-deficient ob/ob mice and in db/db mice with an LR
combined with CPAP (336). These data suggest that exercise
mutation (232). CART expression in the hypothalamus is
could be a therapeutic option for patients with HF and OSA
dependent on the energy status and differs in animals depend-
who refuse or who are intolerant to CPAP.
ing on their susceptibility for diet-induced obesity (157),
Chirinos et al. (56), randomized patients with moderate to
and centrally or peripherally applied leptin directly acts on
severe OSA (without HF) to undergo a weight loss program,
CPAP, and a combination of the two interventions. They CART neurons in distinct nuclei of the rat hypothalamus
report that that subjects in the weight loss program only and (9, 94). Intranasal leptin treatment increases POMC mRNA,
those assigned to the combined interventions had less insulin and POMC reduces appetite and body weight. However,
resistance and lower levels of C-reactive protein and serum intranasal application of leptin downregulates NPY mRNA
triglyceride levels. None of these changes were observed in in the hypothalamus (233, 234, 331), which could lead to
the group receiving CPAP alone. These results emphasize the a reduction in NPY levels, and therefore, less activation of
importance of adjunctive therapy to CPAP for the treatment orexinergic pathways.
of OSA with additional weight loss and exercise programs Since patients with OHS hypoventilate and have high cir-
when applicable. culating leptin levels, they are leptin resistant (246). Berger
et al. (33) demonstrated in an elegant translational study
in leptin-resistant mice with diet-induced obesity, that a
Effect of intranasal leptin treatment on breathing and single dose of intranasal leptin ameliorates the upper airway
energy balance in preclinical model of OSA obstruction and hypoventilation during sleep, independent of
Activating leptin receptors in the CNS via intranasal instilla- metabolism. Intranasal leptin, but not leptin given intraperi-
tion of leptin is potentially a novel therapeutic intervention for toneally, induced leptin receptor signaling in hypothalamic
appetite control. As cited before, obese people and animals and medullary centers involved with respiratory control.
are hyperleptinemic and are leptin resistance when compared Thus, the study suggests that intranasal leptin can be tested
to lean controls (246). Leptin bypasses the blood-brain barrier as a potential therapy in patients with SDB. More research
via nonsaturable direct transport mechanism from nose to is needed to determine the long-term efficacy of intranasal
brain, which is not impeded by elevated peripheral levels leptin in alleviating SDB; whether the dose needs to be
of leptin (105). Leptin resistance likely occurs as a result adjusted based on the therapeutic goal; what mechanisms are
of alterations in leptin receptors signaling in hypothalamic mediating the leptin intranasal effect on respiratory network
neurons, especially in the ARC, and/or decreased leptin need to be more completely described.
transport across the blood-brain barrier (92). In normal adult This novel finding of intranasal leptin treatment alleviat-
rats, intranasal leptin reduces food intake and weight gain and ing hypoventilation and upper-airway obstruction in an obese

Volume 10, July 2020 1071


Leptin in the Control of Breathing Comprehensive Physiology

mouse model is exciting and provides a much-needed novel adiponectin plays a major role in the pathogenesis of MetS
therapeutic approach for the management of SDB. (73, 379, 385).
Circulating adiponectin levels are much lower in indi-
viduals with obesity (15) and are inversely correlated with
Leptin and adiponectin: the Yen and Yang insulin resistance and other metabolic signatures of MeS.
Without affecting leptin levels, adiponectin-deficient mice
We have reviewed what is currently known about the effect are insulin resistance and more susceptible to developing
of leptin on mechanisms that control breathing, and the metabolic and vascular disease (i.e., systemic and pulmonary
association between obesity, leptin resistance, and disordered hypertension) than WT mice (359). Moreover, adiponectin
breathing. While much of this review is focused on the deficient mice when feed a high-calorie diet become even
specific role of leptin on physiology and pathophysiology more insulin-resistant (141). Overexpression of adiponectin
obesity, individuals with obesity also have low plasma levels in ob/ob mice normalized glucose levels even though the
of adiponectin. The adiponectin protein was first identified in animals had a higher volume of fat mass compared to their
1995 by Scherer et al. (329) and named Acrp30. It was also ob/ob littermate controls (176). Therapeutics that improve
discovered by three other investigative teams, independently, insulin sensitivity and glucose regulation, specifically the
and named AdipoQ, apM1, and GBP28 (156, 211, 247). thiazolidinedione (TZD) class of drugs (i.e., rosiglitazone and
Adiponectin, similar to leptin, is produced by adipocytes but pioglitazone) which are PPAR𝛾 agonists, are potent inducers
in much higher concentrations. It is the most abundant plasma of adiponectin gene expression (206, 281, 369). Adiponectin
protein with concentrations between 5 and 10 micrograms/mL KO mice are resistant to the metabolic and cardioprotective
of plasma or 0.01% of total protein. While leptin levels are effects of the TZD class of drugs (201).
directly proportional to fat mass throughout the lifespan, The proposed mechanism for lower levels of adiponectin in
adiponectin levels are only proportional to fat mass prior to obese subjects is through the action of TNFα, which is in high
5 years of age after which time it is inversely related to fat concentrations in adipose tissue (52, 236). Adipocytes pro-
mass (195). Similar to leptin, females have higher circulating duce and secrete TNFα but activated macrophages, within the
levels of adiponectin than men, even after controlling for stromovascular fraction of white fat, are the major source of
fat mass (59, 221); testosterone decreases adiponectin levels this pro-inflammatory cytokine. TNFα inhibits the transcrip-
(109). Lastly, in contrast to leptin, which is pro-inflammatory tion of PPARy and C/EBPα genes (reviewed in Ref. 236),
and pro-oxidant, adiponectin is anti-inflammatory and two major transcription factors for adiponectin expression.
antioxidant (for review see Ref. 100). Thus, TNFα modulates adiponectin expression by both
Adiponectin is a complex protein, comprised of 244 amino paracrine and autocrine mechanisms, indirectly suppressing
acids, encoded by the ADIPOQ gene on the long arm of adiponectin gene expression because it inhibits the transcrip-
chromosome 3 (3q27) (211). It is transcriptionally upreg- tion of PPARy, the “adipogenic master regulator” (73).
ulated by peroxisome proliferator-activated receptor-𝛾
(PPARy), CCAAT/enhancer-binding protein-α (C/EBPs),
sterol-responsive-element-binding protein-1c, forkhead The role of adiponectin and sleep-disordered
box 1, and specificity protein 1 (87, 98, 292, 293, 395), breathing
and downregulated by reactive oxygen species, TNFα and Not much has been published about how adiponectin affects
IL-6 (reviewed in Ref. 205). PPAR𝛾 directly increases central or peripheral systems that control breathing dur-
adiponectin gene transcription through peroxisome prolifer- ing health or disease. A recently published meta-analysis,
ator response elements (PPRE) (205). Adiponectin binds to including 1356 subjects from 28 studies, concluded that
four different receptors: AdipoR1 and AdipoR2, which are plasma/serum adiponectin levels were lower in adults with
widely distributed throughout the body; T-cadherin, which is OSAHS than controls (standard mean difference = −0.71,
expressed in the vasculature but does not have a cytoplasmic 95% CI = −0.92 to −0.49, P < 0.001). Studies included in
domain (112, 222), and calreticulin (calregulin), which is the meta-analysis had previously reported no statistically sig-
expressed on macrophages and when activated removes apop- nificant difference between OSAHS patients and controls in
totic debris (360). Of the adiponectin receptors, AdipoR1 and terms of age, gender and BMI (208). Al Mutairi et al. reported
AdipoR2 are xthe best characterized (389). AdipoR1 is most an inverse relationship between OSAHS severity in adults
abundantly expressed in skeletal muscle, enhancing fatty acid and adiponectin levels after controlling for sex, and BMI (4).
oxidation through phosphorylation of AMPK. AdipoR2 is A similar finding in adolescents has also been reported
most abundantly expressed in the liver and its activation low- (116). Yoshikawa et al., in a small cohort of patients with
ers glucose levels by decreasing hepatic glucose production OSA (n = 22), reported that plasma adiponectin levels were
and reduces oxidative stress (388, 389). Thus, ligand binding inversely correlated with the AHI (r = −0.582, P < 0.005) and
to these receptors on cells throughout the body, regulates % time in oxygen saturation SpO2 less than 90% (r = −0.539,
systemic and pulmonary vascular homeostasis, fatty acid P < 0.01), and after 3 months of CPAP, plasma adiponectin
oxidation, insulin sensitivity, and reduces oxidative stress levels significantly increased in 50% of the patients (396).
and suppresses inflammation (Figure 15) (359). Accordingly, CIH occurs during OSAHS, and CIH decreases adiponectin

1072 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

Oxidative stress
Angiotensin II PPARγ agonist
Adipose
Testosterone
tissue BMI
IL-6
TNF
BMI

Macrophage Adiponectin Liver


Metabolism
NF-κB
TNF
TNF-α
SR-A
Foam cells
AMPK SRF
cAMP-PKA COX-2
AMPK
NF-κB PGE2 mTOR Actin
eNOS

VCAM-1 Proliferation/growth
IL-8 TNF
Endothelial cell Muscle cell

Figure 15 Schematic showing adiponectin signaling in macrophages,


liver, endothelial and muscle cells. Adiponectin is produced by adipose tis-
sue, the agonist to the transcription factor PPAR𝛾 will increase the production of
adiponectin and decrease BMI, while oxidative stress, angiotensin II, testosterone, IL-6,
TNFα, will inhibit the production of adiponectin. Adiponectin binding to adiponectin
receptors on (i) macrophages inhibits the production of NF-𝜅B and SR-A thereby
inhibiting the production of TNFα, and foam cells, (ii) liver cells increases metabolism
and blocks TNFα production, (iii) endothelial cells promoting NO production, and
inhibiting synthesis of chemokines, (iv) muscle cells inhibits the production of TNF and
promotes growth and proliferation. m-TOR, mammalian target of rapamycin; SR-A,
scavenger receptors-A; AMPK, AMP-activated protein kinase; SRF, serum response ele-
ment; COX-2, cyclooxygenase-2; VCAM-1, vascular cell adhesion protein 1; PPAR𝛾,
peroxisome proliferator-activated receptor gamma; BMI, body mass index; IL, Inter-
leukin. Adapted, with permission, from Summer R, et al., 2011 (359).

levels and adiponectin receptor expression (240, 282). The which was associated with a decrease in blood pressure
mechanism by which this downregulation occurs is not (277). Adiponectin receptors have been localized to neurons
known. In the aforementioned study by Yoshikawa et al. in the medial NTS where it is involved in regulation of
(396), they also measured TNFα levels in the cohort of blood pressure (155) but not in the commissural nucleus, an
patients with OSA and reported that TNFα levels were pos- important nucleus that integrates input from the peripheral
itively correlated with the AHI (r = 0.462, P < 0.05). Thus, arterial chemoreceptors. Two recent human studies suggest
one could speculate that elevated TNFα levels in response that adiponectin may be important in positively mediating
to CIH, may decrease adiponectin levels (52), as outlined sympathetic and parasympathetic tone in adolescent girls
above. Exogenous adiponectin given to animals decreased (372) and adults who develop T2D (145).
the consequences of CIH on several organ systems (82, 83). Relatively low levels of adiponectin, in the microenvi-
Of note, thirteen single nucleotide polymorphisms of the ronment of cells and tissues, could be contributing to a
ADIPOQ gene in Chinese Han subjects have been reported persistent pro-inflammatory, pro-oxidant state, which affects
to be associated with the prevalence and severity of OSA and the activity of cells and neurons that control breathing, such
AHI (390). How polymorphisms of the ADIPOQ gene equate as the CB. The CB is surrounded by adipocytes, as shown
to plasma/serum adiponectin levels has not to be reported but in the photomicrograph of an organotypic culture of the CB
would be of considerable interest. from a 2-week old rat (Figure 16) (187). The amount of fat
While much is known about how leptin modulates central that surrounds the CB increases with the size of the animal.
and peripheral cardiorespiratory responses as outlined in Might local release of leptin and adiponectin from this local
this article, less is known about how adiponectin might fat depot surrounding the CB modulate its function? How
modify these responses. Adiponectin can indirectly mod- does this local fat depot differ in humans and models of
ulate blood pressure in the RVLM in animals. Agonist OSA, with respect to local release of TNFα from activated
binding to abnormal cannabidiol G-protein coupled receptor macrophages? Additional studies are needed to understand
18 in the brainstem, specifically in the RVLM, elevated the direct and indirect influences of adiponectin on central and
neuronal adiponectin levels and reduced oxidative stress, peripheral systems that control breathing, and the corollary:

