Comprehensive Physiology Wiley Online Library

Perinatal Hypoxemia and Oxygen Sensing

Full Article on Wiley Online Library



Abstract

The development of the control of breathing begins in utero and continues postnatally. Fetal breathing movements are needed for establishing connectivity between the lungs and central mechanisms controlling breathing. Maturation of the control of breathing, including the increase of hypoxia chemosensitivity, continues postnatally. Insufficient oxygenation, or hypoxia, is a major stressor that can manifest for different reasons in the fetus and neonate. Though the fetus and neonate have different hypoxia sensing mechanisms and respond differently to acute hypoxia, both responses prevent deviations to respiratory and other developmental processes. Intermittent and chronic hypoxia pose much greater threats to the normal developmental respiratory processes. Gestational intermittent hypoxia, due to maternal sleep‐disordered breathing and sleep apnea, increases eupneic breathing and decreases the hypoxic ventilatory response associated with impaired gasping and autoresuscitation postnatally. Chronic fetal hypoxia, due to biologic or environmental (i.e. high‐altitude) factors, is implicated in fetal growth restriction and preterm birth causing a decrease in the postnatal hypoxic ventilatory responses with increases in irregular eupneic breathing. Mechanisms driving these changes include delayed chemoreceptor development, catecholaminergic activity, abnormal myelination, increased astrocyte proliferation in the dorsal respiratory group, among others. Long‐term high‐altitude residents demonstrate favorable adaptations to chronic hypoxia as do their offspring. Neonatal intermittent hypoxia is common among preterm infants due to immature respiratory systems and thus, display a reduced drive to breathe and apneas due to insufficient hypoxic sensitivity. However, ongoing intermittent hypoxia can enhance hypoxic sensitivity causing ventilatory overshoots followed by apnea; the number of apneas is positively correlated with degree of hypoxic sensitivity in preterm infants. Chronic neonatal hypoxia may arise from fetal complications like maternal smoking or from postnatal cardiovascular problems, causing blunting of the hypoxic ventilatory responses throughout at least adolescence due to attenuation of carotid body fibers responses to hypoxia with potential roles of brainstem serotonin, microglia, and inflammation, though these effects depend on the age in which chronic hypoxia initiates. Fetal and neonatal intermittent and chronic hypoxia are implicated in preterm birth and complicate the respiratory system through their direct effects on hypoxia sensing mechanisms and interruptions to the normal developmental processes. Thus, precise regulation of oxygen homeostasis is crucial for normal development of the respiratory control network. © 2021 American Physiological Society. Compr Physiol 11:1653‐1677, 2021.

