Comprehensive Physiology Wiley Online Library

Leptin: Master Regulator of Biological Functions that Affects Breathing

Full Article on Wiley Online Library



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 hypertension. 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 circuits 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.

Figure 1. 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 provide 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.
Figure 2. 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.
Figure 3. 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.
Figure 4. Figure 4. Anatomical localization of serotonin (SERT) and Ob‐R mRNA expression in the rostral brainstem of the monkey. Plates represent sections containing five regions of Raphe nuclei included in the analysis: CLi (A), rostral DR (rDR; B), rostral MR (rMR; B), caudal DR (cDR; C), and caudal MR (cMR; C). Symbols represent the locations of cells containing SERT mRNA (gray triangles), Ob‐R mRNA (empty inverted triangles), or both SERT/Ob‐R mRNAs (black circles). Aq, cerebral aqueduct; CG, central gray; C3, oculomotor nucleus; mlf, medial longitudinal fasciculus; xscp, decussation of superior cerebellar peduncle; IC, inferior colliculus; scp, superior cerebellar peduncle; 4V, fourth ventricle; LC, locus coeruleus. Adapted, with permission, from Finn PD, et al., 2001 102.
Figure 5. Figure 5. Schematic diagram depicting the interrelationship between weight‐dependent and physiological‐dependent mechanisms on metabolic and cardiorespiratory 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, inflammation, or other mechanisms. Adapted, with permission, from Framnes SN and Arble DM, 2018 107.
Figure 6. 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.
Figure 7. 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
Figure 8. 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 neuropeptide 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 brainstem 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 sympathetic 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, arterial 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, paraventricular 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.
Figure 9. 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 arcuate 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.
Figure 10. Figure 10. Serum leptin concentrations in cord blood (ng/mL) according to gestational age (weeks) in fetuses and newborns. Blue symbol represents values obtained from fetuses and newborns with normal growth; red circles, represents values from fetuses and newborns with IUGR. Modified, with permission, from Jaquet D, et al., 1998 168.
Figure 11. Figure 11. Overnutrition and excessive adiposity during pregnancy and lactation alter the fetal programming leading to obesity and hypertension. The altered leptin signaling in the hypothalamus promotes a selective leptin resistance in which the anorexic effects of leptin are lost, whilst the pressor effect of leptin is enhanced. Adapted, with permission, from Taylor PD, et al., 2014 364.
Figure 12. 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 depolarization, 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.
Figure 13. Figure 13. Diagram representing the involvement of carotid body in leptin effects in the control of breathing and on sympathetic overactivation that participates in genesis of insulin resistance and hypertension. Hyperleptinemia produced by adipose tissue dysfunction induced by dysmetabolism, obesity and/or by chronic intermittent hypoxia originates an increase in carotid body sensitization that is on the basis of increased spontaneous ventilation, increased hypoxic ventilatory response, and augmented sympathetic activity.
Figure 14. Figure 14. Graphic depiction of types of bariatric surgery. Printed, with permission, from Gletsu‐Miller N and Wright BN, 2013 126.
Figure 15. Figure 15. Schematic showing adiponectin signaling in macrophages, liver, endothelial and muscle cells. Adiponectin is produced by adipose tissue, 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 element; COX‐2, cyclooxygenase‐2; VCAM‐1, vascular cell adhesion protein 1; PPARγ, peroxisome proliferator‐activated receptor gamma; BMI, body mass index; IL, Interleukin. Adapted, with permission, from Summer R, et al., 2011 359.
Figure 16. Figure 16. Low‐power photomicrograph, using Hoffman contrast microscopy, of an organotypic slice of the carotid body 24 h in culture. Fat cells are in close proximity to the CB in vivo. Tissue was removed from a Sprague Dawley rat at 2 weeks postnatal age. CB, carotid body; CA, carotid artery. Arrows depict fat cells (FC). Adapted, with permission, from Kwak DJ, et al., 2006 187.


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 provide 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.


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.


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.


Figure 4. Anatomical localization of serotonin (SERT) and Ob‐R mRNA expression in the rostral brainstem of the monkey. Plates represent sections containing five regions of Raphe nuclei included in the analysis: CLi (A), rostral DR (rDR; B), rostral MR (rMR; B), caudal DR (cDR; C), and caudal MR (cMR; C). Symbols represent the locations of cells containing SERT mRNA (gray triangles), Ob‐R mRNA (empty inverted triangles), or both SERT/Ob‐R mRNAs (black circles). Aq, cerebral aqueduct; CG, central gray; C3, oculomotor nucleus; mlf, medial longitudinal fasciculus; xscp, decussation of superior cerebellar peduncle; IC, inferior colliculus; scp, superior cerebellar peduncle; 4V, fourth ventricle; LC, locus coeruleus. Adapted, with permission, from Finn PD, et al., 2001 102.


Figure 5. Schematic diagram depicting the interrelationship between weight‐dependent and physiological‐dependent mechanisms on metabolic and cardiorespiratory 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, inflammation, or other mechanisms. Adapted, with permission, from Framnes SN and Arble DM, 2018 107.


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.


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


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 neuropeptide 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 brainstem 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 sympathetic 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, arterial 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, paraventricular 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.


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 arcuate 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.


Figure 10. Serum leptin concentrations in cord blood (ng/mL) according to gestational age (weeks) in fetuses and newborns. Blue symbol represents values obtained from fetuses and newborns with normal growth; red circles, represents values from fetuses and newborns with IUGR. Modified, with permission, from Jaquet D, et al., 1998 168.


Figure 11. Overnutrition and excessive adiposity during pregnancy and lactation alter the fetal programming leading to obesity and hypertension. The altered leptin signaling in the hypothalamus promotes a selective leptin resistance in which the anorexic effects of leptin are lost, whilst the pressor effect of leptin is enhanced. Adapted, with permission, from Taylor PD, et al., 2014 364.


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 depolarization, 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.


Figure 13. Diagram representing the involvement of carotid body in leptin effects in the control of breathing and on sympathetic overactivation that participates in genesis of insulin resistance and hypertension. Hyperleptinemia produced by adipose tissue dysfunction induced by dysmetabolism, obesity and/or by chronic intermittent hypoxia originates an increase in carotid body sensitization that is on the basis of increased spontaneous ventilation, increased hypoxic ventilatory response, and augmented sympathetic activity.


Figure 14. Graphic depiction of types of bariatric surgery. Printed, with permission, from Gletsu‐Miller N and Wright BN, 2013 126.


Figure 15. Schematic showing adiponectin signaling in macrophages, liver, endothelial and muscle cells. Adiponectin is produced by adipose tissue, 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 element; COX‐2, cyclooxygenase‐2; VCAM‐1, vascular cell adhesion protein 1; PPARγ, peroxisome proliferator‐activated receptor gamma; BMI, body mass index; IL, Interleukin. Adapted, with permission, from Summer R, et al., 2011 359.