Volume 10, July 2020 1073


Leptin in the Control of Breathing Comprehensive Physiology

FC Leptin also acts on the CB to modify spontaneous minute


ventilation and hypoxic ventilatory responses in acute and
CA
chronic conditions such as obesity and CIH. The contribution
CB of the CB to ventilatory responses is blunted in obesity, prob-
ably due to leptin resistance within the organ. Information
on leptin signaling pathways in the CB activated in these
acute and chronic conditions would have significant clinical
CA relevance as leptin or its signaling within the CB might
be therapeutic targets for the treatment of obesity-related
100 μm
respiratory diseases. Preliminary data also shows that the
CB contributes to leptin action on the sympathetic nervous
Figure 16 Low-power photomicrograph, using Hoffman system. Knowing that CB contributes to the heightened
contrast microscopy, of an organotypic slice of the carotid
body 24 h in culture. Fat cells are in close proximity to the CB in sympathetic activity present in cardiometabolic diseases, fur-
vivo. Tissue was removed from a Sprague Dawley rat at 2 weeks post- ther research on leptin action on CB-mediated sympathetic
natal age. CB, carotid body; CA, carotid artery. Arrows depict fat cells activity might unravel new specific therapeutic targets for
(FC). Adapted, with permission, from Kwak DJ, et al., 2006 (187).
cardiometabolic diseases.
Breathing disorders also occur in early development, but
how disordered breathing and CIH affect adiponectin levels. essentially nothing is known about how leptin may be mod-
Since adiponectin confers significant and substantial car- ulating breathing during development. While preliminary
dioprotection and protection from atherosclerotic disease, data are presented in this article, showing that leptin is not
studies are needed to determine whether therapeutic modula- likely to increase CO2 sensitivity prior to 3 weeks postnatal
tion of adiponectin levels in humans with OSA may modify age in newborn rats or decrease apnea frequency, more data
cardiovascular morbidities. are needed to substantiate these findings. Most mammalian
species are “leptin resistant” with respect to leptin’s effect
on feeding behavior and energy expenditure during early
development. Instead, leptin’s role during early develop-
Conclusion ment appears to be critical for normal organ development.
Leptin stimulates breathing and leptin deficiency, for SGA infants have lower and large gestational age (LGA)
example, obesity leptin resistance, promotes an impair- infants have higher leptin levels than infants who are AGA
ment of the central and peripheral mechanisms that control at birth, but yet both SGA and LGA are at increased risk of
breathing, which can be reverted by leptin therapy. Evidence developing leptin resistance in adolescence and adulthood
supports that leptin acts mainly in the NTS in the hindbrain with associated cardiorespiratory comorbidities. Thus, both
and dorsal medial nucleus in the hypothalamus. Leptin may excess and absence of leptin during early development pro-
also act in the PeF, DM, and ARC of the hypothalamus grams the system for adverse cardiorespiratory outcomes.
where leptin receptors are also expressed. Administration of The mechanisms of this effect are poorly understood; human
leptin into the lateral ventricle might act in different nuclei studies to better understand how modifying this programming
of the hypothalamus, including in PeF, DM and arcuate could lead to new therapeutic targets to improve long-term
(ARC) nuclei that express leptin receptors. Although the outcomes throughout life are needed. Leptin levels in the
mechanisms and pathways activated by leptin to facilitate fetus exponentially increase after 34 weeks gestation; pre-
breathing are still under investigation, evidence suggests mature infants born at the limit of viability (22–24 weeks of
that the facilitator effects of leptin on breathing require the gestation) have undetectable leptin levels. Carefully designed
brain melanocortin system, including the POMC-MC3/4R studies to better understand the consequences of undetectable
pathway, the same mechanism activated by leptin that leptin levels for many months during organ development in
modulates food intake and sympathetic activity. A better these infants and whether leptin replacement may be a safe
understanding of how leptin controls ventilation, upper and efficacious in improving short and long-term outcomes
airway function and sympathetic activity would inform in this population of high-risk infants are needed.
new approaches for therapeutic interventions to treat obe- Adiponectin is another important adipokine that is mainly
sity related co-morbidites. For example, preclinical studies produced by adipose tissue. Unlike leptin, adiponectin levels
in leptin-resistant mice, showing the benefit of intranasal are low in adults with obesity and obesity-related metabolic
leptin on metabolism, weight loss, and improved CO2 and cardiorespiratory morbidities. Adiponectin promotes
chemosensitivity, and less respiratory disturbances begs the vasoprotection, glucose tolerance, insulin sensitivity, and has
question as to whether intranasal leptin may be feasible antioxidant and anti-inflammatory properties, and balances
and efficacious for the treatment of SDB in humans. How sympathetic and parasympathetic tone (145). Preclinical stud-
intranasal leptin will affect sympathetic overexcitation in ies to determine the specific role of adiponectin on central
individuals with hyperleptinemia would be important to or peripheral regulation of sympathetic tone in normal and
determine. disease models are not known. These studies could contribute

1074 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

to a more balanced understanding of the role of adipokines 18. Athanasakis E, Karavasiliadou S, Styliadis I. The factors contributing
to the risk of sudden infant death syndrome. Hippokratia 15: 127-131,
(leptin and adiponectin) on these physiological responses 2011.
since the biological effects of these adipokines are closely 19. Attig L, Brisard D, Larcher T, Mickiewicz M, Guilloteau P, Boukthir
S, Niamba CN, Gertler A, Djiane J, Monniaux D, Abdennebi-Najar L.
linked. Postnatal leptin promotes organ maturation and development in IUGR
piglets. PLoS One 8: e64616, 2013.
20. Attig L, Djiane J, Gertler A, Rampin O, Larcher T, Boukthir S, Anton
PM, Madec JY, Gourdou I, Abdennebi-Najar L. Study of hypothala-
Related Articles mic leptin receptor expression in low-birth-weight piglets and effects
of leptin supplementation on neonatal growth and development. Am J
Phys Endocrinol Metab 295: E1117-E1125, 2008.
Adiponectin Regulation and Function 21. Attig L, Larcher T, Gertler A, Abdennebi-Najar L, Djiane J. Postnatal
leptin is necessary for maturation of numerous organs in newborn rats.
Obstructive Sleep Apnea and Vascular Diseases Organogenesis 7: 88-94, 2011.
22. Babu AR, Herdegen J, Fogelfeld L, Shott S, Mazzone T. Type 2 dia-
betes, glycemic control, and continuous positive airway pressure in
obstructive sleep apnea. Arch Intern Med 165: 447-452, 2005.
Acknowledgement 23. Baquero AF, de Solis AJ, Lindsley SR, Kirigiti MA, Smith MS, Cow-
ley MA, Zeltser LM, Grove KL. Developmental switch of leptin sig-
naling in arcuate nucleus neurons. J Neurosci Off J Soc Neurosci 34:
The authors would like to acknowledge Ms. Sonia Dos Santos 9982-9994, 2014.
for her exceptional administrative support in preparing this 24. Barcelo A, Barbe F, Llompart E, de la Pena M, Duran-Cantolla J,
Ladaria A, Bosch M, Guerra L, Agusti AG. Neuropeptide Y and lep-
manuscript. tin in patients with obstructive sleep apnea syndrome: Role of obesity.
Am J Respir Crit Care Med 171: 183-187, 2005.
25. Barrington KJ, Finer NN. Periodic breathing and apnea in preterm
infants. Pediatr Res 27: 118-121, 1990.
References 26. Bassi M, Furuya WI, Menani JV, Colombari DS, do Carmo JM, da
Silva AA, Hall JE, Moreira TS, Wenker IC, Mulkey DK, Colombari
1. Abdala AP, Rybak IA, Smith JC, Paton JF. Abdominal expiratory E. Leptin into the ventrolateral medulla facilitates chemorespiratory
activity in the rat brainstem-spinal cord in situ: Patterns, origins response in leptin-deficient (ob/ob) mice. Acta Physiol 211: 240-248,
and implications for respiratory rhythm generation. J Physiol 587: 2014.
3539-3559, 2009. 27. Bassi M, Furuya WI, Zoccal DB, Menani JV, Colombari DS, Mulkey
2. Abraham KA, Feingold H, Fuller DD, Jenkins M, Mateika JH, DK, Colombari E. Facilitation of breathing by leptin effects in the
Fregosi RF. Respiratory-related activation of human abdominal central nervous system. J Physiol 594: 1617-1625, 2016.
muscles during exercise. J Physiol 541: 653-663, 2002. 28. Bassi M, Furuya WI, Zoccal DB, Menani JV, Colombari E, Hall JE,
3. Ahima RS, Saper CB, Flier JS, Elmquist JK. Leptin regulation of da Silva AA, do Carmo JM, Colombari DS. Control of respiratory and
neuroendocrine systems. Front Neuroendocrinol 21: 263-307, 2000. cardiovascular functions by leptin. Life Sci 125: 25-31, 2015.
4. Al Mutairi S, Mojiminiyi OA, Al Alawi A, Al Rammah T, Abdella 29. Bassi M, Giusti H, Leite CM, Anselmo-Franci JA, do Carmo JM, da
N. Study of leptin and adiponectin as disease markers in subjects with Silva AA, Hall JE, Colombari E, Glass ML. Central leptin replacement
obstructive sleep apnea. Dis Markers 2014: 706314, 2014. enhances chemorespiratory responses in leptin-deficient mice inde-
5. Allen AM. Angiotensin AT1 receptor-mediated excitation of rat pendent of changes in body weight. Pflugers Arch Eur J Physiol 464:
carotid body chemoreceptor afferent activity. J Physiol 510 (Pt 3): 145-153, 2012.
773-781, 1998. 30. Bassi M, Nakamura NB, Furuya WI, Colombari DS, Menani JV, do
6. Alvarez-Buylla R, Alvarez-Buylla E, Mendoza H, Montero SA, Carmo JM, da Silva AA, Hall JE, Colombari E. Activation of the
Alvarez-Buylla A. Pituitary and adrenals are required for hyper- brain melanocortin system is required for leptin-induced modulation
glycemic reflex initiated by stimulation of CBR with cyanide. Am J of chemorespiratory function. Acta Physiol 213: 893-901, 2015.
Phys 272: R392-R399, 1997. 31. Batacan RB Jr, Duncan MJ, Dalbo VJ, Tucker PS, Fenning AS. Effects
7. Alvarez-Buylla R, de Alvarez-Buylla ER. Carotid sinus receptors par- of high-intensity interval training on cardiometabolic health: A sys-
ticipate in glucose homeostasis. Respir Physiol 72: 347-359, 1988. tematic review and meta-analysis of intervention studies. Br J Sports
8. Alvarez-Buylla R, de Alvarez-Buylla ER. Changes in blood glucose Med 51: 494-503, 2017.
concentration in the carotid body modify brain glucose retention. 32. Bell BB, Rahmouni K. Leptin as a mediator of obesity-induced hyper-
Adv Exp Med Biol 360: 293-296, 1994. tension. Curr Obes Rep 5: 397-404, 2016.
9. Ambati S, Duan J, Duff E, Choi YH, Hartzell DL, Della-Fera MA, 33. Berger S, Pho H, Fleury-Curado T, Bevans-Fonti S, Younas H, Shin
Baile CA. Gene expression in arcuate nucleus-median eminence of MK, Jun JC, Anokye-Danso F, Ahima RS, Enquist LW, Mendelowitz
rats treated with leptin or ciliary neurotrophic factor. Biofactors 31: D, Schwartz AR, Polotsky VY. Intranasal leptin relieves sleep-
133-144, 2007. disordered breathing in mice with diet-induced obesity. Am J Respir
10. Anderson TM, Garcia AJ 3rd, Baertsch NA, Pollak J, Bloom JC, Wei Crit Care Med 199: 773-783, 2019.
AD, Rai KG, Ramirez JM. A novel excitatory network for the control 34. Bernardi L, Bianchi L. Integrated cardio-respiratory control: Insight in
of breathing. Nature 536: 76-80, 2016. diabetes. Curr Diab Rep 16: 107, 2016.
11. Anderson TM, Ramirez JM. Respiratory rhythm generation: Triple 35. Bianchi AL, Denavit-Saubie M, Champagnat J. Central control of
oscillator hypothesis. F1000Research 6: 139, 2017. breathing in mammals: Neuronal circuitry, membrane properties,
12. Andresen MC, Doyle MW, Bailey TW, Jin YH. Differentiation of and neurotransmitters. Physiol Rev 75: 1-45, 1995.
autonomic reflex control begins with cellular mechanisms at the first 36. Biro FM, Wien M. Childhood obesity and adult morbidities. Am J Clin
synapse within the nucleus tractus solitarius. Braz J Med Biol Res 37: Nutr 91: 1499S-1505S, 2010.
549-558, 2004. 37. Blackmore HL, Niu Y, Fernandez-Twinn DS, Tarry-Adkins JL,
13. Arble DM, Sandoval DA, Seeley RJ. Mechanisms underlying weight Giussani DA, Ozanne SE. Maternal diet-induced obesity programs
loss and metabolic improvements in rodent models of bariatric surgery. cardiovascular dysfunction in adult male mouse offspring independent
Diabetologia 58: 211-220, 2015. of current body weight. Endocrinology 155: 3970-3980, 2014.
14. Arble DM, Schwartz AR, Polotsky VY, Sandoval DA, Seeley RJ. 38. Bluher S, Kapplinger J, Herget S, Reichardt S, Bottcher Y, Grimm A,
Vertical sleeve gastrectomy improves ventilatory drive through a Kratzsch J, Petroff D. Cardiometabolic risk markers, adipocyte fatty
leptin-dependent mechanism. JCI Insight 4 (1): e124469, 2019. acid binding protein (aFABP) and the impact of high-intensity interval
15. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa training (HIIT) in obese adolescents. Metab Clin Exp 68: 77-87, 2017.
J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, 39. Bonsignore MR, Borel AL, Machan E, Grunstein R. Sleep apnoea and
Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, metabolic dysfunction. Eur Respir Rev 22: 353-364, 2013.
Ohmoto Y, Funahashi T, Matsuzawa Y. Paradoxical decrease of an 40. Bouassida A, Zalleg D, Bouassida S, Zaouali M, Feki Y, Zbidi A,
adipose-specific protein, adiponectin, in obesity. Biochem Biophys Tabka Z. Leptin, its implication in physical exercise and training:
Res Commun 257: 79-83, 1999. A short review. J Sports Sci Med 5: 172-181, 2006.
16. Arnold AC, Shaltout HA, Gallagher PE, Diz DI. Leptin impairs car- 41. Bouret SG, Gorski JN, Patterson CM, Chen S, Levin BE, Simerly RB.
diovagal baroreflex function at the level of the solitary tract nucleus. Hypothalamic neural projections are permanently disrupted in diet-
Hypertension 54: 1001-1008, 2009. induced obese rats. Cell Metab 7: 179-185, 2008.
17. Ashworth CJ, Hoggard N, Thomas L, Mercer JG, Wallace JM, Lea 42. Bouret SG, Simerly RB. Minireview: Leptin and development of
RG. Placental leptin. Rev Reprod 5: 18-24, 2000. hypothalamic feeding circuits. Endocrinology 145: 2621-2626, 2004.