Figure 1. Figure 1. Adaptation of the fetus to low oxygen environment present in utero. Fetal oxygen supply is provided by maternal blood bathing the chorionic villi in the placenta. The umbilical vein carries the oxygenated blood to inferior vena cava (IVC) and eventually across the foramen ovale to left atrium and left ventricle to be pumped into coronary and cerebral circulations. The oxygen saturation of fetal blood in different sites are indicated by dark shaded boxes. The percent of cardiac output distributed to each organ is indicated in plain text. The mean values for fetal biventricular output, range of normal Hb concentrations and fetal PaO2 are indicated in the text box to the right, along with the HbP50 for fetal Hb 197,257,268. Reused, with permission, from Richard A. Polin and William W Fox, 2016, Fetal and Neonatal Physiology, Ed: Polin, Abman, Rowitch, Benitz and Fox, 5th edition, Lakshminrusimha and Steinhorn “Pathophysiology of PPHN,” pp 1576‐1587. Copyright Satyan Lakshminrusimha.
Figure 2. Figure 2. Effects of ibotenic acid lesioning within or sparing the thalamic parafascicular (Pf) nuclear complex in neonatal lambs on the depression phase of the hypoxic ventilatory response. Lesioning the Pf removes the ventilatory depression at 10 and 15 min (decline 10 and 15, respectively) expressed as a change in ventilation from prelesion values (upper left) or as a percent of the augmentation phase (lower left) (A). Lesions sparing the Pf have no effect on the hypoxic ventilatory decline (B). *P < 0.005, **P < 0.03 compared with augmentation phase. P < 0.005 versus prelesion at same time, P < 0.05 versus thalamic lesions sparing Pf at same time. Adapted, with permission, from Koos BJ, et al., 2016 164.
Figure 3. Figure 3. The thalamic parafascicular nuclear region mediates hypoxic depression of fetal breathing movements in fetal sheep. Hypoxia‐induced suppression of breathing and associated eye movements is completely removed following lesioning of the thalamic parafascicular nuclear region by ibotenic (IBO) acid (A). Adapted, with permission, from Koos BJ, 2002 163. In a similar experiment using exogenous adenosine, the known neurochemical mediating hypoxia‐induced suppression of fetal breathing suppresses breathing and associated eye movements before IBO injection but is significantly impaired following IBO lesioning of the thalamic parafascicular nuclear region (B). Adapted, with permission, from Koos BJ, et al., 2000 162.
Figure 4. Figure 4. Schematic summary of in‐text descriptions of the effects fetal normoxia and fetal acute, intermittent, and chronic hypoxia have on breathing. During normal development, the periodicity and regularity of FBM activity steadily rises and approaches postnatal breathing regularity at birth. Upon birth, few apneas are present and quickly diminishes with age. The hypoxic ventilatory response (HVR) begins with a significant secondary role‐off pertaining to a decrease in metabolic rate following an initial increase in ventilation (VE). This biphasic HVR quickly matures (within weeks of birth) to a sustained increase in ventilation as the metabolic roll‐off diminishes in effect. Fetal acute hypoxia causes a decrease in FBM incidence. Fetal intermittent hypoxia, also called gestational intermittent hypoxia, decreases FBM hourly incidence but increases FBM amplitude and inspiratory efforts during the hypoxic episodes. Postnatally, fetal intermittent hypoxia reduces the HVR and capacity for gasping and autoresuscitation and an increase in eupneic breathing. Fetal chronic hypoxia is typically observed in high‐altitude births where infants are born with lower weight. Infants also have increased apneas at birth associated with diminished and delayed development of the HVR. Eupneic breathing is fairly normal aside from the apneas. See text for references.
Figure 5. Figure 5. Rate of IUGR at altitude and differences in uterine blood flow during pregnancy between nonnative (European) and native (Andean) high‐altitude populations. The rate of IUGR increases with altitude and at high‐altitude Europeans have a fivefold greater occurrence of IUGR compared to Andeans after adjusting for other fetal growth factors (shown in the graph are unadjusted values; A). A potential explanation for the IUGR rate disparity at altitude between Andeans and Europeans is the compensatory twofold greater increase in uteroplacental oxygen delivery in Andean compared to European women at 36 weeks' gestation indicating potential genetic adaptions across generations (B). NP, nonpregnant. *P < 0.05, **P < 0.01. Reused, with permission, from Julian CG, 2011 138.
Figure 6. Figure 6. Distribution of apnea types lasting 3 to 15 s in eight term (gestational age: 39.5 ± 0.3 weeks) and eight preterm (gestational age: 34.3 ± 0.4 weeks) infants measured between birth and 56 weeks old. A total of 783 and 4086 apneas were recorded in term and preterm infant groups, respectively. Reused, with permission, from Lee D, et al., 1987 170. © 1987, Springer Nature.
Figure 7. Figure 7. Schematic summary of in‐text descriptions of the effects neonatal intermittent and chronic hypoxia have on breathing. Infants born premature (24‐32 weeks) retain a biphasic fetal hypoxic ventilatory response (HVR) which is associated with decreased hypoxic arousal and autorescuscitation but increased number of apneas (which plateaus around 10 weeks after birth). The increased apneas are a reflection of apnea of prematurity but also enhanced hypoxic sensitivity which can cause ventilatory overshoots and trigger the CO2 apneic threshold. Term infants exposed to neonatal intermittent hypoxia have increased eupneic breathing, HVR, and more apneas though arousal from hypoxia is reduced. Exposure to chronic neonatal hypoxia commencing after birth is associated with reduced HVR [in lambs and male (M) rats]. The effects on eupneic breathing are equivocal and thus remain to be fully elucidated. Infants that continue to be exposed to chronic hypoxia after birth (i.e. high‐altitude births) demonstrate increased periodic breathing but the effects on the HVR and eupneic breathing are variable across high altitude populations. See text for references.
Figure 8. Figure 8. Effects of intermittent hypoxic episodes on carotid body hypoxic sensitivity and long‐term facilitation in neonatal versus adult rats. Progressive increases in intermittent hypoxic episodes (36, 72, 216, and 720) cause progressive hypoxic sensitization of the carotid bodies in neonates (A) whereas hypoxic sensitization requires 720 episodes in adults and is not as robust as in neonates (B). The hypoxic sensitization in the neonatal (C) but not the adult (D) rats is sustained after their return to normoxia for 10 days. Intermittent hypoxia causes long‐term facilitation in adult rats (E) but not in neonatal rats (F) despite having enhanced carotid body hypoxic sensitivity. Reused, with permission, from Pawar A, et al., 2008 243.
Figure 9. Figure 9. Schematic of the hypothesized cellular and molecular mechanisms of central oxygen sensing by the astrocyte. Hypoxia sensed from perfusing blood by the astrocyte inhibits the mitochondria and thus stimulating mitochondrial reactive oxygen species (ROS) production, leading to an increase in lipid peroxidation. PLC‐IP3 signaling releases intracellular calcium stores and releases ATP onto nearby preBötC neurons that express two ATP receptors, P2YR and P2XR. In parallel, hypoxia may cause opening of the connexin (Cx) hemichannel leading to release of ATP and lactate, the latter with unknown stimulatory effects on the preBötC neurons. Release of ATP causes further release of ATP in autocrine and paracrine manners, increasing respiratory rate and sympathetic activity through preBötC neurons. Reused, with permission, from Gourine AV and Funk GD, 2017 106. © 1985, The American Physiological Society.