Figure 16. Low‐power photomicrograph, using Hoffman contrast microscopy, of an organotypic slice of the carotid body 24 h in culture. Fat cells are in close proximity to the CB in vivo. Tissue was removed from a Sprague Dawley rat at 2 weeks postnatal age. CB, carotid body; CA, carotid artery. Arrows depict fat cells (FC). Adapted, with permission, from Kwak DJ, et al., 2006 187.
References
 1.Abdala AP, Rybak IA, Smith JC, Paton JF. Abdominal expiratory activity in the rat brainstem‐spinal cord in situ: Patterns, origins and implications for respiratory rhythm generation. J Physiol 587: 3539‐3559, 2009.
 2.Abraham KA, Feingold H, Fuller DD, Jenkins M, Mateika JH, Fregosi RF. Respiratory‐related activation of human abdominal muscles during exercise. J Physiol 541: 653‐663, 2002.
 3.Ahima RS, Saper CB, Flier JS, Elmquist JK. Leptin regulation of neuroendocrine systems. Front Neuroendocrinol 21: 263‐307, 2000.
 4.Al Mutairi S, Mojiminiyi OA, Al Alawi A, Al Rammah T, Abdella N. Study of leptin and adiponectin as disease markers in subjects with obstructive sleep apnea. Dis Markers 2014: 706314, 2014.
 5.Allen AM. Angiotensin AT1 receptor‐mediated excitation of rat carotid body chemoreceptor afferent activity. J Physiol 510 (Pt 3): 773‐781, 1998.
 6.Alvarez‐Buylla R, Alvarez‐Buylla E, Mendoza H, Montero SA, Alvarez‐Buylla A. Pituitary and adrenals are required for hyperglycemic reflex initiated by stimulation of CBR with cyanide. Am J Phys 272: R392‐R399, 1997.
 7.Alvarez‐Buylla R, de Alvarez‐Buylla ER. Carotid sinus receptors participate in glucose homeostasis. Respir Physiol 72: 347‐359, 1988.
 8.Alvarez‐Buylla R, de Alvarez‐Buylla ER. Changes in blood glucose concentration in the carotid body modify brain glucose retention. Adv Exp Med Biol 360: 293‐296, 1994.
 9.Ambati S, Duan J, Duff E, Choi YH, Hartzell DL, Della‐Fera MA, Baile CA. Gene expression in arcuate nucleus‐median eminence of rats treated with leptin or ciliary neurotrophic factor. Biofactors 31: 133‐144, 2007.
 10.Anderson TM, Garcia AJ 3rd, Baertsch NA, Pollak J, Bloom JC, Wei AD, Rai KG, Ramirez JM. A novel excitatory network for the control of breathing. Nature 536: 76‐80, 2016.
 11.Anderson TM, Ramirez JM. Respiratory rhythm generation: Triple oscillator hypothesis. F1000Research 6: 139, 2017.
 12.Andresen MC, Doyle MW, Bailey TW, Jin YH. Differentiation of autonomic reflex control begins with cellular mechanisms at the first synapse within the nucleus tractus solitarius. Braz J Med Biol Res 37: 549‐558, 2004.
 13.Arble DM, Sandoval DA, Seeley RJ. Mechanisms underlying weight loss and metabolic improvements in rodent models of bariatric surgery. Diabetologia 58: 211‐220, 2015.
 14.Arble DM, Schwartz AR, Polotsky VY, Sandoval DA, Seeley RJ. Vertical sleeve gastrectomy improves ventilatory drive through a leptin‐dependent mechanism. JCI Insight 4 (1): e124469, 2019.
 15.Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y. Paradoxical decrease of an adipose‐specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 257: 79‐83, 1999.
 16.Arnold AC, Shaltout HA, Gallagher PE, Diz DI. Leptin impairs cardiovagal baroreflex function at the level of the solitary tract nucleus. Hypertension 54: 1001‐1008, 2009.
 17.Ashworth CJ, Hoggard N, Thomas L, Mercer JG, Wallace JM, Lea RG. Placental leptin. Rev Reprod 5: 18‐24, 2000.
 18.Athanasakis E, Karavasiliadou S, Styliadis I. The factors contributing to the risk of sudden infant death syndrome. Hippokratia 15: 127‐131, 2011.
 19.Attig L, Brisard D, Larcher T, Mickiewicz M, Guilloteau P, Boukthir S, Niamba CN, Gertler A, Djiane J, Monniaux D, Abdennebi‐Najar L. 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 hypothalamic 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.
 21.Attig L, Larcher T, Gertler A, Abdennebi‐Najar L, Djiane J. Postnatal leptin is necessary for maturation of numerous organs in newborn rats. Organogenesis 7: 88‐94, 2011.
 22.Babu AR, Herdegen J, Fogelfeld L, Shott S, Mazzone T. Type 2 diabetes, glycemic control, and continuous positive airway pressure in obstructive sleep apnea. Arch Intern Med 165: 447‐452, 2005.
 23.Baquero AF, de Solis AJ, Lindsley SR, Kirigiti MA, Smith MS, Cowley MA, Zeltser LM, Grove KL. Developmental switch of leptin signaling in arcuate nucleus neurons. J Neurosci Off J Soc Neurosci 34: 9982‐9994, 2014.
 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 leptin 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.
 26.Bassi M, Furuya WI, Menani JV, Colombari DS, do Carmo JM, da Silva AA, Hall JE, Moreira TS, Wenker IC, Mulkey DK, Colombari E. Leptin into the ventrolateral medulla facilitates chemorespiratory response in leptin‐deficient (ob/ob) mice. Acta Physiol 211: 240‐248, 2014.
 27.Bassi M, Furuya WI, Zoccal DB, Menani JV, Colombari DS, Mulkey DK, Colombari E. Facilitation of breathing by leptin effects in the central nervous system. J Physiol 594: 1617‐1625, 2016.
 28.Bassi M, Furuya WI, Zoccal DB, Menani JV, Colombari E, Hall JE, da Silva AA, do Carmo JM, Colombari DS. Control of respiratory and cardiovascular functions by leptin. Life Sci 125: 25‐31, 2015.
 29.Bassi M, Giusti H, Leite CM, Anselmo‐Franci JA, do Carmo JM, da Silva AA, Hall JE, Colombari E, Glass ML. Central leptin replacement enhances chemorespiratory responses in leptin‐deficient mice independent of changes in body weight. Pflugers Arch Eur J Physiol 464: 145‐153, 2012.
 30.Bassi M, Nakamura NB, Furuya WI, Colombari DS, Menani JV, do Carmo JM, da Silva AA, Hall JE, Colombari E. Activation of the brain melanocortin system is required for leptin‐induced modulation of chemorespiratory function. Acta Physiol 213: 893‐901, 2015.
 31.Batacan RB Jr, Duncan MJ, Dalbo VJ, Tucker PS, Fenning AS. Effects of high‐intensity interval training on cardiometabolic health: A systematic review and meta‐analysis of intervention studies. Br J Sports Med 51: 494‐503, 2017.
 32.Bell BB, Rahmouni K. Leptin as a mediator of obesity‐induced hypertension. Curr Obes Rep 5: 397‐404, 2016.
 33.Berger S, Pho H, Fleury‐Curado T, Bevans‐Fonti S, Younas H, Shin MK, Jun JC, Anokye‐Danso F, Ahima RS, Enquist LW, Mendelowitz D, Schwartz AR, Polotsky VY. Intranasal leptin relieves sleep‐disordered breathing in mice with diet‐induced obesity. Am J Respir Crit Care Med 199: 773‐783, 2019.
 34.Bernardi L, Bianchi L. Integrated cardio‐respiratory control: Insight in diabetes. Curr Diab Rep 16: 107, 2016.
 35.Bianchi AL, Denavit‐Saubie M, Champagnat J. Central control of breathing in mammals: Neuronal circuitry, membrane properties, and neurotransmitters. Physiol Rev 75: 1‐45, 1995.
 36.Biro FM, Wien M. Childhood obesity and adult morbidities. Am J Clin Nutr 91: 1499S‐1505S, 2010.
 37.Blackmore HL, Niu Y, Fernandez‐Twinn DS, Tarry‐Adkins JL, Giussani DA, Ozanne SE. Maternal diet‐induced obesity programs cardiovascular dysfunction in adult male mouse offspring independent of current body weight. Endocrinology 155: 3970‐3980, 2014.
 38.Bluher S, Kapplinger J, Herget S, Reichardt S, Bottcher Y, Grimm A, Kratzsch J, Petroff D. Cardiometabolic risk markers, adipocyte fatty acid binding protein (aFABP) and the impact of high‐intensity interval training (HIIT) in obese adolescents. Metab Clin Exp 68: 77‐87, 2017.
 39.Bonsignore MR, Borel AL, Machan E, Grunstein R. Sleep apnoea and metabolic dysfunction. Eur Respir Rev 22: 353‐364, 2013.
 40.Bouassida A, Zalleg D, Bouassida S, Zaouali M, Feki Y, Zbidi A, Tabka Z. Leptin, its implication in physical exercise and training: A short review. J Sports Sci Med 5: 172‐181, 2006.
 41.Bouret SG, Gorski JN, Patterson CM, Chen S, Levin BE, Simerly RB. Hypothalamic neural projections are permanently disrupted in diet‐induced obese rats. Cell Metab 7: 179‐185, 2008.
 42.Bouret SG, Simerly RB. Minireview: Leptin and development of hypothalamic feeding circuits. Endocrinology 145: 2621‐2626, 2004.
 43.Bradley TD, Floras JS. Obstructive sleep apnoea and its cardiovascular consequences. Lancet 373: 82‐93, 2009.
 44.Briffa JF, McAinch AJ, Romano T, Wlodek ME, Hryciw DH. Leptin in pregnancy and development: A contributor to adulthood disease? Am J Phys Endocrinol Metab 308: E335‐E350, 2015.
 45.Buchanan GF. Impaired CO2‐induced arousal in SIDS and SUDEP. Trends Neurosci 42: 242‐250, 2019.
 46.Caballero‐Eraso C, Shin MK, Pho H, Kim LJ, Pichard LE, Wu ZJ, Gu C, Berger S, Pham L, Yeung HB, Shirahata M, Schwartz AR, Tang WW, Sham JSK, Polotsky VY. Leptin acts in the carotid bodies to increase minute ventilation during wakefulness and sleep and augment the hypoxic ventilatory response. J Physiol 597: 151‐172, 2019.
 47.Caldeira RS, Panissa VLG, Inoue DS, Campos EZ, Monteiro PA, Giglio BM, Pimentel GD, Hofmann P, Lira FS. Impact to short‐term high intensity intermittent training on different storages of body fat, leptin and soluble leptin receptor levels in physically active non‐obese men: A pilot investigation. Clin Nutr ESPEN 28: 186‐192, 2018.
 48.Carlin AM, Zeni TM, English WJ, Hawasli AA, Genaw JA, Krause KR, Schram JL, Kole KL, Finks JF, Birkmeyer JD, Share D, Birkmeyer NJ, Michigan Bariatric Surgery C. The comparative effectiveness of sleeve gastrectomy, gastric bypass, and adjustable gastric banding procedures for the treatment of morbid obesity. Ann Surg 257: 791‐797, 2013.
 49.Carrive P, Gorissen M. Premotor sympathetic neurons of conditioned fear in the rat. Eur J Neurosci 28: 428‐446, 2008.
 50.Carroll JL, Bureau MA. Peripheral chemoreceptor CO2 response during hyperoxia in the 14‐day‐old awake lamb. Respir Physiol 73: 339‐349, 1988.
 51.Casabiell X, Pineiro V, Tome MA, Peino R, Dieguez C, Casanueva FF. Presence of leptin in colostrum and/or breast milk from lactating mothers: A potential role in the regulation of neonatal food intake. J Clin Endocrinol Metab 82: 4270‐4273, 1997.
 52.Cawthorn WP, Sethi JK. TNF‐alpha and adipocyte biology. FEBS Lett 582: 117‐131, 2008.
 53.Chang Z, Ballou E, Jiao W, McKenna KE, Morrison SF, McCrimmon DR. Systemic leptin produces a long‐lasting increase in respiratory motor output in rats. Front Physiol 4: 16, 2013.
 54.Chen H, Simar D, Morris MJ. Hypothalamic neuroendocrine circuitry is programmed by maternal obesity: Interaction with postnatal nutritional environment. PLoS One 4: e6259, 2009.
 55.Chen L, Pei JH, Chen HM. Effects of continuous positive airway pressure treatment on glycaemic control and insulin sensitivity in patients with obstructive sleep apnoea and type 2 diabetes: A meta‐analysis. Arch Med Sci 10: 637‐642, 2014.
 56.Chirinos JA, Gurubhagavatula I, Teff K, Rader DJ, Wadden TA, Townsend R, Foster GD, Maislin G, Saif H, Broderick P, Chittams J, Hanlon AL, Pack AI. CPAP, weight loss, or both for obstructive sleep apnea. N Engl J Med 370: 2265‐2275, 2014.
 57.Cinaz P, Sen E, Bideci A, Ezgu FS, Atalay Y, Koca E. Plasma leptin levels of large for gestational age and small for gestational age infants. Acta Paediatr 88: 753‐756, 1999.
 58.Ciriello J, Moreau JM. Leptin signaling in the nucleus of the solitary tract alters the cardiovascular responses to activation of the chemoreceptor reflex. Am J Physiol Regul Integr Comp Physiol 303: R727‐R736, 2012.
 59.Cnop M, Havel PJ, Utzschneider KM, Carr DB, Sinha MK, Boyko EJ, Retzlaff BM, Knopp RH, Brunzell JD, Kahn SE. Relationship of adiponectin to body fat distribution, insulin sensitivity and plasma lipoproteins: Evidence for independent roles of age and sex. Diabetologia 46: 459‐469, 2003.