Volume 10, July 2020 1075


Leptin in the Control of Breathing Comprehensive Physiology

43. Bradley TD, Floras JS. Obstructive sleep apnoea and its cardiovascular sympathetic coupling during chemoreflex activation. J Neurophysiol
consequences. Lancet 373: 82-93, 2009. 103: 2095-2106, 2010.
44. Briffa JF, McAinch AJ, Romano T, Wlodek ME, Hryciw DH. Leptin 68. Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath
in pregnancy and development: A contributor to adulthood disease? TL, Cone RD, Low MJ. Leptin activates anorexigenic POMC neurons
Am J Phys Endocrinol Metab 308: E335-E350, 2015. through a neural network in the arcuate nucleus. Nature 411: 480-484,
45. Buchanan GF. Impaired CO2 -induced arousal in SIDS and SUDEP. 2001.
Trends Neurosci 42: 242-250, 2019. 69. Cuhadaroglu C, Utkusavas A, Ozturk L, Salman S, Ece T. Effects of
46. Caballero-Eraso C, Shin MK, Pho H, Kim LJ, Pichard LE, Wu ZJ, Gu nasal CPAP treatment on insulin resistance, lipid profile, and plasma
C, Berger S, Pham L, Yeung HB, Shirahata M, Schwartz AR, Tang leptin in sleep apnea. Lung 187: 75-81, 2009.
WW, Sham JSK, Polotsky VY. Leptin acts in the carotid bodies to 70. Cummings KJ, Li A, Deneris ES, Nattie EE. Bradycardia in serotonin-
increase minute ventilation during wakefulness and sleep and augment deficient Pet-1−/− mice: Influence of respiratory dysfunction and
the hypoxic ventilatory response. J Physiol 597: 151-172, 2019. hyperthermia over the first 2 postnatal weeks. Am J Physiol Regul
47. Caldeira RS, Panissa VLG, Inoue DS, Campos EZ, Monteiro PA, Integr Comp Physiol 298: R1333-R1342, 2010.
Giglio BM, Pimentel GD, Hofmann P, Lira FS. Impact to short-term 71. Cundrle I Jr, Somers VK, Singh P, Johnson BD, Scott CG, Olson LJ.
high intensity intermittent training on different storages of body fat, The relationship between leptin and ventilatory control in heart failure.
leptin and soluble leptin receptor levels in physically active non-obese J Card Fail 19: 756-761, 2013.
men: A pilot investigation. Clin Nutr ESPEN 28: 186-192, 2018. 72. da Silva AA, Freeman JN, Hall JE, do Carmo JM. Control of appetite,
48. Carlin AM, Zeni TM, English WJ, Hawasli AA, Genaw JA, Krause blood glucose, and blood pressure during melanocortin-4 receptor
KR, Schram JL, Kole KL, Finks JF, Birkmeyer JD, Share D, activation in normoglycemic and diabetic NPY-deficient mice. Am J
Birkmeyer NJ, Michigan Bariatric Surgery C. The comparative effec- Physiol Regul Integr Comp Physiol 314: R533-R539, 2018.
tiveness of sleeve gastrectomy, gastric bypass, and adjustable gastric 73. Dadson K, Liu Y, Sweeney G. Adiponectin action: A combination
banding procedures for the treatment of morbid obesity. Ann Surg of endocrine and autocrine/paracrine effects. Front Endocrinol 2: 62,
257: 791-797, 2013. 2011.
49. Carrive P, Gorissen M. Premotor sympathetic neurons of conditioned 74. Daltro C, Gregorio PB, Alves E, Abreu M, Bomfim D, Chicourel MH,
fear in the rat. Eur J Neurosci 28: 428-446, 2008. Araujo L, Cotrim HP. Prevalence and severity of sleep apnea in a group
50. Carroll JL, Bureau MA. Peripheral chemoreceptor CO2 response of morbidly obese patients. Obes Surg 17: 809-814, 2007.
during hyperoxia in the 14-day-old awake lamb. Respir Physiol 73: 75. Darnall RA. The role of CO(2) and central chemoreception in the
339-349, 1988. control of breathing in the fetus and the neonate. Respir Physiol
51. Casabiell X, Pineiro V, Tome MA, Peino R, Dieguez C, Casanueva Neurobiol 173: 201-212, 2010.
FF. Presence of leptin in colostrum and/or breast milk from lactating 76. Dejours P. Control of respiration by arterial chemoreceptors. Ann N Y
mothers: A potential role in the regulation of neonatal food intake. Acad Sci 109: 682-695, 1963.
J Clin Endocrinol Metab 82: 4270-4273, 1997. 77. Del Negro CA, Funk GD, Feldman JL. Breathing matters. Nat Rev
52. Cawthorn WP, Sethi JK. TNF-alpha and adipocyte biology. FEBS Lett Neurosci 19: 351-367, 2018.
582: 117-131, 2008. 78. Deng BS, Nakamura A, Zhang W, Yanagisawa M, Fukuda Y, Kuwaki
53. Chang Z, Ballou E, Jiao W, McKenna KE, Morrison SF, McCrimmon T. Contribution of orexin in hypercapnic chemoreflex: Evidence from
DR. Systemic leptin produces a long-lasting increase in respiratory genetic and pharmacological disruption and supplementation studies
motor output in rats. Front Physiol 4: 16, 2013. in mice. J Appl Physiol 103: 1772-1779, 2007.
54. Chen H, Simar D, Morris MJ. Hypothalamic neuroendocrine circuitry 79. Devaskar SU, Ollesch C, Rajakumar RA, Rajakumar PA. Develop-
is programmed by maternal obesity: Interaction with postnatal mental changes in ob gene expression and circulating leptin peptide
nutritional environment. PLoS One 4: e6259, 2009. concentrations. Biochem Biophys Res Commun 238: 44-47, 1997.
55. Chen L, Pei JH, Chen HM. Effects of continuous positive airway pres- 80. Di Fiore JM, Martin RJ, Gauda EB. Apnea of prematurity—perfect
sure treatment on glycaemic control and insulin sensitivity in patients storm. Respir Physiol Neurobiol 189: 213-222, 2013.
with obstructive sleep apnoea and type 2 diabetes: A meta-analysis. 81. Dibner C, Gachon F. Circadian dysfunction and obesity: Is leptin the
Arch Med Sci 10: 637-642, 2014. missing link? Cell Metab 22: 359-360, 2015.
56. Chirinos JA, Gurubhagavatula I, Teff K, Rader DJ, Wadden TA, 82. Ding W, Cai Y, Wang W, Ji L, Dong Y, Zhang X, Su M, Liu J, Lu G,
Townsend R, Foster GD, Maislin G, Saif H, Broderick P, Chittams J, Zhang X. Adiponectin protects the kidney against chronic intermit-
Hanlon AL, Pack AI. CPAP, weight loss, or both for obstructive sleep tent hypoxia-induced injury through inhibiting endoplasmic reticulum
apnea. N Engl J Med 370: 2265-2275, 2014. stress. Sleep Breath 20: 1069-1074, 2016.
57. Cinaz P, Sen E, Bideci A, Ezgu FS, Atalay Y, Koca E. Plasma leptin 83. Ding W, Zhang X, Huang H, Ding N, Zhang S, Hutchinson SZ, Zhang
levels of large for gestational age and small for gestational age infants. X. Adiponectin protects rat myocardium against chronic intermittent
Acta Paediatr 88: 753-756, 1999. hypoxia-induced injury via inhibition of endoplasmic reticulum stress.
58. Ciriello J, Moreau JM. Leptin signaling in the nucleus of the solitary PLoS One 9: e94545, 2014.
tract alters the cardiovascular responses to activation of the chemore- 84. do Carmo JM, da Silva AA, Cai Z, Lin S, Dubinion JH, Hall JE. Con-
ceptor reflex. Am J Physiol Regul Integr Comp Physiol 303: R727- trol of blood pressure, appetite, and glucose by leptin in mice lacking
R736, 2012. leptin receptors in proopiomelanocortin neurons. Hypertension 57:
59. Cnop M, Havel PJ, Utzschneider KM, Carr DB, Sinha MK, Boyko 918-926, 2011.
EJ, Retzlaff BM, Knopp RH, Brunzell JD, Kahn SE. Relation- 85. do Carmo JM, da Silva AA, Moak SP, da Silva FS, Spradley FT,
ship of adiponectin to body fat distribution, insulin sensitivity and Hall JE. Role of melanocortin 4 receptor in hypertension induced by
plasma lipoproteins: Evidence for independent roles of age and sex. chronic intermittent hypoxia. Acta Physiol 225: e13222, 2019.
Diabetologia 46: 459-469, 2003. 86. Doi A, Ramirez JM. Neuromodulation and the orchestration of the
60. Coleman DL. Effects of parabiosis of obese with diabetes and normal respiratory rhythm. Respir Physiol Neurobiol 164: 96-104, 2008.
mice. Diabetologia 9: 294-298, 1973. 87. Doran AC, Meller N, Cutchins A, Deliri H, Slayton RP, Oldham SN,
61. Collin M, Hakansson-Ovesjo ML, Misane I, Ogren SO, Meister B. Kim JB, Keller SR, McNamara CA. The helix-loop-helix factors Id3
Decreased 5-HT transporter mRNA in neurons of the dorsal raphe and E47 are novel regulators of adiponectin. Circ Res 103: 624-634,
nucleus and behavioral depression in the obese leptin-deficient ob/ob 2008.
mouse. Brain Res Mol Brain Res 81: 51-61, 2000. 88. Drummond M, Winck JC, Guimaraes JT, Santos AC, Almeida J, Mar-
62. Conde SV, Monteiro EC, Sacramento JF. Purines and carotid body: ques JA. Autoadjusting-CPAP effect on serum leptin concentrations in
New roles in pathological conditions. Front Pharmacol 8: 913, 2017. obstructive sleep apnoea patients. BMC Pulm Med 8: 21, 2008.
63. Conde SV, Ribeiro MJ, Melo BF, Guarino MP, Sacramento JF. Insulin 89. Dubern B, Clement K. Leptin and leptin receptor-related monogenic
resistance: A new consequence of altered carotid body chemoreflex? obesity. Biochimie 94: 2111-2115, 2012.
J Physiol 595: 31-41, 2017. 90. Dundar NO, Anal O, Dundar B, Ozkan H, Caliskan S, Buyukgebiz
64. Conde SV, Sacramento JF, Guarino MP. Carotid body: A metabolic A. Longitudinal investigation of the relationship between breast milk
sensor implicated in insulin resistance. Physiol Genomics 50: 208-214, leptin levels and growth in breast-fed infants. J Pediatr Endocrinol
2018. Metab 18: 181-187, 2005.
65. Conde SV, Sacramento JF, Guarino MP, Gonzalez C, Obeso A, Diogo 91. Edwards BA, Sands SA, Berger PJ. Postnatal maturation of breathing
LN, Monteiro EC, Ribeiro MJ. Carotid body, insulin, and metabolic stability and loop gain: The role of carotid chemoreceptor develop-
diseases: Unraveling the links. Front Physiol 5: 418, 2014. ment. Respir Physiol Neurobiol 185: 144-155, 2013.
66. Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens 92. El-Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS. Two
TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL. defects contribute to hypothalamic leptin resistance in mice with diet-
Serum immunoreactive-leptin concentrations in normal-weight and induced obesity. J Clin Invest 105: 1827-1832, 2000.
obese humans. N Engl J Med 334: 292-295, 1996. 93. Elias CF, Kelly JF, Lee CE, Ahima RS, Drucker DJ, Saper CB,
67. Costa-Silva JH, Zoccal DB, Machado BH. Glutamatergic antagonism Elmquist JK. Chemical characterization of leptin-activated neurons in
in the NTS decreases post-inspiratory drive and changes phrenic and the rat brain. J Comp Neurol 423: 261-281, 2000.