Figure 1. Adaptation of the fetus to low oxygen environment present in utero. Fetal oxygen supply is provided by maternal blood bathing the chorionic villi in the placenta. The umbilical vein carries the oxygenated blood to inferior vena cava (IVC) and eventually across the foramen ovale to left atrium and left ventricle to be pumped into coronary and cerebral circulations. The oxygen saturation of fetal blood in different sites are indicated by dark shaded boxes. The percent of cardiac output distributed to each organ is indicated in plain text. The mean values for fetal biventricular output, range of normal Hb concentrations and fetal PaO2 are indicated in the text box to the right, along with the HbP50 for fetal Hb 197,257,268. Reused, with permission, from Richard A. Polin and William W Fox, 2016, Fetal and Neonatal Physiology, Ed: Polin, Abman, Rowitch, Benitz and Fox, 5th edition, Lakshminrusimha and Steinhorn “Pathophysiology of PPHN,” pp 1576‐1587. Copyright Satyan Lakshminrusimha.


Figure 2. Effects of ibotenic acid lesioning within or sparing the thalamic parafascicular (Pf) nuclear complex in neonatal lambs on the depression phase of the hypoxic ventilatory response. Lesioning the Pf removes the ventilatory depression at 10 and 15 min (decline 10 and 15, respectively) expressed as a change in ventilation from prelesion values (upper left) or as a percent of the augmentation phase (lower left) (A). Lesions sparing the Pf have no effect on the hypoxic ventilatory decline (B). *P < 0.005, **P < 0.03 compared with augmentation phase. P < 0.005 versus prelesion at same time, P < 0.05 versus thalamic lesions sparing Pf at same time. Adapted, with permission, from Koos BJ, et al., 2016 164.


Figure 3. The thalamic parafascicular nuclear region mediates hypoxic depression of fetal breathing movements in fetal sheep. Hypoxia‐induced suppression of breathing and associated eye movements is completely removed following lesioning of the thalamic parafascicular nuclear region by ibotenic (IBO) acid (A). Adapted, with permission, from Koos BJ, 2002 163. In a similar experiment using exogenous adenosine, the known neurochemical mediating hypoxia‐induced suppression of fetal breathing suppresses breathing and associated eye movements before IBO injection but is significantly impaired following IBO lesioning of the thalamic parafascicular nuclear region (B). Adapted, with permission, from Koos BJ, et al., 2000 162.


Figure 4. Schematic summary of in‐text descriptions of the effects fetal normoxia and fetal acute, intermittent, and chronic hypoxia have on breathing. During normal development, the periodicity and regularity of FBM activity steadily rises and approaches postnatal breathing regularity at birth. Upon birth, few apneas are present and quickly diminishes with age. The hypoxic ventilatory response (HVR) begins with a significant secondary role‐off pertaining to a decrease in metabolic rate following an initial increase in ventilation (VE). This biphasic HVR quickly matures (within weeks of birth) to a sustained increase in ventilation as the metabolic roll‐off diminishes in effect. Fetal acute hypoxia causes a decrease in FBM incidence. Fetal intermittent hypoxia, also called gestational intermittent hypoxia, decreases FBM hourly incidence but increases FBM amplitude and inspiratory efforts during the hypoxic episodes. Postnatally, fetal intermittent hypoxia reduces the HVR and capacity for gasping and autoresuscitation and an increase in eupneic breathing. Fetal chronic hypoxia is typically observed in high‐altitude births where infants are born with lower weight. Infants also have increased apneas at birth associated with diminished and delayed development of the HVR. Eupneic breathing is fairly normal aside from the apneas. See text for references.