 60.Coleman DL. Effects of parabiosis of obese with diabetes and normal mice. Diabetologia 9: 294‐298, 1973.
 61.Collin M, Hakansson‐Ovesjo ML, Misane I, Ogren SO, Meister B. Decreased 5‐HT transporter mRNA in neurons of the dorsal raphe nucleus and behavioral depression in the obese leptin‐deficient ob/ob mouse. Brain Res Mol Brain Res 81: 51‐61, 2000.
 62.Conde SV, Monteiro EC, Sacramento JF. Purines and carotid body: New roles in pathological conditions. Front Pharmacol 8: 913, 2017.
 63.Conde SV, Ribeiro MJ, Melo BF, Guarino MP, Sacramento JF. Insulin resistance: A new consequence of altered carotid body chemoreflex? J Physiol 595: 31‐41, 2017.
 64.Conde SV, Sacramento JF, Guarino MP. Carotid body: A metabolic sensor implicated in insulin resistance. Physiol Genomics 50: 208‐214, 2018.
 65.Conde SV, Sacramento JF, Guarino MP, Gonzalez C, Obeso A, Diogo LN, Monteiro EC, Ribeiro MJ. Carotid body, insulin, and metabolic diseases: Unraveling the links. Front Physiol 5: 418, 2014.
 66.Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW, Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL. Serum immunoreactive‐leptin concentrations in normal‐weight and obese humans. N Engl J Med 334: 292‐295, 1996.
 67.Costa‐Silva JH, Zoccal DB, Machado BH. Glutamatergic antagonism in the NTS decreases post‐inspiratory drive and changes phrenic and sympathetic coupling during chemoreflex activation. J Neurophysiol 103: 2095‐2106, 2010.
 68.Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath TL, Cone RD, Low MJ. Leptin activates anorexigenic POMC neurons through a neural network in the arcuate nucleus. Nature 411: 480‐484, 2001.
 69.Cuhadaroglu C, Utkusavas A, Ozturk L, Salman S, Ece T. Effects of nasal CPAP treatment on insulin resistance, lipid profile, and plasma leptin in sleep apnea. Lung 187: 75‐81, 2009.
 70.Cummings KJ, Li A, Deneris ES, Nattie EE. Bradycardia in serotonin‐deficient Pet‐1−/− mice: Influence of respiratory dysfunction and hyperthermia over the first 2 postnatal weeks. Am J Physiol Regul Integr Comp Physiol 298: R1333‐R1342, 2010.
 71.Cundrle I Jr, Somers VK, Singh P, Johnson BD, Scott CG, Olson LJ. The relationship between leptin and ventilatory control in heart failure. J Card Fail 19: 756‐761, 2013.
 72.da Silva AA, Freeman JN, Hall JE, do Carmo JM. Control of appetite, blood glucose, and blood pressure during melanocortin‐4 receptor activation in normoglycemic and diabetic NPY‐deficient mice. Am J Physiol Regul Integr Comp Physiol 314: R533‐R539, 2018.
 73.Dadson K, Liu Y, Sweeney G. Adiponectin action: A combination of endocrine and autocrine/paracrine effects. Front Endocrinol 2: 62, 2011.
 74.Daltro C, Gregorio PB, Alves E, Abreu M, Bomfim D, Chicourel MH, Araujo L, Cotrim HP. Prevalence and severity of sleep apnea in a group of morbidly obese patients. Obes Surg 17: 809‐814, 2007.
 75.Darnall RA. The role of CO(2) and central chemoreception in the control of breathing in the fetus and the neonate. Respir Physiol Neurobiol 173: 201‐212, 2010.
 76.Dejours P. Control of respiration by arterial chemoreceptors. Ann N Y Acad Sci 109: 682‐695, 1963.
 77.Del Negro CA, Funk GD, Feldman JL. Breathing matters. Nat Rev Neurosci 19: 351‐367, 2018.
 78.Deng BS, Nakamura A, Zhang W, Yanagisawa M, Fukuda Y, Kuwaki T. Contribution of orexin in hypercapnic chemoreflex: Evidence from genetic and pharmacological disruption and supplementation studies in mice. J Appl Physiol 103: 1772‐1779, 2007.
 79.Devaskar SU, Ollesch C, Rajakumar RA, Rajakumar PA. Developmental changes in ob gene expression and circulating leptin peptide concentrations. Biochem Biophys Res Commun 238: 44‐47, 1997.
 80.Di Fiore JM, Martin RJ, Gauda EB. Apnea of prematurity—perfect storm. Respir Physiol Neurobiol 189: 213‐222, 2013.
 81.Dibner C, Gachon F. Circadian dysfunction and obesity: Is leptin the missing link? Cell Metab 22: 359‐360, 2015.
 82.Ding W, Cai Y, Wang W, Ji L, Dong Y, Zhang X, Su M, Liu J, Lu G, Zhang X. Adiponectin protects the kidney against chronic intermittent hypoxia‐induced injury through inhibiting endoplasmic reticulum stress. Sleep Breath 20: 1069‐1074, 2016.
 83.Ding W, Zhang X, Huang H, Ding N, Zhang S, Hutchinson SZ, Zhang X. Adiponectin protects rat myocardium against chronic intermittent hypoxia‐induced injury via inhibition of endoplasmic reticulum stress. PLoS One 9: e94545, 2014.
 84.do Carmo JM, da Silva AA, Cai Z, Lin S, Dubinion JH, Hall JE. Control of blood pressure, appetite, and glucose by leptin in mice lacking leptin receptors in proopiomelanocortin neurons. Hypertension 57: 918‐926, 2011.
 85.do Carmo JM, da Silva AA, Moak SP, da Silva FS, Spradley FT, Hall JE. Role of melanocortin 4 receptor in hypertension induced by chronic intermittent hypoxia. Acta Physiol 225: e13222, 2019.
 86.Doi A, Ramirez JM. Neuromodulation and the orchestration of the respiratory rhythm. Respir Physiol Neurobiol 164: 96‐104, 2008.
 87.Doran AC, Meller N, Cutchins A, Deliri H, Slayton RP, Oldham SN, Kim JB, Keller SR, McNamara CA. The helix‐loop‐helix factors Id3 and E47 are novel regulators of adiponectin. Circ Res 103: 624‐634, 2008.
 88.Drummond M, Winck JC, Guimaraes JT, Santos AC, Almeida J, Marques JA. Autoadjusting‐CPAP effect on serum leptin concentrations in obstructive sleep apnoea patients. BMC Pulm Med 8: 21, 2008.
 89.Dubern B, Clement K. Leptin and leptin receptor‐related monogenic obesity. Biochimie 94: 2111‐2115, 2012.
 90.Dundar NO, Anal O, Dundar B, Ozkan H, Caliskan S, Buyukgebiz A. Longitudinal investigation of the relationship between breast milk leptin levels and growth in breast‐fed infants. J Pediatr Endocrinol Metab 18: 181‐187, 2005.
 91.Edwards BA, Sands SA, Berger PJ. Postnatal maturation of breathing stability and loop gain: The role of carotid chemoreceptor development. Respir Physiol Neurobiol 185: 144‐155, 2013.
 92.El‐Haschimi K, Pierroz DD, Hileman SM, Bjorbaek C, Flier JS. Two defects contribute to hypothalamic leptin resistance in mice with diet‐induced obesity. J Clin Invest 105: 1827‐1832, 2000.
 93.Elias CF, Kelly JF, Lee CE, Ahima RS, Drucker DJ, Saper CB, Elmquist JK. Chemical characterization of leptin‐activated neurons in the rat brain. J Comp Neurol 423: 261‐281, 2000.
 94.Elias CF, Lee CE, Kelly JF, Ahima RS, Kuhar M, Saper CB, Elmquist JK. Characterization of CART neurons in the rat and human hypothalamus. J Comp Neurol 432: 1‐19, 2001.
 95.Elmquist JK, Maratos‐Flier E, Saper CB, Flier JS. Unraveling the central nervous system pathways underlying responses to leptin. Nat Neurosci 1: 445‐450, 1998.
 96.Ertl T, Funke S, Sarkany I, Szabo I, Rascher W, Blum WF, Sulyok E. Postnatal changes of leptin levels in full‐term and preterm neonates: Their relation to intrauterine growth, gender and testosterone. Biol Neonate 75: 167‐176, 1999.
 97.Essig DA, Alderson NL, Ferguson MA, Bartoli WP, Durstine JL. Delayed effects of exercise on the plasma leptin concentration. Metab Clin Exp 49: 395‐399, 2000.
 98.Fajas L, Fruchart JC, Auwerx J. Transcriptional control of adipogenesis. Curr Opin Cell Biol 10: 165‐173, 1998.
 99.Famulla S, Horrighs A, Cramer A, Sell H, Eckel J. Hypoxia reduces the response of human adipocytes towards TNFalpha resulting in reduced NF‐kappaB signaling and MCP‐1 secretion. Int J Obes 36: 986‐992, 2012.
 100.Fang H, Judd RL. Adiponectin regulation and function. Compr Physiol 8: 1031‐1063, 2018.
 101.Feldman JL, Mitchell GS, Nattie EE. Breathing: Rhythmicity, plasticity, chemosensitivity. Annu Rev Neurosci 26: 239‐266, 2003.
 102.Finn PD, Cunningham MJ, Rickard DG, Clifton DK, Steiner RA. Serotonergic neurons are targets for leptin in the monkey. J Clin Endocrinol Metab 86: 422‐426, 2001.
 103.Flak JN, Arble D, Pan W, Patterson C, Lanigan T, Goforth PB, Sacksner J, Joosten M, Morgan DA, Allison MB, Hayes J, Feldman E, Seeley RJ, Olson DP, Rahmouni K, Myers MG Jr. A leptin‐regulated circuit controls glucose mobilization during noxious stimuli. J Clin Invest 127: 3103‐3113, 2017.
 104.Fletcher EC, Lesske J, Qian W, Miller CC 3rd, Unger T. Repetitive, episodic hypoxia causes diurnal elevation of blood pressure in rats. Hypertension 19: 555‐561, 1992.
 105.Fliedner S, Schulz C, Lehnert H. Brain uptake of intranasally applied radioiodinated leptin in Wistar rats. Endocrinology 147: 2088‐2094, 2006.
 106.Fortuna MG, Stornetta RL, West GH, Guyenet PG. Activation of the retrotrapezoid nucleus by posterior hypothalamic stimulation. J Physiol 587: 5121‐5138, 2009.
 107.Framnes SN, Arble DM. The bidirectional relationship between obstructive sleep apnea and metabolic disease. Front Endocrinol 9: 440, 2018.
 108.Frantz ID 3rd, Adler SM, Thach BT, Taeusch HW Jr. Maturational effects on respiratory responses to carbon dioxide in premature infants. J Appl Physiol 41: 41‐45, 1976.
 109.Frederiksen L, Hojlund K, Hougaard DM, Mosbech TH, Larsen R, Flyvbjerg A, Frystyk J, Brixen K, Andersen M. Testosterone therapy decreases subcutaneous fat and adiponectin in aging men. Eur J Endocrinol 166: 469‐476, 2012.
 110.Freet CS, Stoner JF, Tang X. Baroreflex and chemoreflex controls of sympathetic activity following intermittent hypoxia. Auton Neurosci Basic Clin 174: 8‐14, 2013.
 111.Fruhbeck G. Intracellular signalling pathways activated by leptin. Biochem J 393: 7‐20, 2006.
 112.Fujishima Y, Maeda N, Matsuda K, Masuda S, Mori T, Fukuda S, Sekimoto R, Yamaoka M, Obata Y, Kita S, Nishizawa H, Funahashi T, Ranscht B, Shimomura I. Adiponectin association with T‐cadherin protects against neointima proliferation and atherosclerosis. FASEB J 31: 1571‐1583, 2017.
 113.Fukushi I, Yokota S, Okada Y. The role of the hypothalamus in modulation of respiration. Respir Physiol Neurobiol 265: 172‐179, 2019.
 114.Furlong TM, McDowall LM, Horiuchi J, Polson JW, Dampney RA. The effect of air puff stress on c‐Fos expression in rat hypothalamus and brainstem: Central circuitry mediating sympathoexcitation and baroreflex resetting. Eur J Neurosci 39: 1429‐1438, 2014.
 115.Fuster JJ, Ouchi N, Gokce N, Walsh K. Obesity‐induced changes in adipose tissue microenvironment and their impact on cardiovascular disease. Circ Res 118: 1786‐1807, 2016.
 116.Gaines J, Vgontzas AN, Fernandez‐Mendoza J, Calhoun SL, He F, Liao D, Sawyer MD, Bixler EO. Inflammation mediates the association between visceral adiposity and obstructive sleep apnea in adolescents. Am J Phys Endocrinol Metab 311: E851‐E858, 2016.
 117.Gamber KM, Huo L, Ha S, Hairston JE, Greeley S, Bjorbaek C. Over‐expression of leptin receptors in hypothalamic POMC neurons increases susceptibility to diet‐induced obesity. PLoS One 7: e30485, 2012.
 118.Gao L, Ortega‐Saenz P, Garcia‐Fernandez M, Gonzalez‐Rodriguez P, Caballero‐Eraso C, Lopez‐Barneo J. Glucose sensing by carotid body glomus cells: Potential implications in disease. Front Physiol 5: 398, 2014.
 119.Garcia‐Rio F, Pino JM, Ramirez T, Alvaro D, Alonso A, Villasante C, Villamor J. Inspiratory neural drive response to hypoxia adequately estimates peripheral chemosensitivity in OSAHS patients. Eur Respir J 20: 724‐732, 2002.
 120.Gauda E, Martin R. Control of Breathing. In: Gleason C, Juul S, editors. Avery's Diseases of the Newborn. Philadelphia, PA: Elsevier, 2018, p. 600‐617.
 121.Gauda EB, Carroll JL, Donnelly DF. Developmental maturation of chemosensitivity to hypoxia of peripheral arterial chemoreceptors – invited article. Adv Exp Med Biol 648: 243‐255, 2009.
 122.Gauda EB, Master Z. Contribution of relative leptin and adiponectin deficiencies in premature infants to chronic intermittent hypoxia: Exploring a new hypothesis. Respir Physiol Neurobiol 256: 119‐127, 2018.