1076 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

94. Elias CF, Lee CE, Kelly JF, Ahima RS, Kuhar M, Saper CB, Elmquist estimates peripheral chemosensitivity in OSAHS patients. Eur Respir
JK. Characterization of CART neurons in the rat and human hypotha- J 20: 724-732, 2002.
lamus. J Comp Neurol 432: 1-19, 2001. 120. Gauda E, Martin R. Control of Breathing. In: Gleason C, Juul S,
95. Elmquist JK, Maratos-Flier E, Saper CB, Flier JS. Unraveling the editors. Avery’s Diseases of the Newborn. Philadelphia, PA: Elsevier,
central nervous system pathways underlying responses to leptin. Nat 2018, p. 600-617.
Neurosci 1: 445-450, 1998. 121. Gauda EB, Carroll JL, Donnelly DF. Developmental maturation
96. Ertl T, Funke S, Sarkany I, Szabo I, Rascher W, Blum WF, Sulyok E. of chemosensitivity to hypoxia of peripheral arterial chemorecep-
Postnatal changes of leptin levels in full-term and preterm neonates: tors – invited article. Adv Exp Med Biol 648: 243-255, 2009.
Their relation to intrauterine growth, gender and testosterone. Biol 122. Gauda EB, Master Z. Contribution of relative leptin and adiponectin
Neonate 75: 167-176, 1999. deficiencies in premature infants to chronic intermittent hypoxia:
97. Essig DA, Alderson NL, Ferguson MA, Bartoli WP, Durstine JL. Exploring a new hypothesis. Respir Physiol Neurobiol 256: 119-127,
Delayed effects of exercise on the plasma leptin concentration. Metab 2018.
Clin Exp 49: 395-399, 2000. 123. Gauda EB, McLemore GL, Tolosa J, Marston-Nelson J, Kwak D. Mat-
98. Fajas L, Fruchart JC, Auwerx J. Transcriptional control of adipogene- uration of peripheral arterial chemoreceptors in relation to neonatal
sis. Curr Opin Cell Biol 10: 165-173, 1998. apnoea. Semin Neonatol 9: 181-194, 2004.
99. Famulla S, Horrighs A, Cramer A, Sell H, Eckel J. Hypoxia reduces the 124. Gerosa-Neto J, Panissa VLG, Monteiro PA, Inoue DS, Ribeiro JPJ,
response of human adipocytes towards TNFalpha resulting in reduced Figueiredo C, Zagatto AM, Little JP, Lira FS. High- or moderate-
NF-kappaB signaling and MCP-1 secretion. Int J Obes 36: 986-992, intensity training promotes change in cardiorespiratory fitness, but not
2012. visceral fat, in obese men: A randomised trial of equal energy expen-
100. Fang H, Judd RL. Adiponectin regulation and function. Compr Physiol diture exercise. Respir Physiol Neurobiol 266: 150-155, 2019.
8: 1031-1063, 2018. 125. Glavas MM, Kirigiti MA, Xiao XQ, Enriori PJ, Fisher SK, Evans AE,
101. Feldman JL, Mitchell GS, Nattie EE. Breathing: Rhythmicity, Grayson BE, Cowley MA, Smith MS, Grove KL. Early overnutrition
plasticity, chemosensitivity. Annu Rev Neurosci 26: 239-266, 2003. results in early-onset arcuate leptin resistance and increased sensitiv-
102. Finn PD, Cunningham MJ, Rickard DG, Clifton DK, Steiner RA. Sero- ity to high-fat diet. Endocrinology 151: 1598-1610, 2010.
tonergic neurons are targets for leptin in the monkey. J Clin Endocrinol 126. Gletsu-Miller N, Wright BN. Mineral malnutrition following bariatric
Metab 86: 422-426, 2001. surgery. Adv Nutr 4: 506-517, 2013.
103. Flak JN, Arble D, Pan W, Patterson C, Lanigan T, Goforth PB, 127. Global BMIMC, Di Angelantonio E, Bhupathiraju Sh N, Wormser D,
Sacksner J, Joosten M, Morgan DA, Allison MB, Hayes J, Feldman E, Gao P, Kaptoge S, Berrington de Gonzalez A, Cairns BJ, Huxley R,
Seeley RJ, Olson DP, Rahmouni K, Myers MG Jr. A leptin-regulated Jackson Ch L, Joshy G, Lewington S, Manson JE, Murphy N, Patel
circuit controls glucose mobilization during noxious stimuli. J Clin AV, Samet JM, Woodward M, Zheng W, Zhou M, Bansal N, Barri-
Invest 127: 3103-3113, 2017. carte A, Carter B, Cerhan JR, Smith GD, Fang X, Franco OH, Green J,
104. Fletcher EC, Lesske J, Qian W, Miller CC 3rd, Unger T. Repetitive, Halsey J, Hildebrand JS, Jung KJ, Korda RJ, McLerran DF, Moore SC,
episodic hypoxia causes diurnal elevation of blood pressure in rats. O’Keeffe LM, Paige E, Ramond A, Reeves GK, Rolland B, Sacerdote
Hypertension 19: 555-561, 1992. C, Sattar N, Sofianopoulou E, Stevens J, Thun M, Ueshima H, Yang
105. Fliedner S, Schulz C, Lehnert H. Brain uptake of intranasally applied L, Yun YD, Willeit P, Banks E, Beral V, Chen Z, Gapstur SM, Gunter
radioiodinated leptin in Wistar rats. Endocrinology 147: 2088-2094, MJ, Hartge P, Jee SH, Lam TH, Peto R, Potter JD, Willett WC, Thomp-
2006. son SG, Danesh J, Hu FB. Body-mass index and all-cause mortality:
106. Fortuna MG, Stornetta RL, West GH, Guyenet PG. Activation of the Individual-participant-data meta-analysis of 239 prospective studies in
retrotrapezoid nucleus by posterior hypothalamic stimulation. J Phys- four continents. Lancet 388: 776-786, 2016.
iol 587: 5121-5138, 2009. 128. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ,
107. Framnes SN, Arble DM. The bidirectional relationship between Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern
obstructive sleep apnea and metabolic disease. Front Endocrinol 9: SM, Heit JA, Howard VJ, Huffman MD, Judd SE, Kissela BM, Kit-
440, 2018.
108. Frantz ID 3rd, Adler SM, Thach BT, Taeusch HW Jr. Maturational tner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Mackey RH, Magid
effects on respiratory responses to carbon dioxide in premature infants. DJ, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER
J Appl Physiol 41: 41-45, 1976. 3rd, Moy CS, Mussolino ME, Neumar RW, Nichol G, Pandey DK,
109. Frederiksen L, Hojlund K, Hougaard DM, Mosbech TH, Larsen R, Paynter NP, Reeves MJ, Sorlie PD, Stein J, Towfighi A, Turan TN,
Flyvbjerg A, Frystyk J, Brixen K, Andersen M. Testosterone therapy Virani SS, Wong ND, Woo D, Turner MB, American Heart Associa-
decreases subcutaneous fat and adiponectin in aging men. Eur J tion Statistics Committee and Stroke Statistics Subcommittee. Heart
Endocrinol 166: 469-476, 2012. disease and stroke statistics—2014 update: A report from the Ameri-
110. Freet CS, Stoner JF, Tang X. Baroreflex and chemoreflex controls of can Heart Association. Circulation 129: e28-e292, 2014.
sympathetic activity following intermittent hypoxia. Auton Neurosci 129. Gonzalez C, Almaraz L, Obeso A, Rigual R. Oxygen and acid
Basic Clin 174: 8-14, 2013. chemoreception in the carotid body chemoreceptors. Trends Neurosci
111. Fruhbeck G. Intracellular signalling pathways activated by leptin. 15: 146-153, 1992.
Biochem J 393: 7-20, 2006. 130. Gonzalez C, Almaraz L, Obeso A, Rigual R. Carotid body chemore-
112. Fujishima Y, Maeda N, Matsuda K, Masuda S, Mori T, Fukuda S, ceptors: From natural stimuli to sensory discharges. Physiol Rev 74:
Sekimoto R, Yamaoka M, Obata Y, Kita S, Nishizawa H, Funahashi 829-898, 1994.
T, Ranscht B, Shimomura I. Adiponectin association with T-cadherin 131. Gonzalez-Martin MC, Vega-Agapito MV, Conde SV, Castaneda
protects against neointima proliferation and atherosclerosis. FASEB J J, Bustamante R, Olea E, Perez-Vizcaino F, Gonzalez C, Obeso
31: 1571-1583, 2017. A. Carotid body function and ventilatory responses in intermittent
113. Fukushi I, Yokota S, Okada Y. The role of the hypothalamus in hypoxia. Evidence for anomalous brainstem integration of arterial
modulation of respiration. Respir Physiol Neurobiol 265: 172-179, chemoreceptor input. J Cell Physiol 226: 1961-1969, 2011.
2019. 132. Goossens GH, Bizzarri A, Venteclef N, Essers Y, Cleutjens JP,
114. Furlong TM, McDowall LM, Horiuchi J, Polson JW, Dampney RA. Konings E, Jocken JW, Cajlakovic M, Ribitsch V, Clement K, Blaak
The effect of air puff stress on c-Fos expression in rat hypothalamus EE. Increased adipose tissue oxygen tension in obese compared with
and brainstem: Central circuitry mediating sympathoexcitation and lean men is accompanied by insulin resistance, impaired adipose
baroreflex resetting. Eur J Neurosci 39: 1429-1438, 2014. tissue capillarization, and inflammation. Circulation 124: 67-76,
115. Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity-induced changes in 2011.
adipose tissue microenvironment and their impact on cardiovascular 133. Gorska E, Popko K, Stelmaszczyk-Emmel A, Ciepiela O, Kucharska
disease. Circ Res 118: 1786-1807, 2016. A, Wasik M. Leptin receptors. Eur J Med Res 15 (Suppl 2): 50-54,
116. Gaines J, Vgontzas AN, Fernandez-Mendoza J, Calhoun SL, He F, 2010.
Liao D, Sawyer MD, Bixler EO. Inflammation mediates the associa- 134. Grattan DR, Ladyman SR, Augustine RA. Hormonal induction of lep-
tion between visceral adiposity and obstructive sleep apnea in adoles- tin resistance during pregnancy. Physiol Behav 91: 366-374, 2007.
cents. Am J Phys Endocrinol Metab 311: E851-E858, 2016. 135. Grill HJ, Schwartz MW, Kaplan JM, Foxhall JS, Breininger J, Baskin
117. Gamber KM, Huo L, Ha S, Hairston JE, Greeley S, Bjorbaek C. DG. Evidence that the caudal brainstem is a target for the inhibitory
Over-expression of leptin receptors in hypothalamic POMC neurons effect of leptin on food intake. Endocrinology 143: 239-246, 2002.
increases susceptibility to diet-induced obesity. PLoS One 7: e30485, 136. Groeben H, Meier S, Brown RH, O’Donnell CP, Mitzner W, Tanker-
2012. sley CG. The effect of leptin on the ventilatory responseto hyperoxia.
118. Gao L, Ortega-Saenz P, Garcia-Fernandez M, Gonzalez-Rodriguez P, Exp Lung Res 30: 559-570, 2004.
Caballero-Eraso C, Lopez-Barneo J. Glucose sensing by carotid body 137. Grove KL, Grayson BE, Glavas MM, Xiao XQ, Smith MS. Develop-
glomus cells: Potential implications in disease. Front Physiol 5: 398, ment of metabolic systems. Physiol Behav 86: 646-660, 2005.
2014. 138. Guertzenstein PG, Silver A. Fall in blood pressure produced from
119. Garcia-Rio F, Pino JM, Ramirez T, Alvaro D, Alonso A, Villasante discrete regions of the ventral surface of the medulla by glycine and
C, Villamor J. Inspiratory neural drive response to hypoxia adequately lesions. J Physiol 242: 489-503, 1974.

Volume 10, July 2020 1077


Leptin in the Control of Breathing Comprehensive Physiology

139. Guilleminault C, Quo SD. Sleep-disordered breathing. A view at the 164. Inyushkina EM, Merkulova NA, Inyushkin AN. Mechanisms of the
beginning of the new Millennium. Dent Clin N Am 45: 643-656, 2001. respiratory activity of leptin at the level of the solitary tract nucleus.
140. Guimaraes KSL, de Araujo EV, Aquino JS, Gadelha DA, Balarini CM, Neurosci Behav Physiol 40: 707-713, 2010.
Costa-Silva JH, Magnani M, Vidal H, Braga VA, de Brito Alves JL. 165. Ip MS, Lam KS, Ho C, Tsang KW, Lam W. Serum leptin and vascular
Effect of maternal dyslipidaemia on the cardiorespiratory physiology risk factors in obstructive sleep apnea. Chest 118: 580-586, 2000.
and biochemical parameters in male rat offspring. Br J Nutr 118: 930- 166. Iturriaga R, Oyarce MP, Dias ACR. Role of carotid body in intermit-
941, 2017. tent hypoxia-related hypertension. Curr Hypertens Rep 19: 38, 2017.
141. Guo R, Zhang Y, Turdi S, Ren J. Adiponectin knockout accentu- 167. Janczewski WA, Feldman JL. Distinct rhythm generators for inspira-
ates high fat diet-induced obesity and cardiac dysfunction: Role of tion and expiration in the juvenile rat. J Physiol 570: 407-420, 2006.
autophagy. Biochim Biophys Acta 1832: 1136-1148, 2013. 168. Jaquet D, Leger J, Levy-Marchal C, Oury JF, Czernichow P. Ontogeny
142. Guyenet PG. Regulation of breathing and autonomic outflows by of leptin in human fetuses and newborns: Effect of intrauterine growth
chemoreceptors. Compr Physiol 4: 1511-1562, 2014. retardation on serum leptin concentrations. J Clin Endocrinol Metab
143. Ha S, Baver S, Huo L, Gata A, Hairston J, Huntoon N, Li W, Zhang T, 83: 1243-1246, 1998.
Benecchi EJ, Ericsson M, Hentges ST, Bjorbaek C. Somato-dendritic 169. Jin G, Wang W, Kang J, Wu Y, Hou X, Yu R. Study of serum leptin
localization and signaling by leptin receptors in hypothalamic POMC level in patients with obstructive sleep apnea. Zhonghua jie he he hu
and AgRP neurons. PLoS One 8: e77622, 2013. xi za zhi 25: 204-206, 2002.
144. Hall JE, da Silva AA, do Carmo JM, Dubinion J, Hamza S, Munusamy 170. Jordan AS, McSharry DG, Malhotra A. Adult obstructive sleep
S, Smith G, Stec DE. Obesity-induced hypertension: Role of sympa- apnoea. Lancet 383: 736-747, 2014.
thetic nervous system, leptin, and melanocortins. J Biol Chem 285: 171. Kaar JL, Brinton JT, Crume T, Hamman RF, Glueck DH, Dabelea D.
17271-17276, 2010. Leptin levels at birth and infant growth: The EPOCH study. J Dev Orig
145. Hansen CS, Vistisen D, Jorgensen ME, Witte DR, Brunner EJ, Health Dis 5: 214-218, 2014.
Tabak AG, Kivimaki M, Roden M, Malik M, Herder C. Adiponectin, 172. Karvonen MK, Pesonen U, Heinonen P, Laakso M, Rissanen A,
biomarkers of inflammation and changes in cardiac autonomic Naukkarinen H, Valve R, Uusitupa MI, Koulu M. Identification of
function: Whitehall II study. Cardiovasc Diabetol 16: 153, 2017. new sequence variants in the leptin gene. J Clin Endocrinol Metab
146. Harsch IA, Konturek PC, Koebnick C, Kuehnlein PP, Fuchs FS, Pour 83: 3239-3242, 1998.
Schahin S, Wiest GH, Hahn EG, Lohmann T, Ficker JH. Leptin and 173. Kelesidis I, Mantzoros CS. Leptin and its emerging role in children
ghrelin levels in patients with obstructive sleep apnoea: Effect of CPAP and adolescents. Clin Pediatr Endocrinol 15: 1-14, 2006.
treatment. Eur Respir J 22: 251-257, 2003. 174. Kesavan K, Devaskar SU. Intrauterine growth restriction: Postnatal
147. Haselton JR, Guyenet PG. Central respiratory modulation of monitoring and outcomes. Pediatr Clin N Am 66: 403-423, 2019.
medullary sympathoexcitatory neurons in rat. Am J Phys 256: 175. Kim DK, Summers BA, Prabhakar NR, Kumar GK. Hypoxia does
R739-R750, 1989. not uniformly facilitate the release of multiple transmitters from the
148. Hauck FR, Thompson JM, Tanabe KO, Moon RY, Vennemann MM. carotid body. Adv Exp Med Biol 536: 291-296, 2003.
Breastfeeding and reduced risk of sudden infant death syndrome: A 176. Kim JY, van de Wall E, Laplante M, Azzara A, Trujillo ME, Hof-
meta-analysis. Pediatrics 128: 103-110, 2011. mann SM, Schraw T, Durand JL, Li H, Li G, Jelicks LA, Mehler MF,
149. Haynes WG. Interaction between leptin and sympathetic nervous Hui DY, Deshaies Y, Shulman GI, Schwartz GJ, Scherer PE. Obesity-
system in hypertension. Curr Hypertens Rep 2: 311-318, 2000. associated improvements in metabolic profile through expansion of
150. Hellgren G, Engstrom E, Smith LE, Lofqvist C, Hellstrom A. Effect of adipose tissue. J Clin Invest 117: 2621-2637, 2007.
preterm birth on postnatal apolipoprotein and adipocytokine profiles. 177. Kim S, Whelan J, Claycombe K, Reath DB, Moustaid-Moussa N.
Neonatology 108: 16-22, 2015. Angiotensin II increases leptin secretion by 3T3-L1 and human
151. Henry RR, Wallace P, Olefsky JM. Effects of weight loss on mech- adipocytes via a prostaglandin-independent mechanism. J Nutr 132:
anisms of hyperglycemia in obese non-insulin-dependent diabetes 1135-1140, 2002.
mellitus. Diabetes 35: 990-998, 1986. 178. Kirk SL, Samuelsson AM, Argenton M, Dhonye H, Kalamatianos T,
152. Hilaire G, Viemari JC, Coulon P, Simonneau M, Bevengut M. Modu- Poston L, Taylor PD, Coen CW. Maternal obesity induced by diet in
lation of the respiratory rhythm generator by the pontine noradrenergic rats permanently influences central processes regulating food intake in
A5 and A6 groups in rodents. Respir Physiol Neurobiol 143: 187-197, offspring. PLoS One 4: e5870, 2009.
2004. 179. Kishi T, Aschkenasi CJ, Lee CE, Mountjoy KG, Saper CB, Elmquist
153. Hitzig BM, Perng WC, Burt T, Okunieff P, Johnson DC. 1H-NMR JK. Expression of melanocortin 4 receptor mRNA in the central ner-
measurement of fractional dissociation of imidazole in intact animals. vous system of the rat. J Comp Neurol 457: 213-235, 2003.
Am J Phys 266: R1008-R1015, 1994. 180. Kokkinos P. Cardiorespiratory fitness, exercise, and blood pressure.
154. Hodson L, Humphreys SM, Karpe F, Frayn KN. Metabolic signatures Hypertension 64: 1160-1164, 2014.
of human adipose tissue hypoxia in obesity. Diabetes 62: 1417-1425, 181. Kotsis V, Stabouli S, Papakatsika S, Rizos Z, Parati G. Mechanisms
2013. of obesity-induced hypertension. Hypertens Res 33: 386-393, 2010.
155. Hoyda TD, Smith PM, Ferguson AV. Adiponectin acts in the nucleus 182. Koyama Y, Coker RH, Denny JC, Lacy DB, Jabbour K, Williams
of the solitary tract to decrease blood pressure by modulating the PE, Wasserman DH. Role of carotid bodies in control of the neuro-
excitability of neuropeptide Y neurons. Brain Res 1256: 76-84, 2009. endocrine response to exercise. Am J Phys Endocrinol Metab 281:
156. Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific E742-E748, 2001.
gene dysregulated in obesity. J Biol Chem 271: 10697-10703, 1996. 183. Koyama Y, Coker RH, Stone EE, Lacy DB, Jabbour K, Williams PE,
157. Huang XF, Han M, South T, Storlien L. Altered levels of POMC, Wasserman DH. Evidence that carotid bodies play an important role
AgRP and MC4-R mRNA expression in the hypothalamus and other in glucoregulation in vivo. Diabetes 49: 1434-1442, 2000.
parts of the limbic system of mice prone or resistant to chronic high- 184. Kugananthan S, Lai CT, Gridneva Z, Mark PJ, Geddes DT, Kakulas
energy diet-induced obesity. Brain Res 992: 9-19, 2003. F. Leptin levels are higher in whole compared to skim human milk,
158. Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in supporting a cellular contribution. Nutrients 8: 711, 2016.
the mouse. Science 153: 1127-1128, 1966. 185. Kumar P, Prabhakar NR. Peripheral chemoreceptors: Function and
159. Hutter MM, Schirmer BD, Jones DB, Ko CY, Cohen ME, Merkow plasticity of the carotid body. Compr Physiol 2: 141-219, 2012.
RP, Nguyen NT. First report from the American College of Surgeons 186. Kuwaki T. Orexinergic modulation of breathing across vigilance
Bariatric Surgery Center Network: Laparoscopic sleeve gastrectomy states. Respir Physiol Neurobiol 164: 204-212, 2008.
has morbidity and effectiveness positioned between the band and the 187. Kwak DJ, Kwak SD, Gauda EB. The effect of hyperoxia on reactive
bypass. Ann Surg 254: 410-420; discussion 420-412, 2011. oxygen species (ROS) in rat petrosal ganglion neurons during devel-
160. Iftikhar IH, Valentine CW, Bittencourt LR, Cohen DL, Fedson AC, opment using organotypic slices. Pediatr Res 60: 371-376, 2006.
Gislason T, Penzel T, Phillips CL, Yu-sheng L, Pack AI, Magalang 188. Lage M, Baldelli R, Camina JP, Rodriguez-Garci J, Penalva A,
UJ. Effects of continuous positive airway pressure on blood pressure Dieguez C, Casanueva FF. Presence of bovine leptin in edible com-
in patients with resistant hypertension and obstructive sleep apnea: mercial milk and infant formula. J Endocrinol Investig 25: 670-674,
A meta-analysis. J Hypertens 32: 2341-2350; discussion 2350, 2014. 2002.
161. Ingalls AM, Dickie MM, Snell GD. Obese, a new mutation in the house 189. Lahiri S, DeLaney RG. Relationship between carotid chemorecep-
mouse. J Hered 41: 317-318, 1950. tor activity and ventilation in the cat. Respir Physiol 24: 267-286,
162. Inoue DS, Panissa VL, Antunes BM, Oliveira FP, Malta RB, Caldeira 1975.
RS, Campos EZ, Pimentel GD, Franchini E, Lira FS. Reduced leptin 190. Lambert E, Straznicky NE, Dawood T, Ika-Sari C, Grima M, Esler
level is independent of fat mass changes and hunger scores from high- MD, Schlaich MP, Lambert GW. Change in sympathetic nerve firing
intensity intermittent plus strength training. J Sport Med Phys Fit 58: pattern associated with dietary weight loss in the metabolic syndrome.
1045-1051, 2018. Front Physiol 2: 52, 2011.
163. Inyushkin AN, Inyushkina EM, Merkulova NA. Respiratory responses 191. Lambert EA, Straznicky NE, Dixon JB, Lambert GW. Should the sym-
to microinjections of leptin into the solitary tract nucleus. Neurosci pathetic nervous system be a target to improve cardiometabolic risk in
Behav Physiol 39: 231-240, 2009. obesity? Am J Phys Heart Circ Phys 309: H244-H258, 2015.