Figure 5. Rate of IUGR at altitude and differences in uterine blood flow during pregnancy between nonnative (European) and native (Andean) high‐altitude populations. The rate of IUGR increases with altitude and at high‐altitude Europeans have a fivefold greater occurrence of IUGR compared to Andeans after adjusting for other fetal growth factors (shown in the graph are unadjusted values; A). A potential explanation for the IUGR rate disparity at altitude between Andeans and Europeans is the compensatory twofold greater increase in uteroplacental oxygen delivery in Andean compared to European women at 36 weeks' gestation indicating potential genetic adaptions across generations (B). NP, nonpregnant. *P < 0.05, **P < 0.01. Reused, with permission, from Julian CG, 2011 138.


Figure 6. Distribution of apnea types lasting 3 to 15 s in eight term (gestational age: 39.5 ± 0.3 weeks) and eight preterm (gestational age: 34.3 ± 0.4 weeks) infants measured between birth and 56 weeks old. A total of 783 and 4086 apneas were recorded in term and preterm infant groups, respectively. Reused, with permission, from Lee D, et al., 1987 170. © 1987, Springer Nature.


Figure 7. Schematic summary of in‐text descriptions of the effects neonatal intermittent and chronic hypoxia have on breathing. Infants born premature (24‐32 weeks) retain a biphasic fetal hypoxic ventilatory response (HVR) which is associated with decreased hypoxic arousal and autorescuscitation but increased number of apneas (which plateaus around 10 weeks after birth). The increased apneas are a reflection of apnea of prematurity but also enhanced hypoxic sensitivity which can cause ventilatory overshoots and trigger the CO2 apneic threshold. Term infants exposed to neonatal intermittent hypoxia have increased eupneic breathing, HVR, and more apneas though arousal from hypoxia is reduced. Exposure to chronic neonatal hypoxia commencing after birth is associated with reduced HVR [in lambs and male (M) rats]. The effects on eupneic breathing are equivocal and thus remain to be fully elucidated. Infants that continue to be exposed to chronic hypoxia after birth (i.e. high‐altitude births) demonstrate increased periodic breathing but the effects on the HVR and eupneic breathing are variable across high altitude populations. See text for references.


Figure 8. Effects of intermittent hypoxic episodes on carotid body hypoxic sensitivity and long‐term facilitation in neonatal versus adult rats. Progressive increases in intermittent hypoxic episodes (36, 72, 216, and 720) cause progressive hypoxic sensitization of the carotid bodies in neonates (A) whereas hypoxic sensitization requires 720 episodes in adults and is not as robust as in neonates (B). The hypoxic sensitization in the neonatal (C) but not the adult (D) rats is sustained after their return to normoxia for 10 days. Intermittent hypoxia causes long‐term facilitation in adult rats (E) but not in neonatal rats (F) despite having enhanced carotid body hypoxic sensitivity. Reused, with permission, from Pawar A, et al., 2008 243.