 123.Gauda EB, McLemore GL, Tolosa J, Marston‐Nelson J, Kwak D. Maturation of peripheral arterial chemoreceptors in relation to neonatal apnoea. Semin Neonatol 9: 181‐194, 2004.
 124.Gerosa‐Neto J, Panissa VLG, Monteiro PA, Inoue DS, Ribeiro JPJ, Figueiredo C, Zagatto AM, Little JP, Lira FS. High‐ or moderate‐intensity training promotes change in cardiorespiratory fitness, but not visceral fat, in obese men: A randomised trial of equal energy expenditure exercise. Respir Physiol Neurobiol 266: 150‐155, 2019.
 125.Glavas MM, Kirigiti MA, Xiao XQ, Enriori PJ, Fisher SK, Evans AE, Grayson BE, Cowley MA, Smith MS, Grove KL. Early overnutrition results in early‐onset arcuate leptin resistance and increased sensitivity to high‐fat diet. Endocrinology 151: 1598‐1610, 2010.
 126.Gletsu‐Miller N, Wright BN. Mineral malnutrition following bariatric surgery. Adv Nutr 4: 506‐517, 2013.
 127.Global BMIMC, Di Angelantonio E, Bhupathiraju Sh N, Wormser D, Gao P, Kaptoge S, Berrington de Gonzalez A, Cairns BJ, Huxley R, Jackson Ch L, Joshy G, Lewington S, Manson JE, Murphy N, Patel AV, Samet JM, Woodward M, Zheng W, Zhou M, Bansal N, Barricarte A, Carter B, Cerhan JR, Smith GD, Fang X, Franco OH, Green J, Halsey J, Hildebrand JS, Jung KJ, Korda RJ, McLerran DF, Moore SC, O'Keeffe LM, Paige E, Ramond A, Reeves GK, Rolland B, Sacerdote C, Sattar N, Sofianopoulou E, Stevens J, Thun M, Ueshima H, Yang L, Yun YD, Willeit P, Banks E, Beral V, Chen Z, Gapstur SM, Gunter MJ, Hartge P, Jee SH, Lam TH, Peto R, Potter JD, Willett WC, Thompson SG, Danesh J, Hu FB. Body‐mass index and all‐cause mortality: Individual‐participant‐data meta‐analysis of 239 prospective studies in four continents. Lancet 388: 776‐786, 2016.
 128.Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES, Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ, Huffman MD, Judd SE, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Mackey RH, Magid DJ, Marcus GM, Marelli A, Matchar DB, McGuire DK, Mohler ER 3rd, Moy CS, Mussolino ME, Neumar RW, Nichol G, Pandey DK, Paynter NP, Reeves MJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Wong ND, Woo D, Turner MB, American Heart Association Statistics Committee and Stroke Statistics Subcommittee. Heart disease and stroke statistics—2014 update: A report from the American Heart Association. Circulation 129: e28‐e292, 2014.
 129.Gonzalez C, Almaraz L, Obeso A, Rigual R. Oxygen and acid chemoreception in the carotid body chemoreceptors. Trends Neurosci 15: 146‐153, 1992.
 130.Gonzalez C, Almaraz L, Obeso A, Rigual R. Carotid body chemoreceptors: From natural stimuli to sensory discharges. Physiol Rev 74: 829‐898, 1994.
 131.Gonzalez‐Martin MC, Vega‐Agapito MV, Conde SV, Castaneda J, Bustamante R, Olea E, Perez‐Vizcaino F, Gonzalez C, Obeso A. Carotid body function and ventilatory responses in intermittent hypoxia. Evidence for anomalous brainstem integration of arterial chemoreceptor input. J Cell Physiol 226: 1961‐1969, 2011.
 132.Goossens GH, Bizzarri A, Venteclef N, Essers Y, Cleutjens JP, Konings E, Jocken JW, Cajlakovic M, Ribitsch V, Clement K, Blaak EE. Increased adipose tissue oxygen tension in obese compared with lean men is accompanied by insulin resistance, impaired adipose tissue capillarization, and inflammation. Circulation 124: 67‐76, 2011.
 133.Gorska E, Popko K, Stelmaszczyk‐Emmel A, Ciepiela O, Kucharska A, Wasik M. Leptin receptors. Eur J Med Res 15 (Suppl 2): 50‐54, 2010.
 134.Grattan DR, Ladyman SR, Augustine RA. Hormonal induction of leptin resistance during pregnancy. Physiol Behav 91: 366‐374, 2007.
 135.Grill HJ, Schwartz MW, Kaplan JM, Foxhall JS, Breininger J, Baskin DG. Evidence that the caudal brainstem is a target for the inhibitory effect of leptin on food intake. Endocrinology 143: 239‐246, 2002.
 136.Groeben H, Meier S, Brown RH, O'Donnell CP, Mitzner W, Tankersley CG. The effect of leptin on the ventilatory responseto hyperoxia. Exp Lung Res 30: 559‐570, 2004.
 137.Grove KL, Grayson BE, Glavas MM, Xiao XQ, Smith MS. Development of metabolic systems. Physiol Behav 86: 646‐660, 2005.
 138.Guertzenstein PG, Silver A. Fall in blood pressure produced from discrete regions of the ventral surface of the medulla by glycine and lesions. J Physiol 242: 489‐503, 1974.
 139.Guilleminault C, Quo SD. Sleep‐disordered breathing. A view at the beginning of the new Millennium. Dent Clin N Am 45: 643‐656, 2001.
 140.Guimaraes KSL, de Araujo EV, Aquino JS, Gadelha DA, Balarini CM, Costa‐Silva JH, Magnani M, Vidal H, Braga VA, de Brito Alves JL. Effect of maternal dyslipidaemia on the cardiorespiratory physiology and biochemical parameters in male rat offspring. Br J Nutr 118: 930‐941, 2017.
 141.Guo R, Zhang Y, Turdi S, Ren J. Adiponectin knockout accentuates high fat diet‐induced obesity and cardiac dysfunction: Role of autophagy. Biochim Biophys Acta 1832: 1136‐1148, 2013.
 142.Guyenet PG. Regulation of breathing and autonomic outflows by chemoreceptors. Compr Physiol 4: 1511‐1562, 2014.
 143.Ha S, Baver S, Huo L, Gata A, Hairston J, Huntoon N, Li W, Zhang T, Benecchi EJ, Ericsson M, Hentges ST, Bjorbaek C. Somato‐dendritic localization and signaling by leptin receptors in hypothalamic POMC and AgRP neurons. PLoS One 8: e77622, 2013.
 144.Hall JE, da Silva AA, do Carmo JM, Dubinion J, Hamza S, Munusamy S, Smith G, Stec DE. Obesity‐induced hypertension: Role of sympathetic nervous system, leptin, and melanocortins. J Biol Chem 285: 17271‐17276, 2010.
 145.Hansen CS, Vistisen D, Jorgensen ME, Witte DR, Brunner EJ, Tabak AG, Kivimaki M, Roden M, Malik M, Herder C. Adiponectin, biomarkers of inflammation and changes in cardiac autonomic function: Whitehall II study. Cardiovasc Diabetol 16: 153, 2017.
 146.Harsch IA, Konturek PC, Koebnick C, Kuehnlein PP, Fuchs FS, Pour Schahin S, Wiest GH, Hahn EG, Lohmann T, Ficker JH. Leptin and ghrelin levels in patients with obstructive sleep apnoea: Effect of CPAP treatment. Eur Respir J 22: 251‐257, 2003.
 147.Haselton JR, Guyenet PG. Central respiratory modulation of medullary sympathoexcitatory neurons in rat. Am J Phys 256: R739‐R750, 1989.
 148.Hauck FR, Thompson JM, Tanabe KO, Moon RY, Vennemann MM. Breastfeeding and reduced risk of sudden infant death syndrome: A meta‐analysis. Pediatrics 128: 103‐110, 2011.
 149.Haynes WG. Interaction between leptin and sympathetic nervous system in hypertension. Curr Hypertens Rep 2: 311‐318, 2000.
 150.Hellgren G, Engstrom E, Smith LE, Lofqvist C, Hellstrom A. Effect of preterm birth on postnatal apolipoprotein and adipocytokine profiles. Neonatology 108: 16‐22, 2015.
 151.Henry RR, Wallace P, Olefsky JM. Effects of weight loss on mechanisms of hyperglycemia in obese non‐insulin‐dependent diabetes mellitus. Diabetes 35: 990‐998, 1986.
 152.Hilaire G, Viemari JC, Coulon P, Simonneau M, Bevengut M. Modulation of the respiratory rhythm generator by the pontine noradrenergic A5 and A6 groups in rodents. Respir Physiol Neurobiol 143: 187‐197, 2004.
 153.Hitzig BM, Perng WC, Burt T, Okunieff P, Johnson DC. 1H‐NMR measurement of fractional dissociation of imidazole in intact animals. Am J Phys 266: R1008‐R1015, 1994.
 154.Hodson L, Humphreys SM, Karpe F, Frayn KN. Metabolic signatures of human adipose tissue hypoxia in obesity. Diabetes 62: 1417‐1425, 2013.
 155.Hoyda TD, Smith PM, Ferguson AV. Adiponectin acts in the nucleus of the solitary tract to decrease blood pressure by modulating the excitability of neuropeptide Y neurons. Brain Res 1256: 76‐84, 2009.
 156.Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose‐specific gene dysregulated in obesity. J Biol Chem 271: 10697‐10703, 1996.
 157.Huang XF, Han M, South T, Storlien L. Altered levels of POMC, AgRP and MC4‐R mRNA expression in the hypothalamus and other parts of the limbic system of mice prone or resistant to chronic high‐energy diet‐induced obesity. Brain Res 992: 9‐19, 2003.
 158.Hummel KP, Dickie MM, Coleman DL. Diabetes, a new mutation in the mouse. Science 153: 1127‐1128, 1966.
 159.Hutter MM, Schirmer BD, Jones DB, Ko CY, Cohen ME, Merkow RP, Nguyen NT. First report from the American College of Surgeons Bariatric Surgery Center Network: Laparoscopic sleeve gastrectomy has morbidity and effectiveness positioned between the band and the bypass. Ann Surg 254: 410‐420; discussion 420‐412, 2011.
 160.Iftikhar IH, Valentine CW, Bittencourt LR, Cohen DL, Fedson AC, Gislason T, Penzel T, Phillips CL, Yu‐sheng L, Pack AI, Magalang UJ. Effects of continuous positive airway pressure on blood pressure in patients with resistant hypertension and obstructive sleep apnea: A meta‐analysis. J Hypertens 32: 2341‐2350; discussion 2350, 2014.
 161.Ingalls AM, Dickie MM, Snell GD. Obese, a new mutation in the house mouse. J Hered 41: 317‐318, 1950.
 162.Inoue DS, Panissa VL, Antunes BM, Oliveira FP, Malta RB, Caldeira RS, Campos EZ, Pimentel GD, Franchini E, Lira FS. Reduced leptin level is independent of fat mass changes and hunger scores from high‐intensity intermittent plus strength training. J Sport Med Phys Fit 58: 1045‐1051, 2018.
 163.Inyushkin AN, Inyushkina EM, Merkulova NA. Respiratory responses to microinjections of leptin into the solitary tract nucleus. Neurosci Behav Physiol 39: 231‐240, 2009.
 164.Inyushkina EM, Merkulova NA, Inyushkin AN. Mechanisms of the respiratory activity of leptin at the level of the solitary tract nucleus. Neurosci Behav Physiol 40: 707‐713, 2010.
 165.Ip MS, Lam KS, Ho C, Tsang KW, Lam W. Serum leptin and vascular risk factors in obstructive sleep apnea. Chest 118: 580‐586, 2000.
 166.Iturriaga R, Oyarce MP, Dias ACR. Role of carotid body in intermittent hypoxia‐related hypertension. Curr Hypertens Rep 19: 38, 2017.
 167.Janczewski WA, Feldman JL. Distinct rhythm generators for inspiration and expiration in the juvenile rat. J Physiol 570: 407‐420, 2006.
 168.Jaquet D, Leger J, Levy‐Marchal C, Oury JF, Czernichow P. Ontogeny of leptin in human fetuses and newborns: Effect of intrauterine growth retardation on serum leptin concentrations. J Clin Endocrinol Metab 83: 1243‐1246, 1998.
 169.Jin G, Wang W, Kang J, Wu Y, Hou X, Yu R. Study of serum leptin level in patients with obstructive sleep apnea. Zhonghua jie he he hu xi za zhi 25: 204‐206, 2002.
 170.Jordan AS, McSharry DG, Malhotra A. Adult obstructive sleep apnoea. Lancet 383: 736‐747, 2014.
 171.Kaar JL, Brinton JT, Crume T, Hamman RF, Glueck DH, Dabelea D. Leptin levels at birth and infant growth: The EPOCH study. J Dev Orig Health Dis 5: 214‐218, 2014.
 172.Karvonen MK, Pesonen U, Heinonen P, Laakso M, Rissanen A, Naukkarinen H, Valve R, Uusitupa MI, Koulu M. Identification of new sequence variants in the leptin gene. J Clin Endocrinol Metab 83: 3239‐3242, 1998.
 173.Kelesidis I, Mantzoros CS. Leptin and its emerging role in children and adolescents. Clin Pediatr Endocrinol 15: 1‐14, 2006.
 174.Kesavan K, Devaskar SU. Intrauterine growth restriction: Postnatal monitoring and outcomes. Pediatr Clin N Am 66: 403‐423, 2019.
 175.Kim DK, Summers BA, Prabhakar NR, Kumar GK. Hypoxia does not uniformly facilitate the release of multiple transmitters from the carotid body. Adv Exp Med Biol 536: 291‐296, 2003.
 176.Kim JY, van de Wall E, Laplante M, Azzara A, Trujillo ME, Hofmann SM, Schraw T, Durand JL, Li H, Li G, Jelicks LA, Mehler MF, Hui DY, Deshaies Y, Shulman GI, Schwartz GJ, Scherer PE. Obesity‐associated improvements in metabolic profile through expansion of adipose tissue. J Clin Invest 117: 2621‐2637, 2007.