1078 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

192. Lambert GW, Straznicky NE, Lambert EA, Dixon JB, Schlaich 216. Marchenko V, Koizumi H, Mosher B, Koshiya N, Tariq MF, Bezdud-
MP. Sympathetic nervous activation in obesity and the metabolic naya TG, Zhang R, Molkov YI, Rybak IA, Smith JC. Perturbations of
syndrome-causes, consequences and therapeutic implications. respiratory rhythm and pattern by disrupting synaptic inhibition within
Pharmacol Ther 126: 159-172, 2010. pre-Botzinger and Botzinger complexes. eNeuro 3: ENEURO.0011-
193. Landsberg L. Insulin resistance and the metabolic syndrome. 16.2016, 2016.
Diabetologia 48: 1244-1246, 2005. 217. Marin P, Rebuffe-Scrive M, Smith U, Bjorntorp P. Glucose uptake in
194. Landt M, Lawson GM, Helgeson JM, Davila-Roman VG, Ladenson human adipose tissue. Metab Clin Exp 36: 1154-1160, 1987.
JH, Jaffe AS, Hickner RC. Prolonged exercise decreases serum leptin 218. Marjanovic M, Elliott AC, Dawson MJ. The temperature dependence
concentrations. Metab Clin Exp 46: 1109-1112, 1997. of intracellular pH in isolated frog skeletal muscle: Lessons concerning
195. Lecke SB, Morsch DM, Spritzer PM. Leptin and adiponectin in the the Na(+)-H+ exchanger. J Membr Biol 161: 215-225, 1998.
female life course. Braz J Med Biol Res 44: 381-387, 2011. 219. Mark AL, Agassandian K, Morgan DA, Liu X, Cassell MD, Rahmouni
196. Leite RD, Durigan Rde C, de Souza Lino AD, de Souza Campos MV, K. Leptin signaling in the nucleus tractus solitarii increases sympa-
Souza M, Selistre-de-Araujo HS, Bouskela E, Kraemer-Aguiar LG. thetic nerve activity to the kidney. Hypertension 53: 375-380, 2009.
Resistance training may concomitantly benefit body composition, 220. Marshall JM. Peripheral chemoreceptors and cardiovascular regula-
blood pressure and muscle MMP-2 activity on the left ventricle of tion. Physiol Rev 74: 543-594, 1994.
high-fat fed diet rats. Metab Clin Exp 62: 1477-1484, 2013. 221. Martos-Moreno GA, Barrios V, Argente J. Normative data for
197. Levitzky M. Control of Breathing. In: Michael G. Levitzky, editor. adiponectin, resistin, interleukin 6, and leptin/receptor ratio in a
Pulmonary Physiology; [traduction of the Marcos Ikeda]. 6th ed. healthy Spanish pediatric population: Relationship with sex steroids.
Manole, Barueri: Sao Paulo, 2004. Eur J Endocrinol 155: 429-434, 2006.
198. Li A, Nattie E. CO2 dialysis in one chemoreceptor site, the RTN: Stim- 222. Matsuda K, Fujishima Y, Maeda N, Mori T, Hirata A, Sekimoto R,
ulus intensity and sensitivity in the awake rat. Respir Physiol Neurobiol Tsushima Y, Masuda S, Yamaoka M, Inoue K, Nishizawa H, Kita
133: 11-22, 2002. S, Ranscht B, Funahashi T, Shimomura I. Positive feedback regula-
199. Li A, Nattie E. Serotonin transporter knockout mice have a reduced tion between adiponectin and T-cadherin impacts adiponectin levels in
ventilatory response to hypercapnia (predominantly in males) but not tissue and plasma of male mice. Endocrinology 156: 934-946, 2015.
to hypoxia. J Physiol 586: 2321-2329, 2008. 223. McAllen RM. Central respiratory modulation of subretrofacial bul-
200. Li P, Cui BP, Zhang LL, Sun HJ, Liu TY, Zhu GQ. Melanocortin 3/4 bospinal neurones in the cat. J Physiol 388: 533-545, 1987.
receptors in paraventricular nucleus modulate sympathetic outflow and 224. McDowall LM, Horiuchi J, Dampney RA. Effects of disinhibition of
blood pressure. Exp Physiol 98: 435-443, 2013. neurons in the dorsomedial hypothalamus on central respiratory drive.
201. Li P, Shibata R, Unno K, Shimano M, Furukawa M, Ohashi T, Cheng Am J Physiol Regul Integr Comp Physiol 293: R1728-R1735, 2007.
X, Nagata K, Ouchi N, Murohara T. Evidence for the importance of 225. McEvoy RD, Antic NA, Heeley E, Luo Y, Ou Q, Zhang X, Mediano
adiponectin in the cardioprotective effects of pioglitazone. Hyperten- O, Chen R, Drager LF, Liu Z, Chen G, Du B, McArdle N, Mukherjee
sion 55: 69-75, 2010. S, Tripathi M, Billot L, Li Q, Lorenzi-Filho G, Barbe F, Redline S,
202. Li YL, Schultz HD. Enhanced sensitivity of Kv channels to hypoxia Wang J, Arima H, Neal B, White DP, Grunstein RR, Zhong N, Ander-
in the rabbit carotid body in heart failure: Role of angiotensin II. son CS, SAVE Investigators and Coordinators. CPAP for prevention of
J Physiol 575: 215-227, 2006. cardiovascular events in obstructive sleep apnea. N Engl J Med 375:
203. Li YL, Xia XH, Zheng H, Gao L, Li YF, Liu D, Patel KP, Wang W, 919-931, 2016.
Schultz HD. Angiotensin II enhances carotid body chemoreflex con- 226. Mesarwi OA, Sharma EV, Jun JC, Polotsky VY. Metabolic dysfunc-
trol of sympathetic outflow in chronic heart failure rabbits. Cardiovasc tion in obstructive sleep apnea: A critical examination of underlying
Res 71: 129-138, 2006. mechanisms. Sleep Biol Rhythms 13: 2-17, 2015.
204. Lin M, Liu R, Gozal D, Wead WB, Chapleau MW, Wurster R, Cheng 227. Messenger SA, Ciriello J. Effects of intermittent hypoxia on leptin
ZJ. Chronic intermittent hypoxia impairs baroreflex control of heart signalling in the carotid body. Neuroscience 232: 216-225, 2013.
rate but enhances heart rate responses to vagal efferent stimulation 228. Messenger SA, Moreau JM, Ciriello J. Intermittent hypoxia and
in anesthetized mice. Am J Phys Heart Circ Phys 293: H997-H1006, systemic leptin administration induces pSTAT3 and Fos/Fra-1 in the
2007. carotid body. Brain Res 1446: 56-70, 2012.
205. Liu M, Liu F. Transcriptional and post-translational regulation of 229. Messenger SA, Moreau JM, Ciriello J. Effect of chronic intermittent
adiponectin. Biochem J 425: 41-52, 2009. hypoxia on leptin and leptin receptor protein expression in the carotid
206. Liu Y, Chewchuk S, Lavigne C, Brule S, Pilon G, Houde V, Xu A, body. Brain Res 1513: 51-60, 2013.
Marette A, Sweeney G. Functional significance of skeletal muscle 230. Mistry AM, Swick A, Romsos DR. Leptin alters metabolic rates before
adiponectin production, changes in animal models of obesity and acquisition of its anorectic effect in developing neonatal mice. Am J
diabetes, and regulation by rosiglitazone treatment. Am J Phys Phys 277: R742-R747, 1999.
Endocrinol Metab 297: E657-E664, 2009. 231. Miyawaki T, Pilowsky P, Sun QJ, Minson J, Suzuki S, Arnolda L,
207. Loredo JS, Clausen JL, Nelesen RA, Ancoli-Israel S, Ziegler MG, Llewellyn-Smith I, Chalmers J. Central inspiration increases barosen-
Dimsdale JE. Obstructive sleep apnea and hypertension: Are periph- sitivity of neurons in rat rostral ventrolateral medulla. Am J Phys 268:
eral chemoreceptors involved? Med Hypotheses 56: 17-19, 2001. R909-R918, 1995.
208. Lu M, Fang F, Wang Z, Wei P, Hu C, Wei Y. Association between 232. Mizuno TM, Kleopoulos SP, Bergen HT, Roberts JL, Priest CA,
serum/plasma levels of adiponectin and obstructive sleep apnea Mobbs CV. Hypothalamic pro-opiomelanocortin mRNA is reduced
hypopnea syndrome: A meta-analysis. Lipids Health Dis 18: 30, by fasting and [corrected] in ob/ob and db/db mice, but is stimulated
2019. by leptin. Diabetes 47: 294-297, 1998.
209. MacFarlane PM, Vinit S, Mitchell GS. Enhancement of phrenic 233. Mizuno TM, Makimura H, Silverstein J, Roberts JL, Lopingco
long-term facilitation following repetitive acute intermittent hypoxia T, Mobbs CV. Fasting regulates hypothalamic neuropeptide Y,
is blocked by the glycolytic inhibitor 2-deoxyglucose. Am J Physiol agouti-related peptide, and proopiomelanocortin in diabetic mice
Regul Integr Comp Physiol 314: R135-R144, 2018. independent of changes in leptin or insulin. Endocrinology 140:
210. Mack SO, Kc P, Wu M, Coleman BR, Tolentino-Silva FP, Haxhiu MA. 4551-4557, 1999.
Paraventricular oxytocin neurons are involved in neural modulation of 234. Mizuno TM, Mobbs CV. Hypothalamic agouti-related protein mes-
breathing. J Appl Physiol 92: 826-834, 2002. senger ribonucleic acid is inhibited by leptin and stimulated by fasting.
211. Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y, Mat- Endocrinology 140: 814-817, 1999.
subara K. cDNA cloning and expression of a novel adipose specific 235. Mohr MA, Fairchild KD, Patel M, Sinkin RA, Clark MT, Moorman
collagen-like factor, apM1 (adipose most abundant gene transcript 1). JR, Lake DE, Kattwinkel J, Delos JB. Quantification of periodic
1996. Biochem Biophys Res Commun 425: 556-559, 2012. breathing in premature infants. Physiol Meas 36: 1415-1427, 2015.
212. Makinodan K, Yoshikawa M, Fukuoka A, Tamaki S, Koyama N, 236. Moller DE. Potential role of TNF-alpha in the pathogenesis of insulin
Yamauchi M, Tomoda K, Hamada K, Kimura H. Effect of serum resistance and type 2 diabetes. Trends Endocrinol Metab 11: 212-217,
leptin levels on hypercapnic ventilatory response in obstructive sleep 2000.
apnea. Respiration 75: 257-264, 2008. 237. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham
213. Maki-Nunes C, Toschi-Dias E, Cepeda FX, Rondon MU, Alves NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH,
MJ, Fraga RF, Braga AM, Aguilar AM, Amaro AC, Drager LF, Earley AR, Barnett AH, Prins JB, O’Rahilly S. Congenital leptin defi-
Lorenzi-Filho G, Negrao CE, Trombetta IC. Diet and exercise ciency is associated with severe early-onset obesity in humans. Nature
improve chemoreflex sensitivity in patients with metabolic syndrome 387: 903-908, 1997.
and obstructive sleep apnea. Obesity 23: 1582-1590, 2015. 238. Montesi SB, Edwards BA, Malhotra A, Bakker JP. The effect of
214. Malli F, Papaioannou AI, Gourgoulianis KI, Daniil Z. The role of continuous positive airway pressure treatment on blood pressure: A
leptin in the respiratory system: An overview. Respir Res 11: 152, systematic review and meta-analysis of randomized controlled trials.
2010. J Clin Sleep Med 8: 587-596, 2012.
215. Mansukhani MP, Kara T, Caples SM, Somers VK. Chemoreflexes, 239. Moraes DJ, Zoccal DB, Machado BH. Medullary respiratory network
sleep apnea, and sympathetic dysregulation. Curr Hypertens Rep 16: drives sympathetic overactivity and hypertension in rats submitted
476, 2014. to chronic intermittent hypoxia. Hypertension 60: 1374-1380, 2012.