Figure 9. Schematic of the hypothesized cellular and molecular mechanisms of central oxygen sensing by the astrocyte. Hypoxia sensed from perfusing blood by the astrocyte inhibits the mitochondria and thus stimulating mitochondrial reactive oxygen species (ROS) production, leading to an increase in lipid peroxidation. PLC‐IP3 signaling releases intracellular calcium stores and releases ATP onto nearby preBötC neurons that express two ATP receptors, P2YR and P2XR. In parallel, hypoxia may cause opening of the connexin (Cx) hemichannel leading to release of ATP and lactate, the latter with unknown stimulatory effects on the preBötC neurons. Release of ATP causes further release of ATP in autocrine and paracrine manners, increasing respiratory rate and sympathetic activity through preBötC neurons. Reused, with permission, from Gourine AV and Funk GD, 2017 106. © 1985, The American Physiological Society.
References
 1.Abu‐Shaweesh JM, Martin RJ. Neonatal apnea: What's new? Pediatr Pulmonol 43 (10): 937‐944, 2008.
 2.Adzick NS, Harrison MR, Glick PL, Villa RL, Finkbeiner W. Experimental pulmonary hypoplasia and oligohydramnios: Relative contributions of lung fluid and fetal breathing movements. J Pediatr Surg 19 (6): 658‐665, 1984.
 3.Alcorn D, Adamson TM, Maloney JE, Robinson PM. Morphological effects of chronic bilateral phrenectomy or vagotomy in the fetal lamb lung. J Anat 130 (Pt 4): 683‐695, 1980.
 4.Al‐Matary A, Kutbi I, Qurashi M, Khalil M, Alvaro R, Kwiatkowski K, Cates D, Rigatto H. Increased peripheral chemoreceptor activity may be critical in destabilizing breathing in neonates. Semin Perinatol 28 (4): 264‐272, 2004.
 5.Alvaro R, Rigatto H. Control of breathing in newborns. In: Bracci R, Buonocore G, Weindling M, editors. Neonatology. Milano: Springer, 2012.
 6.Angelova PR, Kasymov V, Christie I, Sheikhbahaei S, Turovsky E, Marina N, Korsak A, Zwicker J, Teschemacher AG, Ackland GL, Funk GD, Kasparov S, Abramov AY, Gourine AV. Functional oxygen sensitivity of astrocytes. J Neurosci 35 (29): 10460‐10473, 2015.
 7.Aucott SW, Donohue PK, Northington FJ. Increased morbidity in severe early intrauterine growth restriction. J Perinatol 24 (7): 435‐440, 2004.
 8.Bach KB, Mitchell GS. Hypoxia‐induced long‐term facilitation of respiratory activity is serotonin dependent. Respir Physiol 104 (2–3): 251‐260, 1996.
 9.Baguma‐Nibasheka M, Gugic D, Saraga‐Babic M, Kablar B. Role of skeletal muscle in lung development. Histol Histopathol 27 (7): 817‐826, 2012.
 10.Bairam A, Basson H, Marchal F, Cottet‐Emard JM, Pequignot JM, Hascoet JM, Lahiri S. Effects of hypoxia on carotid body dopamine content and release in developing rabbits. J Appl Physiol (1985) 80 (1): 20‐24, 1996.
 11.Balan KV, Kc P, Hoxha Z, Mayer CA, Wilson CG, Martin RJ. Vagal afferents modulate cytokine‐mediated respiratory control at the neonatal medulla oblongata. Respir Physiol Neurobiol 178 (3): 458‐464, 2011.
 12.Balan KV, Kc P, Mayer CA, Wilson CG, Belkadi A, Martin RJ. Intrapulmonary lipopolysaccharide exposure upregulates cytokine expression in the neonatal brainstem. Acta Paediatr 101 (5): 466‐471, 2012.
 13.Barcroft J, Barron DH. Movements in midfoetal life in the sheep embryo. J Physiol 91 (3): 329‐351, 1937.
 14.Barrett KT, Dosumu‐Johnson RT, Daubenspeck JA, Brust RD, Kreouzis V, Kim JC, Li A, Dymecki SM, Nattie EE. Partial raphe dysfunction in neurotransmission is sufficient to increase mortality after anoxic exposures in mice at a critical period in postnatal development. J Neurosci 36 (14): 3943‐3953, 2016.
 15.Bartelds B, van Bel F, Teitel DF, Rudolph AM. Carotid, not aortic, chemoreceptors mediate the fetal cardiovascular response to acute hypoxemia in lambs. Pediatr Res 34 (1): 51‐55, 1993.
 16.Bates ML, Pillers DA, Palta M, Farrell ET, Eldridge MW. Ventilatory control in infants, children, and adults with bronchopulmonary dysplasia. Respir Physiol Neurobiol 189 (2): 329‐337, 2013.
 17.Battaglia FC, Lubchenco LO. A practical classification of newborn infants by weight and gestational age. J Pediatr 71 (2): 159‐163, 1967.