 177.Kim S, Whelan J, Claycombe K, Reath DB, Moustaid‐Moussa N. Angiotensin II increases leptin secretion by 3T3‐L1 and human adipocytes via a prostaglandin‐independent mechanism. J Nutr 132: 1135‐1140, 2002.
 178.Kirk SL, Samuelsson AM, Argenton M, Dhonye H, Kalamatianos T, Poston L, Taylor PD, Coen CW. Maternal obesity induced by diet in rats permanently influences central processes regulating food intake in offspring. PLoS One 4: e5870, 2009.
 179.Kishi T, Aschkenasi CJ, Lee CE, Mountjoy KG, Saper CB, Elmquist JK. Expression of melanocortin 4 receptor mRNA in the central nervous system of the rat. J Comp Neurol 457: 213‐235, 2003.
 180.Kokkinos P. Cardiorespiratory fitness, exercise, and blood pressure. Hypertension 64: 1160‐1164, 2014.
 181.Kotsis V, Stabouli S, Papakatsika S, Rizos Z, Parati G. Mechanisms of obesity‐induced hypertension. Hypertens Res 33: 386‐393, 2010.
 182.Koyama Y, Coker RH, Denny JC, Lacy DB, Jabbour K, Williams PE, Wasserman DH. Role of carotid bodies in control of the neuroendocrine response to exercise. Am J Phys Endocrinol Metab 281: E742‐E748, 2001.
 183.Koyama Y, Coker RH, Stone EE, Lacy DB, Jabbour K, Williams PE, Wasserman DH. Evidence that carotid bodies play an important role in glucoregulation in vivo. Diabetes 49: 1434‐1442, 2000.
 184.Kugananthan S, Lai CT, Gridneva Z, Mark PJ, Geddes DT, Kakulas F. Leptin levels are higher in whole compared to skim human milk, supporting a cellular contribution. Nutrients 8: 711, 2016.
 185.Kumar P, Prabhakar NR. Peripheral chemoreceptors: Function and plasticity of the carotid body. Compr Physiol 2: 141‐219, 2012.
 186.Kuwaki T. Orexinergic modulation of breathing across vigilance states. Respir Physiol Neurobiol 164: 204‐212, 2008.
 187.Kwak DJ, Kwak SD, Gauda EB. The effect of hyperoxia on reactive oxygen species (ROS) in rat petrosal ganglion neurons during development using organotypic slices. Pediatr Res 60: 371‐376, 2006.
 188.Lage M, Baldelli R, Camina JP, Rodriguez‐Garci J, Penalva A, Dieguez C, Casanueva FF. Presence of bovine leptin in edible commercial milk and infant formula. J Endocrinol Investig 25: 670‐674, 2002.
 189.Lahiri S, DeLaney RG. Relationship between carotid chemoreceptor activity and ventilation in the cat. Respir Physiol 24: 267‐286, 1975.
 190.Lambert E, Straznicky NE, Dawood T, Ika‐Sari C, Grima M, Esler MD, Schlaich MP, Lambert GW. Change in sympathetic nerve firing pattern associated with dietary weight loss in the metabolic syndrome. Front Physiol 2: 52, 2011.
 191.Lambert EA, Straznicky NE, Dixon JB, Lambert GW. Should the sympathetic nervous system be a target to improve cardiometabolic risk in obesity? Am J Phys Heart Circ Phys 309: H244‐H258, 2015.
 192.Lambert GW, Straznicky NE, Lambert EA, Dixon JB, Schlaich MP. Sympathetic nervous activation in obesity and the metabolic syndrome‐causes, consequences and therapeutic implications. Pharmacol Ther 126: 159‐172, 2010.
 193.Landsberg L. Insulin resistance and the metabolic syndrome. Diabetologia 48: 1244‐1246, 2005.
 194.Landt M, Lawson GM, Helgeson JM, Davila‐Roman VG, Ladenson JH, Jaffe AS, Hickner RC. Prolonged exercise decreases serum leptin concentrations. Metab Clin Exp 46: 1109‐1112, 1997.
 195.Lecke SB, Morsch DM, Spritzer PM. Leptin and adiponectin in the female life course. Braz J Med Biol Res 44: 381‐387, 2011.
 196.Leite RD, Durigan Rde C, de Souza Lino AD, de Souza Campos MV, Souza M, Selistre‐de‐Araujo HS, Bouskela E, Kraemer‐Aguiar LG. Resistance training may concomitantly benefit body composition, blood pressure and muscle MMP‐2 activity on the left ventricle of high‐fat fed diet rats. Metab Clin Exp 62: 1477‐1484, 2013.
 197.Levitzky M. Control of Breathing. In: Michael G. Levitzky, editor. Pulmonary Physiology; [traduction of the Marcos Ikeda]. 6th ed. Manole, Barueri: Sao Paulo, 2004.
 198.Li A, Nattie E. CO2 dialysis in one chemoreceptor site, the RTN: Stimulus intensity and sensitivity in the awake rat. Respir Physiol Neurobiol 133: 11‐22, 2002.
 199.Li A, Nattie E. Serotonin transporter knockout mice have a reduced ventilatory response to hypercapnia (predominantly in males) but not to hypoxia. J Physiol 586: 2321‐2329, 2008.
 200.Li P, Cui BP, Zhang LL, Sun HJ, Liu TY, Zhu GQ. Melanocortin 3/4 receptors in paraventricular nucleus modulate sympathetic outflow and blood pressure. Exp Physiol 98: 435‐443, 2013.
 201.Li P, Shibata R, Unno K, Shimano M, Furukawa M, Ohashi T, Cheng X, Nagata K, Ouchi N, Murohara T. Evidence for the importance of adiponectin in the cardioprotective effects of pioglitazone. Hypertension 55: 69‐75, 2010.
 202.Li YL, Schultz HD. Enhanced sensitivity of Kv channels to hypoxia in the rabbit carotid body in heart failure: Role of angiotensin II. J Physiol 575: 215‐227, 2006.
 203.Li YL, Xia XH, Zheng H, Gao L, Li YF, Liu D, Patel KP, Wang W, Schultz HD. Angiotensin II enhances carotid body chemoreflex control of sympathetic outflow in chronic heart failure rabbits. Cardiovasc Res 71: 129‐138, 2006.
 204.Lin M, Liu R, Gozal D, Wead WB, Chapleau MW, Wurster R, Cheng ZJ. Chronic intermittent hypoxia impairs baroreflex control of heart rate but enhances heart rate responses to vagal efferent stimulation in anesthetized mice. Am J Phys Heart Circ Phys 293: H997‐H1006, 2007.
 205.Liu M, Liu F. Transcriptional and post‐translational regulation of adiponectin. Biochem J 425: 41‐52, 2009.
 206.Liu Y, Chewchuk S, Lavigne C, Brule S, Pilon G, Houde V, Xu A, Marette A, Sweeney G. Functional significance of skeletal muscle adiponectin production, changes in animal models of obesity and diabetes, and regulation by rosiglitazone treatment. Am J Phys Endocrinol Metab 297: E657‐E664, 2009.
 207.Loredo JS, Clausen JL, Nelesen RA, Ancoli‐Israel S, Ziegler MG, Dimsdale JE. Obstructive sleep apnea and hypertension: Are peripheral chemoreceptors involved? Med Hypotheses 56: 17‐19, 2001.
 208.Lu M, Fang F, Wang Z, Wei P, Hu C, Wei Y. Association between serum/plasma levels of adiponectin and obstructive sleep apnea hypopnea syndrome: A meta‐analysis. Lipids Health Dis 18: 30, 2019.
 209.MacFarlane PM, Vinit S, Mitchell GS. Enhancement of phrenic long‐term facilitation following repetitive acute intermittent hypoxia is blocked by the glycolytic inhibitor 2‐deoxyglucose. Am J Physiol Regul Integr Comp Physiol 314: R135‐R144, 2018.
 210.Mack SO, Kc P, Wu M, Coleman BR, Tolentino‐Silva FP, Haxhiu MA. Paraventricular oxytocin neurons are involved in neural modulation of breathing. J Appl Physiol 92: 826‐834, 2002.
 211.Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y, Matsubara K. cDNA cloning and expression of a novel adipose specific collagen‐like factor, apM1 (adipose most abundant gene transcript 1). 1996. Biochem Biophys Res Commun 425: 556‐559, 2012.
 212.Makinodan K, Yoshikawa M, Fukuoka A, Tamaki S, Koyama N, Yamauchi M, Tomoda K, Hamada K, Kimura H. Effect of serum leptin levels on hypercapnic ventilatory response in obstructive sleep apnea. Respiration 75: 257‐264, 2008.
 213.Maki‐Nunes C, Toschi‐Dias E, Cepeda FX, Rondon MU, Alves MJ, Fraga RF, Braga AM, Aguilar AM, Amaro AC, Drager LF, Lorenzi‐Filho G, Negrao CE, Trombetta IC. Diet and exercise improve chemoreflex sensitivity in patients with metabolic syndrome and obstructive sleep apnea. Obesity 23: 1582‐1590, 2015.
 214.Malli F, Papaioannou AI, Gourgoulianis KI, Daniil Z. The role of leptin in the respiratory system: An overview. Respir Res 11: 152, 2010.
 215.Mansukhani MP, Kara T, Caples SM, Somers VK. Chemoreflexes, sleep apnea, and sympathetic dysregulation. Curr Hypertens Rep 16: 476, 2014.
 216.Marchenko V, Koizumi H, Mosher B, Koshiya N, Tariq MF, Bezdudnaya TG, Zhang R, Molkov YI, Rybak IA, Smith JC. Perturbations of respiratory rhythm and pattern by disrupting synaptic inhibition within pre‐Botzinger and Botzinger complexes. eNeuro 3: ENEURO.0011‐16.2016, 2016.
 217.Marin P, Rebuffe‐Scrive M, Smith U, Bjorntorp P. Glucose uptake in human adipose tissue. Metab Clin Exp 36: 1154‐1160, 1987.
 218.Marjanovic M, Elliott AC, Dawson MJ. The temperature dependence of intracellular pH in isolated frog skeletal muscle: Lessons concerning the Na(+)‐H+ exchanger. J Membr Biol 161: 215‐225, 1998.
 219.Mark AL, Agassandian K, Morgan DA, Liu X, Cassell MD, Rahmouni K. Leptin signaling in the nucleus tractus solitarii increases sympathetic nerve activity to the kidney. Hypertension 53: 375‐380, 2009.
 220.Marshall JM. Peripheral chemoreceptors and cardiovascular regulation. Physiol Rev 74: 543‐594, 1994.
 221.Martos‐Moreno GA, Barrios V, Argente J. Normative data for adiponectin, resistin, interleukin 6, and leptin/receptor ratio in a healthy Spanish pediatric population: Relationship with sex steroids. Eur J Endocrinol 155: 429‐434, 2006.
 222.Matsuda K, Fujishima Y, Maeda N, Mori T, Hirata A, Sekimoto R, Tsushima Y, Masuda S, Yamaoka M, Inoue K, Nishizawa H, Kita S, Ranscht B, Funahashi T, Shimomura I. Positive feedback regulation between adiponectin and T‐cadherin impacts adiponectin levels in tissue and plasma of male mice. Endocrinology 156: 934‐946, 2015.
 223.McAllen RM. Central respiratory modulation of subretrofacial bulbospinal neurones in the cat. J Physiol 388: 533‐545, 1987.
 224.McDowall LM, Horiuchi J, Dampney RA. Effects of disinhibition of neurons in the dorsomedial hypothalamus on central respiratory drive. Am J Physiol Regul Integr Comp Physiol 293: R1728‐R1735, 2007.
 225.McEvoy RD, Antic NA, Heeley E, Luo Y, Ou Q, Zhang X, Mediano O, Chen R, Drager LF, Liu Z, Chen G, Du B, McArdle N, Mukherjee S, Tripathi M, Billot L, Li Q, Lorenzi‐Filho G, Barbe F, Redline S, Wang J, Arima H, Neal B, White DP, Grunstein RR, Zhong N, Anderson CS, SAVE Investigators and Coordinators. CPAP for prevention of cardiovascular events in obstructive sleep apnea. N Engl J Med 375: 919‐931, 2016.
 226.Mesarwi OA, Sharma EV, Jun JC, Polotsky VY. Metabolic dysfunction in obstructive sleep apnea: A critical examination of underlying mechanisms. Sleep Biol Rhythms 13: 2‐17, 2015.
 227.Messenger SA, Ciriello J. Effects of intermittent hypoxia on leptin signalling in the carotid body. Neuroscience 232: 216‐225, 2013.
 228.Messenger SA, Moreau JM, Ciriello J. Intermittent hypoxia and systemic leptin administration induces pSTAT3 and Fos/Fra‐1 in the carotid body. Brain Res 1446: 56‐70, 2012.
 229.Messenger SA, Moreau JM, Ciriello J. Effect of chronic intermittent hypoxia on leptin and leptin receptor protein expression in the carotid body. Brain Res 1513: 51‐60, 2013.
 230.Mistry AM, Swick A, Romsos DR. Leptin alters metabolic rates before acquisition of its anorectic effect in developing neonatal mice. Am J Phys 277: R742‐R747, 1999.
 231.Miyawaki T, Pilowsky P, Sun QJ, Minson J, Suzuki S, Arnolda L, Llewellyn‐Smith I, Chalmers J. Central inspiration increases barosensitivity of neurons in rat rostral ventrolateral medulla. Am J Phys 268: R909‐R918, 1995.
 232.Mizuno TM, Kleopoulos SP, Bergen HT, Roberts JL, Priest CA, Mobbs CV. Hypothalamic pro‐opiomelanocortin mRNA is reduced by fasting and [corrected] in ob/ob and db/db mice, but is stimulated by leptin. Diabetes 47: 294‐297, 1998.
 233.Mizuno TM, Makimura H, Silverstein J, Roberts JL, Lopingco T, Mobbs CV. Fasting regulates hypothalamic neuropeptide Y, agouti‐related peptide, and proopiomelanocortin in diabetic mice independent of changes in leptin or insulin. Endocrinology 140: 4551‐4557, 1999.