Volume 10, July 2020 1079


Leptin in the Control of Breathing Comprehensive Physiology

240. Moreau JM, Ciriello J. Effects of acute intermittent hypoxia on energy 269. Pardal R, Lopez-Barneo J. Low glucose-sensing cells in the carotid
balance and hypothalamic feeding pathways. Neuroscience 253: 350- body. Nat Neurosci 5: 197-198, 2002.
360, 2013. 270. Pardal R, Ortega-Saenz P, Duran R, Lopez-Barneo J. Glia-like stem
241. Moreau JM, Messenger SA, Ciriello J. Effects of angiotensin II on cells sustain physiologic neurogenesis in the adult mammalian carotid
leptin and downstream leptin signaling in the carotid body during acute body. Cell 131: 364-377, 2007.
intermittent hypoxia. Neuroscience 310: 430-441, 2015. 271. Pasarica M, Sereda OR, Redman LM, Albarado DC, Hymel DT, Roan
242. Moreira TS, Takakura AC, Colombari E, Guyenet PG. Central LE, Rood JC, Burk DH, Smith SR. Reduced adipose tissue oxygena-
chemoreceptors and sympathetic vasomotor outflow. J Physiol 577: tion in human obesity: Evidence for rarefaction, macrophage chemo-
369-386, 2006. taxis, and inflammation without an angiogenic response. Diabetes 58:
243. Morton GJ, Schwartz MW. Leptin and the central nervous system con- 718-725, 2009.
trol of glucose metabolism. Physiol Rev 91: 389-411, 2011. 272. Patel SR, Palmer LJ, Larkin EK, Jenny NS, White DP, Redline S. Rela-
244. Munzberg H, Morrison CD. Structure, production and signaling of tionship between obstructive sleep apnea and diurnal leptin rhythms.
leptin. Metab Clin Exp 64: 13-23, 2015. Sleep 27: 235-239, 2004.
245. Murphy AM, Thomas A, Crinion SJ, Kent BD, Tambuwala MM, Fabre 273. Patel SR, White DP, Malhotra A, Stanchina ML, Ayas NT. Continuous
A, Pepin JL, Roche HM, Arnaud C, Ryan S. Intermittent hypoxia in positive airway pressure therapy for treating sleepiness in a diverse
obstructive sleep apnoea mediates insulin resistance through adipose population with obstructive sleep apnea: Results of a meta-analysis.
tissue inflammation. Eur Respir J 49 (4): 1601731, 2017. Arch Intern Med 163: 565-571, 2003.
246. Myers MG, Cowley MA, Munzberg H. Mechanisms of leptin action 274. Paz-Filho G, Mastronardi C, Delibasi T, Wong ML, Licinio J. Con-
and leptin resistance. Annu Rev Physiol 70: 537-556, 2008. genital leptin deficiency: Diagnosis and effects of leptin replacement
247. Nakano Y, Tobe T, Choi-Miura NH, Mazda T, Tomita M. Isolation therapy. Arquivos brasileiros de endocrinologia e metabologia 54:
and characterization of GBP28, a novel gelatin-binding protein puri- 690-697, 2010.
fied from human plasma. J Biochem 120: 803-812, 1996. 275. Peng YJ, Overholt JL, Kline D, Kumar GK, Prabhakar NR. Induction
248. Nambu T, Sakurai T, Mizukami K, Hosoya Y, Yanagisawa M, Goto of sensory long-term facilitation in the carotid body by intermittent
K. Distribution of orexin neurons in the adult rat brain. Brain Res 827: hypoxia: Implications for recurrent apneas. Proc Natl Acad Sci U S A
243-260, 1999. 100: 10073-10078, 2003.
249. Narkiewicz K, Somers VK. Cardiovascular variability characteristics 276. Peng YJ, Prabhakar NR. Effect of two paradigms of chronic inter-
in obstructive sleep apnea. Auton Neurosci Basic Clin 90: 89-94, 2001. mittent hypoxia on carotid body sensory activity. J Appl Physiol 96:
250. Narkiewicz K, van de Borne PJ, Pesek CA, Dyken ME, Montano N, 1236-1242; discussion 1196, 2004.
Somers VK. Selective potentiation of peripheral chemoreflex sensitiv- 277. Penumarti A, Abdel-Rahman AA. Neuronal nitric oxide synthase-
ity in obstructive sleep apnea. Circulation 99: 1183-1189, 1999. dependent elevation in adiponectin in the rostral ventrolateral medulla
251. Nattie E. CO2 , brainstem chemoreceptors and breathing. Prog Neuro- underlies g protein-coupled receptor 18-mediated hypotension in
biol 59: 299-331, 1999. conscious rats. J Pharmacol Exp Ther 351: 44-53, 2014.
252. Nattie E, Li A. Respiration and autonomic regulation and orexin. Prog 278. Peppard PE, Young T, Palta M, Dempsey J, Skatrud J. Longitudi-
Brain Res 198: 25-46, 2012. nal study of moderate weight change and sleep-disordered breathing.
253. Nattie EE, Li A. Retrotrapezoid nucleus lesions decrease phrenic activ- JAMA 284: 3015-3021, 2000.
ity and CO2 sensitivity in rats. Respir Physiol 97: 63-77, 1994. 279. Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC, Sutcliffe
254. Nattie EE, Li A. Central chemoreception in the region of the ventral JG, Kilduff TS. Neurons containing hypocretin (orexin) project to mul-
respiratory group in the rat. J Appl Physiol 81: 1987-1995, 1996. tiple neuronal systems. J Neurosci Off J Soc Neurosci 18: 9996-10015,
255. Newhouse LP, Joyner MJ, Curry TB, Laurenti MC, Man CD, Cobelli 1998.
C, Vella A, Limberg JK. Three hours of intermittent hypoxia increases 280. Phillips BG, Kato M, Narkiewicz K, Choe I, Somers VK. Increases in
circulating glucose levels in healthy adults. Phys Rep 5 (1): e13106, leptin levels, sympathetic drive, and weight gain in obstructive sleep
2017. apnea. Am J Phys Heart Circ Phys 279: H234-H237, 2000.
256. Newton AJ, Hess S, Paeger L, Vogt MC, Fleming Lascano J, Nillni EA,
Bruning JC, Kloppenburg P, Xu AW. AgRP innervation onto POMC 281. Phillips SA, Ciaraldi TP, Oh DK, Savu MK, Henry RR. Adiponectin
neurons increases with age and is accelerated with chronic high-fat secretion and response to pioglitazone is depot dependent in cultured
feeding in male mice. Endocrinology 154: 172-183, 2013. human adipose tissue. Am J Phys Endocrinol Metab 295: E842-E850,
257. Nindl BC, Kraemer WJ, Arciero PJ, Samatallee N, Leone CD, Mayo 2008.
MF, Hafeman DL. Leptin concentrations experience a delayed reduc- 282. Pierard M, Tassin A, Conotte S, Zouaoui Boudjeltia K, Legrand A.
tion after resistance exercise in men. Med Sci Sports Exerc 34: 608- Sustained intermittent hypoxemia induces adiponectin oligomers
613, 2002. redistribution and a tissue-specific modulation of adiponectin receptor
258. Nunes M, da Silva CH, Bosa VL, Bernardi JR, Werlang ICR, Goldani in mice. Front Physiol 10: 68, 2019.
MZ, NESCA Group. Could a remarkable decrease in leptin and insulin 283. Pinheiro AR, Cunha AR, Aguila MB, Mandarim-de-Lacerda CA. Ben-
levels from colostrum to mature milk contribute to early growth catch- eficial effects of physical exercise on hypertension and cardiovascular
up of SGA infants? BMC Pregnancy Childbirth 17: 410, 2017. adverse remodeling of diet-induced obese rats. Nutr Metab Cardiovasc
259. Nurse CA, Leonard EM, Salman S. Role of glial-like type II cells Dis 17: 365-375, 2007.
as paracrine modulators of carotid body chemoreception. Physiol 284. Piskuric NA, Nurse CA. Expanding role of ATP as a versatile messen-
Genomics 50: 255-262, 2018. ger at carotid and aortic body chemoreceptors. J Physiol 591: 415-422,
260. O’Donnell CP, Schaub CD, Haines AS, Berkowitz DE, Tankersley 2013.
CG, Schwartz AR, Smith PL. Leptin prevents respiratory depression 285. Platero-Luengo A, Gonzalez-Granero S, Duran R, Diaz-Castro B,
in obesity. Am J Respir Crit Care Med 159: 1477-1484, 1999. Piruat JI, Garcia-Verdugo JM, Pardal R, Lopez-Barneo J. An O2-
261. Ogden CL, Carroll MD, Fryar CD, Flegal KM. Prevalence of obesity sensitive glomus cell-stem cell synapse induces carotid body growth
among adults and youth: United States, 2011-2014. NCHS data brief, in chronic hypoxia. Cell 156: 291-303, 2014.
no 219. Hyattsville, MD: National Center for Health Statistics, 2015. 286. Polak J, Shimoda LA, Drager LF, Undem C, McHugh H, Polotsky
262. Olea E, Ribeiro MJ, Gallego-Martin T, Yubero S, Rigual R, Masa JF, VY, Punjabi NM. Intermittent hypoxia impairs glucose homeostasis in
Obeso A, Conde SV, Gonzalez C. The carotid body does not mediate C57BL6/J mice: Partial improvement with cessation of the exposure.
the acute ventilatory effects of leptin. Adv Exp Med Biol 860: 379-385, Sleep 36: 1483-1490; 1490A-1490B, 2013.
2015. 287. Polotsky VY, Wilson JA, Smaldone MC, Haines AS, Hurn PD,
263. Olive JL, Miller GD. Differential effects of maximal- and moderate- Tankersley CG, Smith PL, Schwartz AR, O’Donnell CP. Female
intensity runs on plasma leptin in healthy trained subjects. Nutrition gender exacerbates respiratory depression in leptin-deficient obesity.
17: 365-369, 2001. Am J Respir Crit Care Med 164: 1470-1475, 2001.
264. Oyamada Y, Ballantyne D, Muckenhoff K, Scheid P. Respiration- 288. Porzionato A, Rucinski M, Macchi V, Stecco C, Castagliuolo I, Mal-
modulated membrane potential and chemosensitivity of locus endowicz LK, De Caro R. Expression of leptin and leptin receptor
coeruleus neurones in the in vitro brainstem-spinal cord of the isoforms in the rat and human carotid body. Brain Res 1385: 56-67,
neonatal rat. J Physiol 513 (Pt 2): 381-398, 1998. 2011.
265. Ozturk L, Unal M, Tamer L, Celikoglu F. The association of the sever- 289. Prabhakar NR, Dick TE, Nanduri J, Kumar GK. Systemic, cellular
ity of obstructive sleep apnea with plasma leptin levels. Arch Otolaryn- and molecular analysis of chemoreflex-mediated sympathoexcitation
gol Head Neck Surg 129: 538-540, 2003. by chronic intermittent hypoxia. Exp Physiol 92: 39-44, 2007.
266. Pan WW, Myers MG Jr. Leptin and the maintenance of elevated body 290. Prabhakar NR, Peng YJ, Kumar GK, Pawar A. Altered carotid body
weight. Nat Rev Neurosci 19: 95-105, 2018. function by intermittent hypoxia in neonates and adults: Relevance to
267. Papp RS, Palkovits M. Brainstem projections of neurons located in var- recurrent apneas. Respir Physiol Neurobiol 157: 148-153, 2007.
ious subdivisions of the dorsolateral hypothalamic area-an anterograde 291. Pye RL, Dunn EJ, Ricker EM, Jurcsisn JG, Barr BL, Wyatt CN.
tract-tracing study. Front Neuroanat 8: 34, 2014. Acutely administered leptin increases [Ca2+] I and BK Ca currents
268. Parameswaran K, Todd DC, Soth M. Altered respiratory physiology but does not alter chemosensory behavior in rat carotid body type I
in obesity. Can Respir J 13: 203-210, 2006. cells. Adv Exp Med Biol 860: 61-67, 2015.

1080 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

292. Qiao L, Maclean PS, Schaack J, Orlicky DJ, Darimont C, Pagliassotti Matafome P, Conde SV. Functional abolition of carotid body activ-
M, Friedman JE, Shao J. C/EBPalpha regulates human adiponectin ity restores insulin action and glucose homeostasis in rats: Key roles
gene transcription through an intronic enhancer. Diabetes 54: 1744- for visceral adipose tissue and the liver. Diabetologia 60: 158-168,
1754, 2005. 2017.
293. Qiao L, Shao J. SIRT1 regulates adiponectin gene expression through 316. Sainz N, Barrenetxe J, Moreno-Aliaga MJ, Martinez JA. Leptin
Foxo1-C/enhancer-binding protein alpha transcriptional complex. resistance and diet-induced obesity: Central and peripheral actions of
J Biol Chem 281: 39915-39924, 2006. leptin. Metab Clin Exp 64: 35-46, 2015.
294. Rahmouni K. Leptin-induced sympathetic nerve activation: Signaling 317. Sakurai T. The neural circuit of orexin (hypocretin): Maintaining sleep
mechanisms and cardiovascular consequences in obesity. Curr Hyper- and wakefulness. Nat Rev Neurosci 8: 171-181, 2007.
tens Rev 6: 104-209, 2010. 318. Sakurai T. The role of orexin in motivated behaviours. Nat Rev Neu-
295. Rahmouni K, Correia ML, Haynes WG, Mark AL. Obesity-associated rosci 15: 719-731, 2014.
hypertension: New insights into mechanisms. Hypertension 45: 9-14, 319. Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka
2005. H, Williams SC, Richarson JA, Kozlowski GP, Wilson S, Arch JR,
296. Rakoczy RJ, Pye RL, Fayyad TH, Santin JM, Barr BL, Wyatt CN. Buckingham RE, Haynes AC, Carr SA, Annan RS, McNulty DE, Liu
High fat feeding in rats alters respiratory parameters by a mechanism WS, Terrett JA, Elshourbagy NA, Bergsma DJ, Yanagisawa M. Orex-
that is unlikely to be mediated by carotid body type I cells. Adv Exp ins and orexin receptors: A family of hypothalamic neuropeptides and
Med Biol 1071: 137-142, 2018. G protein-coupled receptors that regulate feeding behavior. Cell 92: 1
297. Rakoczy RJ, Wyatt CN. Acute oxygen sensing by the carotid body: page following 696, 1998.
A rattlebag of molecular mechanisms. J Physiol 596: 2969-2976, 320. Samuelsson AM, Clark J, Rudyk O, Shattock MJ, Bae SE, South
2018. T, Pombo J, Redington K, Uppal E, Coen CW, Poston L, Taylor
298. Ramadan W, Dewasmes G, Petitjean M, Wiernsperger N, Delanaud PD. Experimental hyperleptinemia in neonatal rats leads to selective
S, Geloen A, Libert JP. Sleep apnea is induced by a high-fat diet and leptin responsiveness, hypertension, and altered myocardial function.
reversed and prevented by metformin in non-obese rats. Obesity 15: Hypertension 62: 627-633, 2013.
1409-1418, 2007. 321. Samuelsson AM, Morris A, Igosheva N, Kirk SL, Pombo JM, Coen
299. Ramirez JM, Garcia AJ 3rd, Anderson TM, Koschnitzky JE, Peng YJ, CW, Poston L, Taylor PD. Evidence for sympathetic origins of hyper-
Kumar GK, Prabhakar NR. Central and peripheral factors contributing tension in juvenile offspring of obese rats. Hypertension 55: 76-82,
to obstructive sleep apneas. Respir Physiol Neurobiol 189: 344-353, 2010.
2013. 322. Samuelsson AS, Mullier A, Maicas N, Oosterhuis NR, Eun Bae S,
300. Reaven GM. Banting lecture 1988. Role of insulin resistance in human Novoselova TV, Chan LF, Pombo JM, Taylor PD, Joles JA, Coen CW,
disease. Diabetes 37: 1595-1607, 1988. Balthasar N, Poston L. Central role for melanocortin-4 receptors in
301. Regazzetti C, Peraldi P, Gremeaux T, Najem-Lendom R, Ben-Sahra offspring hypertension arising from maternal obesity. Proc Natl Acad
I, Cormont M, Bost F, Le Marchand-Brustel Y, Tanti JF, Giorgetti- Sci U S A 113: 12298-12303, 2016.
Peraldi S. Hypoxia decreases insulin signaling pathways in adipocytes. 323. Sanchez J, Oliver P, Miralles O, Ceresi E, Pico C, Palou A. Leptin
Diabetes 58: 95-103, 2009. orally supplied to neonate rats is directly uptaken by the immature
302. Resto M, O’Connor D, Leef K, Funanage V, Spear M, Locke R. Lep- stomach and may regulate short-term feeding. Endocrinology 146:
tin levels in preterm human breast milk and infant formula. Pediatrics 2575-2582, 2005.
108: E15, 2001. 324. Sandoval D. Bariatric surgeries: Beyond restriction and malabsorption.
303. Rey S, Del Rio R, Alcayaga J, Iturriaga R. Chronic intermittent Int J Obes 35 (Suppl 3): S45-S49, 2011.
hypoxia enhances cat chemosensory and ventilatory responses to 325. Sanner BM, Kollhosser P, Buechner N, Zidek W, Tepel M. Influence
hypoxia. J Physiol 560: 577-586, 2004. of treatment on leptin levels in patients with obstructive sleep apnoea.
304. Ribeiro MJ, Guimarães JP, Sacramento JF, Conde S. Contribution Eur Respir J 23: 601-604, 2004.
of carotid body to leptin effects on ventilation and blood pressure in 326. Savino F, Nanni GE, Maccario S, Costamagna M, Oggero R, Silve-
control and obese rats. ERJ Open Res 5 (Suppl 3): P77, 2019. stro L. Breast-fed infants have higher leptin values than formula-fed
305. Ribeiro MJ, Sacramento JF, Gallego-Martin T, Olea E, Melo BF, infants in the first four months of life. J Pediatr Endocrinol Metab 17:
Guarino MP, Yubero S, Obeso A, Conde SV. High fat diet blunts the 1527-1532, 2004.
effects of leptin on ventilation and on carotid body activity. J Physiol 327. Savransky V, Bevans S, Nanayakkara A, Li J, Smith PL, Torbenson
596: 3187-3199, 2018. MS, Polotsky VY. Chronic intermittent hypoxia causes hepatitis in a
306. Ribeiro MJ, Sacramento JF, Gonzalez C, Guarino MP, Monteiro EC, mouse model of diet-induced fatty liver. Am J Physiol Gastrointest
Conde SV. Carotid body denervation prevents the development of Liver Physiol 293: G871-G877, 2007.
insulin resistance and hypertension induced by hypercaloric diets. 328. Schafer H, Pauleit D, Sudhop T, Gouni-Berthold I, Ewig S, Berthold
Diabetes 62: 2905-2916, 2013. HK. Body fat distribution, serum leptin, and cardiovascular risk factors
307. Richter DW, Smith JC. Respiratory rhythm generation in vivo. Physi- in men with obstructive sleep apnea. Chest 122: 829-839, 2002.
ology 29: 58-71, 2014. 329. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel
308. Richter DW, Spyer KM. Studying rhythmogenesis of breathing: serum protein similar to C1q, produced exclusively in adipocytes.
Comparison of in vivo and in vitro models. Trends Neurosci 24: J Biol Chem 270: 26746-26749, 1995.
464-472, 2001. 330. Schultz HD. Angiotensin and carotid body chemoreception in heart
309. Rigatto H, Brady JP, de la Torre Verduzco R. Chemoreceptor reflexes failure. Curr Opin Pharmacol 11: 144-149, 2011.
in preterm infants: II. The effect of gestational and postnatal age on 331. Schulz C, Paulus K, Johren O, Lehnert H. Intranasal leptin reduces
the ventilatory response to inhaled carbon dioxide. Pediatrics 55: 614- appetite and induces weight loss in rats with diet-induced obesity
620, 1975. (DIO). Endocrinology 153: 143-153, 2012.
310. Roberts BL, Bennett CM, Carroll JM, Lindsley SR, Kievit P. Early 332. Schulz C, Paulus K, Lehnert H. Central nervous and metabolic effects
overnutrition alters synaptic signaling and induces leptin resistance of intranasally applied leptin. Endocrinology 145: 2696-2701, 2004.
in arcuate proopiomelanocortin neurons. Physiol Behav 206: 166-174, 333. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central
2019. nervous system control of food intake. Nature 404: 661-671, 2000.
311. Ross CA, Ruggiero DA, Joh TH, Park DH, Reis DJ. Rostral ventro- 334. Semenza GL, Prabhakar NR. Neural regulation of hypoxia-inducible
lateral medulla: Selective projections to the thoracic autonomic cell factors and redox state drives the pathogenesis of hypertension in a
column from the region containing C1 adrenaline neurons. J Comp rodent model of sleep apnea. J Appl Physiol 119: 1152-1156, 2015.
Neurol 228: 168-185, 1984. 335. Senaris R, Garcia-Caballero T, Casabiell X, Gallego R, Castro R, Con-
312. Rubinsztajn R, Kumor M, Byskiniewicz K, Bielicki P, Chazan R. sidine RV, Dieguez C, Casanueva FF. Synthesis of leptin in human
Serum leptin concentration and sympathetic activation estimated on placenta. Endocrinology 138: 4501-4504, 1997.
the adrenaline and noradrenaline serum concentration in patients with 336. Servantes DM, Javaheri S, Kravchychyn ACP, Storti LJ, Almeida DR,
obstructive sleep apnea. Pol Arch Med Wewn 113: 544-551, 2005. de Mello MT, Cintra FD, Tufik S, Bittencourt L. Effects of exercise
313. Sacramento JF, Chew DJ, Melo BF, Donega M, Dopson W, Guarino training and CPAP in patients with heart failure and OSA: A prelimi-
MP, Robinson A, Prieto-Lloret J, Patel S, Holinski BJ, Ramnarain N, nary study. Chest 154: 808-817, 2018.
Pikov V, Famm K, Conde SV. Bioelectronic modulation of carotid 337. Sharma SK, Agrawal S, Damodaran D, Sreenivas V, Kadhiravan T,
sinus nerve activity in the rat: A potential therapeutic approach for Lakshmy R, Jagia P, Kumar A. CPAP for the metabolic syndrome in
type 2 diabetes. Diabetologia 61: 700-710, 2018. patients with obstructive sleep apnea. N Engl J Med 365: 2277-2286,
314. Sacramento JF, Ribeiro MJ, Rodrigues T, Guarino MP, Diogo LN, 2011.
Seica R, Monteiro EC, Matafome P, Conde SV. Insulin resistance 338. Shechter A. Effects of continuous positive airway pressure on energy
is associated with tissue-specific regulation of HIF-1alpha and balance regulation: A systematic review. Eur Respir J 48: 1640-1657,
HIF-2alpha during mild chronic intermittent hypoxia. Respir Physiol 2016.
Neurobiol 228: 30-38, 2016. 339. Shimizu K, Chin K, Nakamura T, Masuzaki H, Ogawa Y, Hosokawa
315. Sacramento JF, Ribeiro MJ, Rodrigues T, Olea E, Melo BF, Guar- R, Niimi A, Hattori N, Nohara R, Sasayama S, Nakao K, Mishima
ino MP, Fonseca-Pinto R, Ferreira CR, Coelho J, Obeso A, Seica R, M, Nakamura T, Ohi M. Plasma leptin levels and cardiac sympathetic