 234.Mizuno TM, Mobbs CV. Hypothalamic agouti‐related protein messenger ribonucleic acid is inhibited by leptin and stimulated by fasting. Endocrinology 140: 814‐817, 1999.
 235.Mohr MA, Fairchild KD, Patel M, Sinkin RA, Clark MT, Moorman JR, Lake DE, Kattwinkel J, Delos JB. Quantification of periodic breathing in premature infants. Physiol Meas 36: 1415‐1427, 2015.
 236.Moller DE. Potential role of TNF‐alpha in the pathogenesis of insulin resistance and type 2 diabetes. Trends Endocrinol Metab 11: 212‐217, 2000.
 237.Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, Earley AR, Barnett AH, Prins JB, O'Rahilly S. Congenital leptin deficiency is associated with severe early‐onset obesity in humans. Nature 387: 903‐908, 1997.
 238.Montesi SB, Edwards BA, Malhotra A, Bakker JP. The effect of continuous positive airway pressure treatment on blood pressure: A systematic review and meta‐analysis of randomized controlled trials. J Clin Sleep Med 8: 587‐596, 2012.
 239.Moraes DJ, Zoccal DB, Machado BH. Medullary respiratory network drives sympathetic overactivity and hypertension in rats submitted to chronic intermittent hypoxia. Hypertension 60: 1374‐1380, 2012.
 240.Moreau JM, Ciriello J. Effects of acute intermittent hypoxia on energy balance and hypothalamic feeding pathways. Neuroscience 253: 350‐360, 2013.
 241.Moreau JM, Messenger SA, Ciriello J. Effects of angiotensin II on leptin and downstream leptin signaling in the carotid body during acute intermittent hypoxia. Neuroscience 310: 430‐441, 2015.
 242.Moreira TS, Takakura AC, Colombari E, Guyenet PG. Central chemoreceptors and sympathetic vasomotor outflow. J Physiol 577: 369‐386, 2006.
 243.Morton GJ, Schwartz MW. Leptin and the central nervous system control of glucose metabolism. Physiol Rev 91: 389‐411, 2011.
 244.Munzberg H, Morrison CD. Structure, production and signaling of leptin. Metab Clin Exp 64: 13‐23, 2015.
 245.Murphy AM, Thomas A, Crinion SJ, Kent BD, Tambuwala MM, Fabre A, Pepin JL, Roche HM, Arnaud C, Ryan S. Intermittent hypoxia in obstructive sleep apnoea mediates insulin resistance through adipose tissue inflammation. Eur Respir J 49 (4): 1601731, 2017.
 246.Myers MG, Cowley MA, Munzberg H. Mechanisms of leptin action and leptin resistance. Annu Rev Physiol 70: 537‐556, 2008.
 247.Nakano Y, Tobe T, Choi‐Miura NH, Mazda T, Tomita M. Isolation and characterization of GBP28, a novel gelatin‐binding protein purified from human plasma. J Biochem 120: 803‐812, 1996.
 248.Nambu T, Sakurai T, Mizukami K, Hosoya Y, Yanagisawa M, Goto K. Distribution of orexin neurons in the adult rat brain. Brain Res 827: 243‐260, 1999.
 249.Narkiewicz K, Somers VK. Cardiovascular variability characteristics in obstructive sleep apnea. Auton Neurosci Basic Clin 90: 89‐94, 2001.
 250.Narkiewicz K, van de Borne PJ, Pesek CA, Dyken ME, Montano N, Somers VK. Selective potentiation of peripheral chemoreflex sensitivity in obstructive sleep apnea. Circulation 99: 1183‐1189, 1999.
 251.Nattie E. CO2, brainstem chemoreceptors and breathing. Prog Neurobiol 59: 299‐331, 1999.
 252.Nattie E, Li A. Respiration and autonomic regulation and orexin. Prog Brain Res 198: 25‐46, 2012.
 253.Nattie EE, Li A. Retrotrapezoid nucleus lesions decrease phrenic activity and CO2 sensitivity in rats. Respir Physiol 97: 63‐77, 1994.
 254.Nattie EE, Li A. Central chemoreception in the region of the ventral respiratory group in the rat. J Appl Physiol 81: 1987‐1995, 1996.
 255.Newhouse LP, Joyner MJ, Curry TB, Laurenti MC, Man CD, Cobelli C, Vella A, Limberg JK. Three hours of intermittent hypoxia increases circulating glucose levels in healthy adults. Phys Rep 5 (1): e13106, 2017.
 256.Newton AJ, Hess S, Paeger L, Vogt MC, Fleming Lascano J, Nillni EA, Bruning JC, Kloppenburg P, Xu AW. AgRP innervation onto POMC neurons increases with age and is accelerated with chronic high‐fat feeding in male mice. Endocrinology 154: 172‐183, 2013.
 257.Nindl BC, Kraemer WJ, Arciero PJ, Samatallee N, Leone CD, Mayo MF, Hafeman DL. Leptin concentrations experience a delayed reduction after resistance exercise in men. Med Sci Sports Exerc 34: 608‐613, 2002.
 258.Nunes M, da Silva CH, Bosa VL, Bernardi JR, Werlang ICR, Goldani MZ, NESCA Group. Could a remarkable decrease in leptin and insulin levels from colostrum to mature milk contribute to early growth catch‐up of SGA infants? BMC Pregnancy Childbirth 17: 410, 2017.
 259.Nurse CA, Leonard EM, Salman S. Role of glial‐like type II cells as paracrine modulators of carotid body chemoreception. Physiol Genomics 50: 255‐262, 2018.
 260.O'Donnell CP, Schaub CD, Haines AS, Berkowitz DE, Tankersley CG, Schwartz AR, Smith PL. Leptin prevents respiratory depression in obesity. Am J Respir Crit Care Med 159: 1477‐1484, 1999.
 261.Ogden CL, Carroll MD, Fryar CD, Flegal KM. Prevalence of obesity among adults and youth: United States, 2011‐2014. NCHS data brief, no 219. Hyattsville, MD: National Center for Health Statistics, 2015.
 262.Olea E, Ribeiro MJ, Gallego‐Martin T, Yubero S, Rigual R, Masa JF, Obeso A, Conde SV, Gonzalez C. The carotid body does not mediate the acute ventilatory effects of leptin. Adv Exp Med Biol 860: 379‐385, 2015.
 263.Olive JL, Miller GD. Differential effects of maximal‐ and moderate‐intensity runs on plasma leptin in healthy trained subjects. Nutrition 17: 365‐369, 2001.
 264.Oyamada Y, Ballantyne D, Muckenhoff K, Scheid P. Respiration‐modulated membrane potential and chemosensitivity of locus coeruleus neurones in the in vitro brainstem‐spinal cord of the neonatal rat. J Physiol 513 (Pt 2): 381‐398, 1998.
 265.Ozturk L, Unal M, Tamer L, Celikoglu F. The association of the severity of obstructive sleep apnea with plasma leptin levels. Arch Otolaryngol Head Neck Surg 129: 538‐540, 2003.
 266.Pan WW, Myers MG Jr. Leptin and the maintenance of elevated body weight. Nat Rev Neurosci 19: 95‐105, 2018.
 267.Papp RS, Palkovits M. Brainstem projections of neurons located in various subdivisions of the dorsolateral hypothalamic area‐an anterograde tract‐tracing study. Front Neuroanat 8: 34, 2014.
 268.Parameswaran K, Todd DC, Soth M. Altered respiratory physiology in obesity. Can Respir J 13: 203‐210, 2006.
 269.Pardal R, Lopez‐Barneo J. Low glucose‐sensing cells in the carotid body. Nat Neurosci 5: 197‐198, 2002.
 270.Pardal R, Ortega‐Saenz P, Duran R, Lopez‐Barneo J. Glia‐like stem cells sustain physiologic neurogenesis in the adult mammalian carotid body. Cell 131: 364‐377, 2007.
 271.Pasarica M, Sereda OR, Redman LM, Albarado DC, Hymel DT, Roan LE, Rood JC, Burk DH, Smith SR. Reduced adipose tissue oxygenation in human obesity: Evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes 58: 718‐725, 2009.
 272.Patel SR, Palmer LJ, Larkin EK, Jenny NS, White DP, Redline S. Relationship between obstructive sleep apnea and diurnal leptin rhythms. Sleep 27: 235‐239, 2004.
 273.Patel SR, White DP, Malhotra A, Stanchina ML, Ayas NT. Continuous positive airway pressure therapy for treating sleepiness in a diverse population with obstructive sleep apnea: Results of a meta‐analysis. Arch Intern Med 163: 565‐571, 2003.
 274.Paz‐Filho G, Mastronardi C, Delibasi T, Wong ML, Licinio J. Congenital leptin deficiency: Diagnosis and effects of leptin replacement therapy. Arquivos brasileiros de endocrinologia e metabologia 54: 690‐697, 2010.
 275.Peng YJ, Overholt JL, Kline D, Kumar GK, Prabhakar NR. Induction of sensory long‐term facilitation in the carotid body by intermittent hypoxia: Implications for recurrent apneas. Proc Natl Acad Sci U S A 100: 10073‐10078, 2003.
 276.Peng YJ, Prabhakar NR. Effect of two paradigms of chronic intermittent hypoxia on carotid body sensory activity. J Appl Physiol 96: 1236‐1242; discussion 1196, 2004.
 277.Penumarti A, Abdel‐Rahman AA. Neuronal nitric oxide synthase‐dependent elevation in adiponectin in the rostral ventrolateral medulla underlies g protein‐coupled receptor 18‐mediated hypotension in conscious rats. J Pharmacol Exp Ther 351: 44‐53, 2014.
 278.Peppard PE, Young T, Palta M, Dempsey J, Skatrud J. Longitudinal study of moderate weight change and sleep‐disordered breathing. JAMA 284: 3015‐3021, 2000.
 279.Peyron C, Tighe DK, van den Pol AN, de Lecea L, Heller HC, Sutcliffe JG, Kilduff TS. Neurons containing hypocretin (orexin) project to multiple neuronal systems. J Neurosci Off J Soc Neurosci 18: 9996‐10015, 1998.
 280.Phillips BG, Kato M, Narkiewicz K, Choe I, Somers VK. Increases in leptin levels, sympathetic drive, and weight gain in obstructive sleep apnea. Am J Phys Heart Circ Phys 279: H234‐H237, 2000.
 281.Phillips SA, Ciaraldi TP, Oh DK, Savu MK, Henry RR. Adiponectin secretion and response to pioglitazone is depot dependent in cultured human adipose tissue. Am J Phys Endocrinol Metab 295: E842‐E850, 2008.
 282.Pierard M, Tassin A, Conotte S, Zouaoui Boudjeltia K, Legrand A. Sustained intermittent hypoxemia induces adiponectin oligomers redistribution and a tissue‐specific modulation of adiponectin receptor in mice. Front Physiol 10: 68, 2019.
 283.Pinheiro AR, Cunha AR, Aguila MB, Mandarim‐de‐Lacerda CA. Beneficial effects of physical exercise on hypertension and cardiovascular adverse remodeling of diet‐induced obese rats. Nutr Metab Cardiovasc Dis 17: 365‐375, 2007.
 284.Piskuric NA, Nurse CA. Expanding role of ATP as a versatile messenger at carotid and aortic body chemoreceptors. J Physiol 591: 415‐422, 2013.
 285.Platero‐Luengo A, Gonzalez‐Granero S, Duran R, Diaz‐Castro B, Piruat JI, Garcia‐Verdugo JM, Pardal R, Lopez‐Barneo J. An O2‐sensitive glomus cell‐stem cell synapse induces carotid body growth in chronic hypoxia. Cell 156: 291‐303, 2014.
 286.Polak J, Shimoda LA, Drager LF, Undem C, McHugh H, Polotsky VY, Punjabi NM. Intermittent hypoxia impairs glucose homeostasis in C57BL6/J mice: Partial improvement with cessation of the exposure. Sleep 36: 1483‐1490; 1490A‐1490B, 2013.
 287.Polotsky VY, Wilson JA, Smaldone MC, Haines AS, Hurn PD, Tankersley CG, Smith PL, Schwartz AR, O'Donnell CP. Female gender exacerbates respiratory depression in leptin‐deficient obesity. Am J Respir Crit Care Med 164: 1470‐1475, 2001.
 288.Porzionato A, Rucinski M, Macchi V, Stecco C, Castagliuolo I, Malendowicz LK, De Caro R. Expression of leptin and leptin receptor isoforms in the rat and human carotid body. Brain Res 1385: 56‐67, 2011.
 289.Prabhakar NR, Dick TE, Nanduri J, Kumar GK. Systemic, cellular and molecular analysis of chemoreflex‐mediated sympathoexcitation by chronic intermittent hypoxia. Exp Physiol 92: 39‐44, 2007.
 290.Prabhakar NR, Peng YJ, Kumar GK, Pawar A. Altered carotid body function by intermittent hypoxia in neonates and adults: Relevance to recurrent apneas. Respir Physiol Neurobiol 157: 148‐153, 2007.
 291.Pye RL, Dunn EJ, Ricker EM, Jurcsisn JG, Barr BL, Wyatt CN. Acutely administered leptin increases [Ca2+] I and BK Ca currents but does not alter chemosensory behavior in rat carotid body type I cells. Adv Exp Med Biol 860: 61‐67, 2015.
 292.Qiao L, Maclean PS, Schaack J, Orlicky DJ, Darimont C, Pagliassotti M, Friedman JE, Shao J. C/EBPalpha regulates human adiponectin gene transcription through an intronic enhancer. Diabetes 54: 1744‐1754, 2005.
 293.Qiao L, Shao J. SIRT1 regulates adiponectin gene expression through Foxo1‐C/enhancer‐binding protein alpha transcriptional complex. J Biol Chem 281: 39915‐39924, 2006.
 294.Rahmouni K. Leptin‐induced sympathetic nerve activation: Signaling mechanisms and cardiovascular consequences in obesity. Curr Hypertens Rev 6: 104‐209, 2010.