Volume 10, July 2020 1081


Leptin in the Control of Breathing Comprehensive Physiology

function in patients with obstructive sleep apnoea-hypopnoea 364. Taylor PD, Samuelsson AM, Poston L. Maternal obesity and the devel-
syndrome. Thorax 57: 429-434, 2002. opmental programming of hypertension: A role for leptin. Acta Physiol
340. Shin MK, Yao Q, Jun JC, Bevans-Fonti S, Yoo DY, Han W, Mesarwi 210: 508-523, 2014.
O, Richardson R, Fu YY, Pasricha PJ, Schwartz AR, Shirahata 365. Thorp AA, Schlaich MP. Relevance of sympathetic nervous system
M, Polotsky VY. Carotid body denervation prevents fasting hyper- activation in obesity and metabolic syndrome. J Diabetes Res 2015:
glycemia during chronic intermittent hypoxia. J Appl Physiol 117: 341583, 2015.
765-776, 2014. 366. Trayhurn P. Hypoxia and adipose tissue function and dysfunction in
341. Shinohara E, Kihara S, Yamashita S, Yamane M, Nishida M, Arai obesity. Physiol Rev 93: 1-21, 2013.
T, Kotani K, Nakamura T, Takemura K, Matsuzawa Y. Visceral fat 367. Trayhurn P, Wood IS. Adipokines: Inflammation and the pleiotropic
accumulation as an important risk factor for obstructive sleep apnoea role of white adipose tissue. Br J Nutr 92: 347-355, 2004.
syndrome in obese subjects. J Intern Med 241: 11-18, 1997. 368. Trombetta IC, Batalha LT, Rondon MU, Laterza MC, Kuniyoshi FH,
342. Shirahata M, Tang WY, Shin MK, Polotsky VY. Is the carotid body a Gowdak MM, Barretto AC, Halpern A, Villares SM, Negrao CE.
metabolic monitor? Adv Exp Med Biol 860: 153-159, 2015. Weight loss improves neurovascular and muscle metaboreflex control
343. Skurk T, van Harmelen V, Blum WF, Hauner H. Angiotensin in obesity. Am J Phys Heart Circ Phys 285: H974-H982, 2003.
II promotes leptin production in cultured human fat cells by an 369. Tsuchida A, Yamauchi T, Takekawa S, Hada Y, Ito Y, Maki T,
ERK1/2-dependent pathway. Obes Res 13: 969-973, 2005. Kadowaki T. Peroxisome proliferator-activated receptor (PPAR)alpha
344. Smith HR, Leibold NK, Rappoport DA, Ginapp CM, Purnell BS, activation increases adiponectin receptors and reduces obesity-related
Bode NM, Alberico SL, Kim YC, Audero E, Gross CT, Buchanan inflammation in adipose tissue: Comparison of activation of PPARal-
GF. Dorsal raphe serotonin neurons mediate CO2 -induced arousal pha, PPARgamma, and their combination. Diabetes 54: 3358-3370,
from sleep. J Neurosci Off J Soc Neurosci 38: 1915-1925, 2018. 2005.
345. Smith JC, Abdala AP, Koizumi H, Rybak IA, Paton JF. Spatial and 370. Ueno LM, Drager LF, Rodrigues AC, Rondon MU, Braga AM, Math-
functional architecture of the mammalian brain stem respiratory ias W Jr, Krieger EM, Barretto AC, Middlekauff HR, Lorenzi-Filho G,
network: A hierarchy of three oscillatory mechanisms. J Neurophysiol Negrao CE. Effects of exercise training in patients with chronic heart
98: 3370-3387, 2007. failure and sleep apnea. Sleep 32: 637-647, 2009.
346. Smith JC, Ellenberger HH, Ballanyi K, Richter DW, Feldman JL. Pre- 371. Valuniene M, Verkauskiene R, Boguszewski M, Dahlgren J, Lasiene
Botzinger complex: A brainstem region that may generate respiratory D, Lasas L, Wikland KA. Leptin levels at birth and in early postnatal
rhythm in mammals. Science 254: 726-729, 1991. life in small- and appropriate-for-gestational-age infants. Medicina 43:
347. Smith-Kirwin SM, O’Connor DM, De Johnston J, Lancey ED, Hassink 784-791, 2007.
SG, Funanage VL. Leptin expression in human mammary epithelial 372. Van De Wielle R, Michels N. Longitudinal associations of leptin and
cells and breast milk. J Clin Endocrinol Metab 83: 1810-1813, 1998. adiponectin with heart rate variability in children. Front Physiol 8: 498,
348. Sobhani I, Bado A, Vissuzaine C, Buyse M, Kermorgant S, Laigneau 2017.
JP, Attoub S, Lehy T, Henin D, Mignon M, Lewin MJ. Leptin secretion 373. Van Eyck A, Van Hoorenbeeck K, De Winter BY, Van Gaal L, De
and leptin receptor in the human stomach. Gut 47: 178-183, 2000. Backer W, Verhulst SL. Sleep disordered breathing and autonomic
349. Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neural function in overweight and obese children and adolescents. ERJ Open
mechanisms in obstructive sleep apnea. J Clin Invest 96: 1897-1904, Res 2, 2016.
1995. 374. Varela L, Horvath TL. Leptin and insulin pathways in POMC and
350. Speretta GF, Lemes EV, Vendramini RC, Menani JV, Zoccal DB, AgRP neurons that modulate energy balance and glucose homeostasis.
Colombari E, Colombari DSA, Bassi M. High-fat diet increases EMBO Rep 13: 1079-1086, 2012.
respiratory frequency and abdominal expiratory motor activity during 375. Vgontzas AN, Papanicolaou DA, Bixler EO, Hopper K, Lotsikas A,
hypercapnia. Respir Physiol Neurobiol 258: 32-39, 2018. Lin HM, Kales A, Chrousos GP. Sleep apnea and daytime sleepiness
351. Speretta GF, Silva AA, Vendramini RC, Zanesco A, Delbin MA, and fatigue: Relation to visceral obesity, insulin resistance, and hyper-
Menani JV, Bassi M, Colombari E, Colombari DS. Resistance training cytokinemia. J Clin Endocrinol Metab 85: 1151-1158, 2000.
prevents the cardiovascular changes caused by high-fat diet. Life Sci 376. Wabitsch M, Funcke JB, Lennerz B, Kuhnle-Krahl U, Lahr G, Debatin
146: 154-162, 2016. KM, Vatter P, Gierschik P, Moepps B, Fischer-Posovszky P. Biolog-
352. Stasche N. Selective indication for positive airway pressure (PAP) ically inactive leptin and early-onset extreme obesity. N Engl J Med
in sleep-related breathing disorders with obstruction. GMS Curr Top 372: 48-54, 2015.
Otorhinolaryngol Head Neck Surg 5: Doc06, 2006. 377. Wang B, Wood IS, Trayhurn P. Hypoxia induces leptin gene expres-
353. Stefater MA, Perez-Tilve D, Chambers AP, Wilson-Perez HE, sion and secretion in human preadipocytes: Differential effects of
Sandoval DA, Berger J, Toure M, Tschop M, Woods SC, Seeley RJ. hypoxia on adipokine expression by preadipocytes. J Endocrinol 198:
Sleeve gastrectomy induces loss of weight and fat mass in obese 127-134, 2008.
rats, but does not affect leptin sensitivity. Gastroenterology 138: 378. Wang T, Hartzell DL, Rose BS, Flatt WP, Hulsey MG, Menon NK,
2426-2436, 2436 e2421-2423, 2010. Makula RA, Baile CA. Metabolic responses to intracerebroventricular
354. Steinbrekera B, Roghair R. Modeling the impact of growth and leptin leptin and restricted feeding. Physiol Behav 65: 839-848, 1999.
deficits on the neuronal regulation of blood pressure. J Endocrinol 231: 379. Wang ZV, Scherer PE. Adiponectin, the past two decades. J Mol Cell
R47-R60, 2016. Biol 8: 93-100, 2016.
355. Stunden CE, Filosa JA, Garcia AJ, Dean JB, Putnam RW. Devel- 380. Wauman J, Zabeau L, Tavernier J. The leptin receptor complex: Heav-
opment of in vivo ventilatory and single chemosensitive neuron ier than expected? Front Endocrinol 8: 30, 2017.
responses to hypercapnia in rats. Respir Physiol 127: 135-155, 2001. 381. Wehrwein EA, Basu R, Basu A, Curry TB, Rizza RA, Joyner
356. Sugerman HJ, Baron PL, Fairman RP, Evans CR, Vetrovec GW. MJ. Hyperoxia blunts counterregulation during hypoglycaemia
Hemodynamic dysfunction in obesity hypoventilation syndrome and in humans: Possible role for the carotid bodies? J Physiol 588:
the effects of treatment with surgically induced weight loss. Ann Surg 4593-4601, 2010.
207: 604-613, 1988. 382. Weiszenstein M, Shimoda LA, Koc M, Seda O, Polak J. Inhibition of
357. Sugerman HJ, Fairman RP, Sood RK, Engle K, Wolfe L, Kellum JM. lipolysis ameliorates diabetic phenotype in a mouse model of obstruc-
Long-term effects of gastric surgery for treating respiratory insuffi- tive sleep apnea. Am J Respir Cell Mol Biol 55: 299-307, 2016.
ciency of obesity. Am J Clin Nutr 55: 597S-601S, 1992. 383. Weltman A, Pritzlaff CJ, Wideman L, Considine RV, Fryburg DA,
358. Sullivan CE, Berthon-Jones M, Issa FG. Nocturnal nasal-airway pres- Gutgesell ME, Hartman ML, Veldhuis JD. Intensity of acute exercise
sure for sleep apnea. N Engl J Med 309: 112, 1983. does not affect serum leptin concentrations in young men. Med Sci
359. Summer R, Walsh K, Medoff BD. Obesity and pulmonary arte- Sports Exerc 32: 1556-1561, 2000.
rial hypertension: Is adiponectin the molecular link between these 384. Wewege M, van den Berg R, Ward RE, Keech A. The effects of high-
conditions? Pulm Circ 1: 440-447, 2011. intensity interval training vs. moderate-intensity continuous training
360. Takemura Y, Ouchi N, Shibata R, Aprahamian T, Kirber MT, Summer on body composition in overweight and obese adults: A systematic
RS, Kihara S, Walsh K. Adiponectin modulates inflammatory reac- review and meta-analysis. Obes Rev 18: 635-646, 2017.
tions via calreticulin receptor-dependent clearance of early apoptotic 385. Whitehead JP, Richards AA, Hickman IJ, Macdonald GA, Prins JB.
bodies. J Clin Invest 117: 375-386, 2007. Adiponectin—a key adipokine in the metabolic syndrome. Diabetes
361. Tan W, Janczewski WA, Yang P, Shao XM, Callaway EM, Feldman Obes Metab 8: 264-280, 2006.
JL. Silencing preBotzinger complex somatostatin-expressing neurons 386. WHO. World Health Organization Obesity and Overweight. 2018.
induces persistent apnea in awake rat. Nat Neurosci 11: 538-540, 2008. https://www.who.int/news-room/fact-sheets/detail/obesity-and-
362. Tankersley C, Kleeberger S, Russ B, Schwartz A, Smith P. Modified overweight (accessed 31 Jan 2020).
control of breathing in genetically obese (ob/ob) mice. J Appl Physiol 387. Wolf J, Hering D, Narkiewicz K. Non-dipping pattern of hypertension
81: 716-723, 1996. and obstructive sleep apnea syndrome. Hypertens Res 33: 867-871,
363. Tankersley CG, O’Donnell C, Daood MJ, Watchko JF, Mitzner W, 2010.
Schwartz A, Smith P. Leptin attenuates respiratory complications 388. Yamauchi T, Iwabu M, Okada-Iwabu M, Kadowaki T. Adiponectin
associated with the obese phenotype. J Appl Physiol 85: 2261-2269, receptors: A review of their structure, function and how they work.
1998. Best Pract Res Clin Endocrinol Metab 28: 15-23, 2014.

1082 Volume 10, July 2020


Comprehensive Physiology Leptin in the Control of Breathing

389. Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, 398. Young T, Finn L, Peppard PE, Szklo-Coxe M, Austin D, Nieto
Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K, Ohteki FJ, Stubbs R, Hla KM. Sleep disordered breathing and mortality:
T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, Terauchi Eighteen-year follow-up of the Wisconsin sleep cohort. Sleep 31:
Y, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T, Shimizu T, 1071-1078, 2008.
Nagai R, Kadowaki T. Cloning of adiponectin receptors that mediate 399. Yuan F, Wang H, Feng J, Wei Z, Yu H, Zhang X, Zhang Y, Wang S.
antidiabetic metabolic effects. Nature 423: 762-769, 2003. Leptin signaling in the carotid body regulates a hypoxic ventilatory
390. Yang Y, Yang S, Jiao X, Li J, Wu H, Sun H, Yang Y, Zhang M, Wei response through altering TASK channel expression. Front Physiol 9:
Y, Qin Y. Targeted sequencing analysis of the adiponectin gene iden- 249, 2018.
tifies variants associated with obstructive sleep apnoea in Chinese Han 400. Zhang W, Fukuda Y, Kuwaki T. Respiratory and cardiovascular
population. Medicine 98: e15219, 2019. actions of orexin-A in mice. Neurosci Lett 385: 131-136, 2005.
391. Yao Q, Pho H, Kirkness J, Ladenheim EE, Bi S, Moran TH, Fuller DD, 401. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM.
Schwartz AR, Polotsky VY. Localizing effects of leptin on upper air- Positional cloning of the mouse obese gene and its human homologue.
way and respiratory control during sleep. Sleep 39: 1097-1106, 2016. Nature 372: 425-432, 1994.
392. Ye J. Emerging role of adipose tissue hypoxia in obesity and insulin 402. Zhou SY, Gilbey MP. Respiratory-related activity of lower thoracic
resistance. Int J Obes 33: 54-66, 2009. and upper lumbar sympathetic preganglionic neurones in the rat.
393. Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for chronic J Physiol 451: 631-642, 1992.
inflammation and adiponectin reduction in adipose tissue of ob/ob and 403. Zoccal DB, Bonagamba LG, Paton JF, Machado BH. Sympathetic-
dietary obese mice. Am J Phys Endocrinol Metab 293: E1118-E1128, mediated hypertension of awake juvenile rats submitted to chronic
2007. intermittent hypoxia is not linked to baroreflex dysfunction. Exp Phys-
394. Yeh ER, Erokwu B, LaManna JC, Haxhiu MA. The paraventricular iol 94: 972-983, 2009.
nucleus of the hypothalamus influences respiratory timing and activity 404. Zoccal DB, Furuya WI, Bassi M, Colombari DS, Colombari E. The
in the rat. Neurosci Lett 232: 63-66, 1997. nucleus of the solitary tract and the coordination of respiratory and
395. Yilmaz MI, Sonmez A, Caglar K, Gok DE, Eyileten T, Yenicesu sympathetic activities. Front Physiol 5: 238, 2014.
M, Acikel C, Bingol N, Kilic S, Oguz Y, Vural A. Peroxisome 405. Zoccal DB, Silva JN, Barnett WH, Lemes EV, Falquetto B, Colom-
proliferator-activated receptor gamma (PPAR-gamma) agonist bari E, Molkov YI, Moreira TS, Takakura AC. Interaction between the
increases plasma adiponectin levels in type 2 diabetic patients with retrotrapezoid nucleus and the parafacial respiratory group to regulate
proteinuria. Endocrine 25: 207-214, 2004. active expiration and sympathetic activity in rats. Am J Physiol Lung
396. Yoshikawa M, Yamauchi M, Fujita Y, Koyama N, Fukuoka A, Tamaki Cell Mol Physiol 315: L891-L909, 2018.
S, Yamamoto Y, Tomoda K, Kimura H. The impact of obstructive 406. Zoccal DB, Simms AE, Bonagamba LG, Braga VA, Pickering AE,
sleep apnea and nasal CPAP on circulating adiponectin levels. Lung Paton JF, Machado BH. Increased sympathetic outflow in juvenile rats
192: 289-295, 2014. submitted to chronic intermittent hypoxia correlates with enhanced
397. Yosunkaya S, Okur HK, Can U, Zamani A, Kutlu R. Impact of con- expiratory activity. J Physiol 586: 3253-3265, 2008.
tinuous positive airway pressure treatment on leptin levels in patients
with obstructive sleep apnea syndrome. Metab Syndr Relat Disord 13:
272-277, 2015.

Volume 10, July 2020 1083

You might also like