 295.Rahmouni K, Correia ML, Haynes WG, Mark AL. Obesity‐associated hypertension: New insights into mechanisms. Hypertension 45: 9‐14, 2005.
 296.Rakoczy RJ, Pye RL, Fayyad TH, Santin JM, Barr BL, Wyatt CN. High fat feeding in rats alters respiratory parameters by a mechanism that is unlikely to be mediated by carotid body type I cells. Adv Exp Med Biol 1071: 137‐142, 2018.
 297.Rakoczy RJ, Wyatt CN. Acute oxygen sensing by the carotid body: A rattlebag of molecular mechanisms. J Physiol 596: 2969‐2976, 2018.
 298.Ramadan W, Dewasmes G, Petitjean M, Wiernsperger N, Delanaud S, Geloen A, Libert JP. Sleep apnea is induced by a high‐fat diet and reversed and prevented by metformin in non‐obese rats. Obesity 15: 1409‐1418, 2007.
 299.Ramirez JM, Garcia AJ 3rd, Anderson TM, Koschnitzky JE, Peng YJ, Kumar GK, Prabhakar NR. Central and peripheral factors contributing to obstructive sleep apneas. Respir Physiol Neurobiol 189: 344‐353, 2013.
 300.Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes 37: 1595‐1607, 1988.
 301.Regazzetti C, Peraldi P, Gremeaux T, Najem‐Lendom R, Ben‐Sahra I, Cormont M, Bost F, Le Marchand‐Brustel Y, Tanti JF, Giorgetti‐Peraldi S. Hypoxia decreases insulin signaling pathways in adipocytes. Diabetes 58: 95‐103, 2009.
 302.Resto M, O'Connor D, Leef K, Funanage V, Spear M, Locke R. Leptin levels in preterm human breast milk and infant formula. Pediatrics 108: E15, 2001.
 303.Rey S, Del Rio R, Alcayaga J, Iturriaga R. Chronic intermittent hypoxia enhances cat chemosensory and ventilatory responses to hypoxia. J Physiol 560: 577‐586, 2004.
 304.Ribeiro MJ, Guimarães JP, Sacramento JF, Conde S. Contribution of carotid body to leptin effects on ventilation and blood pressure in control and obese rats. ERJ Open Res 5 (Suppl 3): P77, 2019.
 305.Ribeiro MJ, Sacramento JF, Gallego‐Martin T, Olea E, Melo BF, Guarino MP, Yubero S, Obeso A, Conde SV. High fat diet blunts the effects of leptin on ventilation and on carotid body activity. J Physiol 596: 3187‐3199, 2018.
 306.Ribeiro MJ, Sacramento JF, Gonzalez C, Guarino MP, Monteiro EC, Conde SV. Carotid body denervation prevents the development of insulin resistance and hypertension induced by hypercaloric diets. Diabetes 62: 2905‐2916, 2013.
 307.Richter DW, Smith JC. Respiratory rhythm generation in vivo. Physiology 29: 58‐71, 2014.
 308.Richter DW, Spyer KM. Studying rhythmogenesis of breathing: Comparison of in vivo and in vitro models. Trends Neurosci 24: 464‐472, 2001.
 309.Rigatto H, Brady JP, de la Torre Verduzco R. Chemoreceptor reflexes in preterm infants: II. The effect of gestational and postnatal age on the ventilatory response to inhaled carbon dioxide. Pediatrics 55: 614‐620, 1975.
 310.Roberts BL, Bennett CM, Carroll JM, Lindsley SR, Kievit P. Early overnutrition alters synaptic signaling and induces leptin resistance in arcuate proopiomelanocortin neurons. Physiol Behav 206: 166‐174, 2019.
 311.Ross CA, Ruggiero DA, Joh TH, Park DH, Reis DJ. Rostral ventrolateral medulla: Selective projections to the thoracic autonomic cell column from the region containing C1 adrenaline neurons. J Comp Neurol 228: 168‐185, 1984.
 312.Rubinsztajn R, Kumor M, Byskiniewicz K, Bielicki P, Chazan R. Serum leptin concentration and sympathetic activation estimated on the adrenaline and noradrenaline serum concentration in patients with obstructive sleep apnea. Pol Arch Med Wewn 113: 544‐551, 2005.
 313.Sacramento JF, Chew DJ, Melo BF, Donega M, Dopson W, Guarino MP, Robinson A, Prieto‐Lloret J, Patel S, Holinski BJ, Ramnarain N, Pikov V, Famm K, Conde SV. Bioelectronic modulation of carotid sinus nerve activity in the rat: A potential therapeutic approach for type 2 diabetes. Diabetologia 61: 700‐710, 2018.
 314.Sacramento JF, Ribeiro MJ, Rodrigues T, Guarino MP, Diogo LN, Seica R, Monteiro EC, Matafome P, Conde SV. Insulin resistance is associated with tissue‐specific regulation of HIF‐1alpha and HIF‐2alpha during mild chronic intermittent hypoxia. Respir Physiol Neurobiol 228: 30‐38, 2016.
 315.Sacramento JF, Ribeiro MJ, Rodrigues T, Olea E, Melo BF, Guarino MP, Fonseca‐Pinto R, Ferreira CR, Coelho J, Obeso A, Seica R, Matafome P, Conde SV. Functional abolition of carotid body activity restores insulin action and glucose homeostasis in rats: Key roles for visceral adipose tissue and the liver. Diabetologia 60: 158‐168, 2017.
 316.Sainz N, Barrenetxe J, Moreno‐Aliaga MJ, Martinez JA. Leptin resistance and diet‐induced obesity: Central and peripheral actions of leptin. Metab Clin Exp 64: 35‐46, 2015.
 317.Sakurai T. The neural circuit of orexin (hypocretin): Maintaining sleep and wakefulness. Nat Rev Neurosci 8: 171‐181, 2007.
 318.Sakurai T. The role of orexin in motivated behaviours. Nat Rev Neurosci 15: 719‐731, 2014.
 319.Sakurai T, Amemiya A, Ishii M, Matsuzaki I, Chemelli RM, Tanaka H, Williams SC, Richarson JA, Kozlowski GP, Wilson S, Arch JR, Buckingham RE, Haynes AC, Carr SA, Annan RS, McNulty DE, Liu WS, Terrett JA, Elshourbagy NA, Bergsma DJ, Yanagisawa M. Orexins and orexin receptors: A family of hypothalamic neuropeptides and G protein‐coupled receptors that regulate feeding behavior. Cell 92: 1 page following 696, 1998.
 320.Samuelsson AM, Clark J, Rudyk O, Shattock MJ, Bae SE, South T, Pombo J, Redington K, Uppal E, Coen CW, Poston L, Taylor PD. Experimental hyperleptinemia in neonatal rats leads to selective leptin responsiveness, hypertension, and altered myocardial function. Hypertension 62: 627‐633, 2013.
 321.Samuelsson AM, Morris A, Igosheva N, Kirk SL, Pombo JM, Coen CW, Poston L, Taylor PD. Evidence for sympathetic origins of hypertension in juvenile offspring of obese rats. Hypertension 55: 76‐82, 2010.
 322.Samuelsson AS, Mullier A, Maicas N, Oosterhuis NR, Eun Bae S, Novoselova TV, Chan LF, Pombo JM, Taylor PD, Joles JA, Coen CW, Balthasar N, Poston L. Central role for melanocortin‐4 receptors in offspring hypertension arising from maternal obesity. Proc Natl Acad Sci U S A 113: 12298‐12303, 2016.
 323.Sanchez J, Oliver P, Miralles O, Ceresi E, Pico C, Palou A. Leptin orally supplied to neonate rats is directly uptaken by the immature stomach and may regulate short‐term feeding. Endocrinology 146: 2575‐2582, 2005.
 324.Sandoval D. Bariatric surgeries: Beyond restriction and malabsorption. Int J Obes 35 (Suppl 3): S45‐S49, 2011.
 325.Sanner BM, Kollhosser P, Buechner N, Zidek W, Tepel M. Influence of treatment on leptin levels in patients with obstructive sleep apnoea. Eur Respir J 23: 601‐604, 2004.
 326.Savino F, Nanni GE, Maccario S, Costamagna M, Oggero R, Silvestro L. Breast‐fed infants have higher leptin values than formula‐fed infants in the first four months of life. J Pediatr Endocrinol Metab 17: 1527‐1532, 2004.
 327.Savransky V, Bevans S, Nanayakkara A, Li J, Smith PL, Torbenson MS, Polotsky VY. Chronic intermittent hypoxia causes hepatitis in a mouse model of diet‐induced fatty liver. Am J Physiol Gastrointest Liver Physiol 293: G871‐G877, 2007.
 328.Schafer H, Pauleit D, Sudhop T, Gouni‐Berthold I, Ewig S, Berthold HK. Body fat distribution, serum leptin, and cardiovascular risk factors in men with obstructive sleep apnea. Chest 122: 829‐839, 2002.
 329.Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 270: 26746‐26749, 1995.
 330.Schultz HD. Angiotensin and carotid body chemoreception in heart failure. Curr Opin Pharmacol 11: 144‐149, 2011.
 331.Schulz C, Paulus K, Johren O, Lehnert H. Intranasal leptin reduces appetite and induces weight loss in rats with diet‐induced obesity (DIO). Endocrinology 153: 143‐153, 2012.
 332.Schulz C, Paulus K, Lehnert H. Central nervous and metabolic effects of intranasally applied leptin. Endocrinology 145: 2696‐2701, 2004.
 333.Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature 404: 661‐671, 2000.
 334.Semenza GL, Prabhakar NR. Neural regulation of hypoxia‐inducible factors and redox state drives the pathogenesis of hypertension in a rodent model of sleep apnea. J Appl Physiol 119: 1152‐1156, 2015.
 335.Senaris R, Garcia‐Caballero T, Casabiell X, Gallego R, Castro R, Considine RV, Dieguez C, Casanueva FF. Synthesis of leptin in human placenta. Endocrinology 138: 4501‐4504, 1997.
 336.Servantes DM, Javaheri S, Kravchychyn ACP, Storti LJ, Almeida DR, de Mello MT, Cintra FD, Tufik S, Bittencourt L. Effects of exercise training and CPAP in patients with heart failure and OSA: A preliminary study. Chest 154: 808‐817, 2018.
 337.Sharma SK, Agrawal S, Damodaran D, Sreenivas V, Kadhiravan T, Lakshmy R, Jagia P, Kumar A. CPAP for the metabolic syndrome in patients with obstructive sleep apnea. N Engl J Med 365: 2277‐2286, 2011.
 338.Shechter A. Effects of continuous positive airway pressure on energy balance regulation: A systematic review. Eur Respir J 48: 1640‐1657, 2016.
 339.Shimizu K, Chin K, Nakamura T, Masuzaki H, Ogawa Y, Hosokawa R, Niimi A, Hattori N, Nohara R, Sasayama S, Nakao K, Mishima M, Nakamura T, Ohi M. Plasma leptin levels and cardiac sympathetic function in patients with obstructive sleep apnoea‐hypopnoea syndrome. Thorax 57: 429‐434, 2002.
 340.Shin MK, Yao Q, Jun JC, Bevans‐Fonti S, Yoo DY, Han W, Mesarwi O, Richardson R, Fu YY, Pasricha PJ, Schwartz AR, Shirahata M, Polotsky VY. Carotid body denervation prevents fasting hyperglycemia during chronic intermittent hypoxia. J Appl Physiol 117: 765‐776, 2014.
 341.Shinohara E, Kihara S, Yamashita S, Yamane M, Nishida M, Arai T, Kotani K, Nakamura T, Takemura K, Matsuzawa Y. Visceral fat accumulation as an important risk factor for obstructive sleep apnoea syndrome in obese subjects. J Intern Med 241: 11‐18, 1997.
 342.Shirahata M, Tang WY, Shin MK, Polotsky VY. Is the carotid body a metabolic monitor? Adv Exp Med Biol 860: 153‐159, 2015.
 343.Skurk T, van Harmelen V, Blum WF, Hauner H. Angiotensin II promotes leptin production in cultured human fat cells by an ERK1/2‐dependent pathway. Obes Res 13: 969‐973, 2005.
 344.Smith HR, Leibold NK, Rappoport DA, Ginapp CM, Purnell BS, Bode NM, Alberico SL, Kim YC, Audero E, Gross CT, Buchanan GF. Dorsal raphe serotonin neurons mediate CO2‐induced arousal from sleep. J Neurosci Off J Soc Neurosci 38: 1915‐1925, 2018.
 345.Smith JC, Abdala AP, Koizumi H, Rybak IA, Paton JF. Spatial and functional architecture of the mammalian brain stem respiratory network: A hierarchy of three oscillatory mechanisms. J Neurophysiol 98: 3370‐3387, 2007.
 346.Smith JC, Ellenberger HH, Ballanyi K, Richter DW, Feldman JL. Pre‐Botzinger complex: A brainstem region that may generate respiratory rhythm in mammals. Science 254: 726‐729, 1991.
 347.Smith‐Kirwin SM, O'Connor DM, De Johnston J, Lancey ED, Hassink SG, Funanage VL. Leptin expression in human mammary epithelial cells and breast milk. J Clin Endocrinol Metab 83: 1810‐1813, 1998.
 348.Sobhani I, Bado A, Vissuzaine C, Buyse M, Kermorgant S, Laigneau JP, Attoub S, Lehy T, Henin D, Mignon M, Lewin MJ. Leptin secretion and leptin receptor in the human stomach. Gut 47: 178‐183, 2000.
 349.Somers VK, Dyken ME, Clary MP, Abboud FM. Sympathetic neural mechanisms in obstructive sleep apnea. J Clin Invest 96: 1897‐1904, 1995.
 350.Speretta GF, Lemes EV, Vendramini RC, Menani JV, Zoccal DB, Colombari E, Colombari DSA, Bassi M. High‐fat diet increases respiratory frequency and abdominal expiratory motor activity during hypercapnia. Respir Physiol Neurobiol 258: 32‐39, 2018.
 351.Speretta GF, Silva AA, Vendramini RC, Zanesco A, Delbin MA, Menani JV, Bassi M, Colombari E, Colombari DS. Resistance training prevents the cardiovascular changes caused by high‐fat diet. Life Sci 146: 154‐162, 2016.
 352.Stasche N. Selective indication for positive airway pressure (PAP) in sleep‐related breathing disorders with obstruction. GMS Curr Top Otorhinolaryngol Head Neck Surg 5: Doc06, 2006.
 353.Stefater MA, Perez‐Tilve D, Chambers AP, Wilson‐Perez HE, Sandoval DA, Berger J, Toure M, Tschop M, Woods SC, Seeley RJ. Sleeve gastrectomy induces loss of weight and fat mass in obese rats, but does not affect leptin sensitivity. Gastroenterology 138: 2426‐2436, 2436 e2421‐2423, 2010.
 354.Steinbrekera B, Roghair R. Modeling the impact of growth and leptin deficits on the neuronal regulation of blood pressure. J Endocrinol 231: R47‐R60, 2016.
 355.Stunden CE, Filosa JA, Garcia AJ, Dean JB, Putnam RW. Development of in vivo ventilatory and single chemosensitive neuron responses to hypercapnia in rats. Respir Physiol 127: 135‐155, 2001.
 356.Sugerman HJ, Baron PL, Fairman RP, Evans CR, Vetrovec GW. Hemodynamic dysfunction in obesity hypoventilation syndrome and the effects of treatment with surgically induced weight loss. Ann Surg 207: 604‐613, 1988.
 357.Sugerman HJ, Fairman RP, Sood RK, Engle K, Wolfe L, Kellum JM. Long‐term effects of gastric surgery for treating respiratory insufficiency of obesity. Am J Clin Nutr 55: 597S‐601S, 1992.
 358.Sullivan CE, Berthon‐Jones M, Issa FG. Nocturnal nasal‐airway pressure for sleep apnea. N Engl J Med 309: 112, 1983.
 359.Summer R, Walsh K, Medoff BD. Obesity and pulmonary arterial hypertension: Is adiponectin the molecular link between these conditions? Pulm Circ 1: 440‐447, 2011.
 360.Takemura Y, Ouchi N, Shibata R, Aprahamian T, Kirber MT, Summer RS, Kihara S, Walsh K. Adiponectin modulates inflammatory reactions via calreticulin receptor‐dependent clearance of early apoptotic bodies. J Clin Invest 117: 375‐386, 2007.
 361.Tan W, Janczewski WA, Yang P, Shao XM, Callaway EM, Feldman JL. Silencing preBotzinger complex somatostatin‐expressing neurons induces persistent apnea in awake rat. Nat Neurosci 11: 538‐540, 2008.
 362.Tankersley C, Kleeberger S, Russ B, Schwartz A, Smith P. Modified control of breathing in genetically obese (ob/ob) mice. J Appl Physiol 81: 716‐723, 1996.
 363.Tankersley CG, O'Donnell C, Daood MJ, Watchko JF, Mitzner W, Schwartz A, Smith P. Leptin attenuates respiratory complications associated with the obese phenotype. J Appl Physiol 85: 2261‐2269, 1998.
 364.Taylor PD, Samuelsson AM, Poston L. Maternal obesity and the developmental programming of hypertension: A role for leptin. Acta Physiol 210: 508‐523, 2014.
 365.Thorp AA, Schlaich MP. Relevance of sympathetic nervous system activation in obesity and metabolic syndrome. J Diabetes Res 2015: 341583, 2015.
 366.Trayhurn P. Hypoxia and adipose tissue function and dysfunction in obesity. Physiol Rev 93: 1‐21, 2013.
 367.Trayhurn P, Wood IS. Adipokines: Inflammation and the pleiotropic role of white adipose tissue. Br J Nutr 92: 347‐355, 2004.
 368.Trombetta IC, Batalha LT, Rondon MU, Laterza MC, Kuniyoshi FH, Gowdak MM, Barretto AC, Halpern A, Villares SM, Negrao CE. Weight loss improves neurovascular and muscle metaboreflex control in obesity. Am J Phys Heart Circ Phys 285: H974‐H982, 2003.
 369.Tsuchida A, Yamauchi T, Takekawa S, Hada Y, Ito Y, Maki T, Kadowaki T. Peroxisome proliferator‐activated receptor (PPAR)alpha activation increases adiponectin receptors and reduces obesity‐related inflammation in adipose tissue: Comparison of activation of PPARalpha, PPARgamma, and their combination. Diabetes 54: 3358‐3370, 2005.
 370.Ueno LM, Drager LF, Rodrigues AC, Rondon MU, Braga AM, Mathias W Jr, Krieger EM, Barretto AC, Middlekauff HR, Lorenzi‐Filho G, Negrao CE. Effects of exercise training in patients with chronic heart failure and sleep apnea. Sleep 32: 637‐647, 2009.
 371.Valuniene M, Verkauskiene R, Boguszewski M, Dahlgren J, Lasiene D, Lasas L, Wikland KA. Leptin levels at birth and in early postnatal life in small‐ and appropriate‐for‐gestational‐age infants. Medicina 43: 784‐791, 2007.
 372.Van De Wielle R, Michels N. Longitudinal associations of leptin and adiponectin with heart rate variability in children. Front Physiol 8: 498, 2017.
 373.Van Eyck A, Van Hoorenbeeck K, De Winter BY, Van Gaal L, De Backer W, Verhulst SL. Sleep disordered breathing and autonomic function in overweight and obese children and adolescents. ERJ Open Res 2, 2016.
 374.Varela L, Horvath TL. Leptin and insulin pathways in POMC and AgRP neurons that modulate energy balance and glucose homeostasis. EMBO Rep 13: 1079‐1086, 2012.
 375.Vgontzas AN, Papanicolaou DA, Bixler EO, Hopper K, Lotsikas A, Lin HM, Kales A, Chrousos GP. Sleep apnea and daytime sleepiness and fatigue: Relation to visceral obesity, insulin resistance, and hypercytokinemia. J Clin Endocrinol Metab 85: 1151‐1158, 2000.
 376.Wabitsch M, Funcke JB, Lennerz B, Kuhnle‐Krahl U, Lahr G, Debatin KM, Vatter P, Gierschik P, Moepps B, Fischer‐Posovszky P. Biologically inactive leptin and early‐onset extreme obesity. N Engl J Med 372: 48‐54, 2015.
 377.Wang B, Wood IS, Trayhurn P. Hypoxia induces leptin gene expression and secretion in human preadipocytes: Differential effects of hypoxia on adipokine expression by preadipocytes. J Endocrinol 198: 127‐134, 2008.
 378.Wang T, Hartzell DL, Rose BS, Flatt WP, Hulsey MG, Menon NK, Makula RA, Baile CA. Metabolic responses to intracerebroventricular leptin and restricted feeding. Physiol Behav 65: 839‐848, 1999.
 379.Wang ZV, Scherer PE. Adiponectin, the past two decades. J Mol Cell Biol 8: 93‐100, 2016.
 380.Wauman J, Zabeau L, Tavernier J. The leptin receptor complex: Heavier than expected? Front Endocrinol 8: 30, 2017.
 381.Wehrwein EA, Basu R, Basu A, Curry TB, Rizza RA, Joyner MJ. Hyperoxia blunts counterregulation during hypoglycaemia in humans: Possible role for the carotid bodies? J Physiol 588: 4593‐4601, 2010.
 382.Weiszenstein M, Shimoda LA, Koc M, Seda O, Polak J. Inhibition of lipolysis ameliorates diabetic phenotype in a mouse model of obstructive sleep apnea. Am J Respir Cell Mol Biol 55: 299‐307, 2016.
 383.Weltman A, Pritzlaff CJ, Wideman L, Considine RV, Fryburg DA, Gutgesell ME, Hartman ML, Veldhuis JD. Intensity of acute exercise does not affect serum leptin concentrations in young men. Med Sci Sports Exerc 32: 1556‐1561, 2000.
 384.Wewege M, van den Berg R, Ward RE, Keech A. The effects of high‐intensity interval training vs. moderate‐intensity continuous training on body composition in overweight and obese adults: A systematic review and meta‐analysis. Obes Rev 18: 635‐646, 2017.
 385.Whitehead JP, Richards AA, Hickman IJ, Macdonald GA, Prins JB. Adiponectin—a key adipokine in the metabolic syndrome. Diabetes Obes Metab 8: 264‐280, 2006.
 386.WHO. World Health Organization Obesity and Overweight. 2018. https://www.who.int/news‐room/fact‐sheets/detail/obesity‐and‐overweight (accessed 31 Jan 2020).
 387.Wolf J, Hering D, Narkiewicz K. Non‐dipping pattern of hypertension and obstructive sleep apnea syndrome. Hypertens Res 33: 867‐871, 2010.
 388.Yamauchi T, Iwabu M, Okada‐Iwabu M, Kadowaki T. Adiponectin receptors: A review of their structure, function and how they work. Best Pract Res Clin Endocrinol Metab 28: 15‐23, 2014.
 389.Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, Sugiyama T, Miyagishi M, Hara K, Tsunoda M, Murakami K, Ohteki T, Uchida S, Takekawa S, Waki H, Tsuno NH, Shibata Y, Terauchi Y, Froguel P, Tobe K, Koyasu S, Taira K, Kitamura T, Shimizu T, Nagai R, Kadowaki T. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 423: 762‐769, 2003.
 390.Yang Y, Yang S, Jiao X, Li J, Wu H, Sun H, Yang Y, Zhang M, Wei Y, Qin Y. Targeted sequencing analysis of the adiponectin gene identifies variants associated with obstructive sleep apnoea in Chinese Han population. Medicine 98: e15219, 2019.
 391.Yao Q, Pho H, Kirkness J, Ladenheim EE, Bi S, Moran TH, Fuller DD, Schwartz AR, Polotsky VY. Localizing effects of leptin on upper airway and respiratory control during sleep. Sleep 39: 1097‐1106, 2016.
 392.Ye J. Emerging role of adipose tissue hypoxia in obesity and insulin resistance. Int J Obes 33: 54‐66, 2009.
 393.Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am J Phys Endocrinol Metab 293: E1118‐E1128, 2007.
 394.Yeh ER, Erokwu B, LaManna JC, Haxhiu MA. The paraventricular nucleus of the hypothalamus influences respiratory timing and activity in the rat. Neurosci Lett 232: 63‐66, 1997.
 395.Yilmaz MI, Sonmez A, Caglar K, Gok DE, Eyileten T, Yenicesu M, Acikel C, Bingol N, Kilic S, Oguz Y, Vural A. Peroxisome proliferator‐activated receptor gamma (PPAR‐gamma) agonist increases plasma adiponectin levels in type 2 diabetic patients with proteinuria. Endocrine 25: 207‐214, 2004.
 396.Yoshikawa M, Yamauchi M, Fujita Y, Koyama N, Fukuoka A, Tamaki S, Yamamoto Y, Tomoda K, Kimura H. The impact of obstructive sleep apnea and nasal CPAP on circulating adiponectin levels. Lung 192: 289‐295, 2014.
 397.Yosunkaya S, Okur HK, Can U, Zamani A, Kutlu R. Impact of continuous positive airway pressure treatment on leptin levels in patients with obstructive sleep apnea syndrome. Metab Syndr Relat Disord 13: 272‐277, 2015.
 398.Young T, Finn L, Peppard PE, Szklo‐Coxe M, Austin D, Nieto FJ, Stubbs R, Hla KM. Sleep disordered breathing and mortality: Eighteen‐year follow‐up of the Wisconsin sleep cohort. Sleep 31: 1071‐1078, 2008.
 399.Yuan F, Wang H, Feng J, Wei Z, Yu H, Zhang X, Zhang Y, Wang S. Leptin signaling in the carotid body regulates a hypoxic ventilatory response through altering TASK channel expression. Front Physiol 9: 249, 2018.
 400.Zhang W, Fukuda Y, Kuwaki T. Respiratory and cardiovascular actions of orexin‐A in mice. Neurosci Lett 385: 131‐136, 2005.
 401.Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature 372: 425‐432, 1994.
 402.Zhou SY, Gilbey MP. Respiratory‐related activity of lower thoracic and upper lumbar sympathetic preganglionic neurones in the rat. J Physiol 451: 631‐642, 1992.
 403.Zoccal DB, Bonagamba LG, Paton JF, Machado BH. Sympathetic‐mediated hypertension of awake juvenile rats submitted to chronic intermittent hypoxia is not linked to baroreflex dysfunction. Exp Physiol 94: 972‐983, 2009.
 404.Zoccal DB, Furuya WI, Bassi M, Colombari DS, Colombari E. The nucleus of the solitary tract and the coordination of respiratory and sympathetic activities. Front Physiol 5: 238, 2014.
 405.Zoccal DB, Silva JN, Barnett WH, Lemes EV, Falquetto B, Colombari E, Molkov YI, Moreira TS, Takakura AC. Interaction between the retrotrapezoid nucleus and the parafacial respiratory group to regulate active expiration and sympathetic activity in rats. Am J Physiol Lung Cell Mol Physiol 315: L891‐L909, 2018.
 406.Zoccal DB, Simms AE, Bonagamba LG, Braga VA, Pickering AE, Paton JF, Machado BH. Increased sympathetic outflow in juvenile rats submitted to chronic intermittent hypoxia correlates with enhanced expiratory activity. J Physiol 586: 3253‐3265, 2008.

Contact Editor

Submit a note to the editor about this article by filling in the form below.

* Required Field

How to Cite

Estelle B. Gauda, Silvia Conde, Mirian Bassi, Daniel B. Zoccal, Debora Simoes Almeida Colombari, Eduardo Colombari, Nikola Despotovic. Leptin: Master Regulator of Biological Functions that Affects Breathing. Compr Physiol 2020, 10: 1047-1083. doi: 10.1002/cphy.c190031