 18.Bavis RW. Developmental plasticity of the hypoxic ventilatory response after perinatal hyperoxia and hypoxia. Respir Physiol Neurobiol 149 (1–3): 287‐299, 2005.
 19.Bavis RW, Fallon SC, Dmitrieff EF. Chronic hyperoxia and the development of the carotid body. Respir Physiol Neurobiol 185 (1): 94‐104, 2013.
 20.Bavis RW, MacFarlane PM. Developmental plasticity in the neural control of breathing. Exp Neurol 287 (Pt 2): 176‐191, 2017.
 21.Bavis RW, Mitchell GS. Intermittent hypoxia induces phrenic long‐term facilitation in carotid‐denervated rats. J Appl Physiol (1985) 94 (1): 399‐409, 2003.
 22.Bavis RW, Olson EB Jr, Vidruk EH, Fuller DD, Mitchell GS. Developmental plasticity of the hypoxic ventilatory response in rats induced by neonatal hypoxia. J Physiol 557 (Pt 2): 645‐660, 2004.
 23.Bavis RW, Young KM, Barry KJ, Boller MR, Kim E, Klein PM, Ovrutsky AR, Rampersad DA. Chronic hyperoxia alters the early and late phases of the hypoxic ventilatory response in neonatal rats. J Appl Physiol (1985) 109 (3): 796‐803, 2010.
 24.Bavis RW, Kim I, Pradhan N, Nawreen N, Dmitrieff EF, Carroll JL, Donnelly DF. Recovery of carotid body O2 sensitivity following chronic postnatal hyperoxia in rats. Respir Physiol Neurobiol 177 (1): 47‐55, 2011.
 25.Bavis RW, van Heerden ES, Brackett DG, Harmeling LH, Johnson SM, Blegen HJ, Logan S, Nguyen GN, Fallon SC. Postnatal development of eupneic ventilation and metabolism in rats chronically exposed to moderate hyperoxia. Respir Physiol Neurobiol 198: 1‐12, 2014.
 26.Bavis RW, Li KY, DeAngelis KJ, March RJ, Wallace JA, Logan S, Putnam RW. Ventilatory and chemoreceptor responses to hypercapnia in neonatal rats chronically exposed to moderate hyperoxia. Respir Physiol Neurobiol 237: 22‐34, 2017.
 27.Beall CM, Brittenham GM, Macuaga F, Barragan M. Variation in hemoglobin concentration among samples of high‐altitude natives in the Andes and the Himalayas. Am J Hum Biol 2 (6): 639‐651, 1990.
 28.Beall CM, Strohl KP, Blangero J, Williams‐Blangero S, Almasy LA, Decker MJ, Worthman CM, Goldstein MC, Vargas E, Villena M, Soria R, Alarcon AM, Gonzales C. Ventilation and hypoxic ventilatory response of Tibetan and Aymara high altitude natives. Am J Phys Anthropol 104 (4): 427‐447, 1997.
 29.Beall CM, Cavalleri GL, Deng L, Elston RC, Gao Y, Knight J, Li C, Li JC, Liang Y, McCormack M, Montgomery HE, Pan H, Robbins PA, Shianna KV, Tam SC, Tsering N, Veeramah KR, Wang W, Wangdui P, Weale ME, Xu Y, Xu Z, Yang L, Zaman MJ, Zeng C, Zhang L, Zhang X, Zhaxi P, Zheng YT. Natural selection on EPAS1 (HIF2alpha) associated with low hemoglobin concentration in Tibetan highlanders. Proc Natl Acad Sci USA 107 (25): 11459‐11464, 2010.
 30.Berra E, Benizri E, Ginouves A, Volmat V, Roux D, Pouyssegur J. HIF prolyl‐hydroxylase 2 is the key oxygen sensor setting low steady‐state levels of HIF‐1alpha in normoxia. EMBO J 22 (16): 4082‐4090, 2003.
 31.Bigham AW, Wilson MJ, Julian CG, Kiyamu M, Vargas E, Leon‐Velarde F, Rivera‐Chira M, Rodriquez C, Browne VA, Parra E, Brutsaert TD, Moore LG, Shriver MD. Andean and Tibetan patterns of adaptation to high altitude. Am J Hum Biol 25 (2): 190‐197, 2013.
 32.Bisgard GE, Olson EB Jr, Bavis RW, Wenninger J, Nordheim EV, Mitchell GS. Carotid chemoafferent plasticity in adult rats following developmental hyperoxia. Respir Physiol Neurobiol 145 (1): 3‐11, 2005.
 33.Bishop T, Ratcliffe PJ. HIF hydroxylase pathways in cardiovascular physiology and medicine. Circ Res 117 (1): 65‐79, 2015.
 34.Bishop T, Talbot NP, Turner PJ, Nicholls LG, Pascual A, Hodson EJ, Douglas G, Fielding JW, Smith TG, Demetriades M, Schofield CJ, Robbins PA, Pugh CW, Buckler KJ, Ratcliffe PJ. Carotid body hyperplasia and enhanced ventilatory responses to hypoxia in mice with heterozygous deficiency of PHD2. J Physiol 591 (14): 3565‐3577, 2013.
 35.Blanco CE, Dawes GS, Hanson MA, McCooke HB. The response to hypoxia of arterial chemoreceptors in fetal sheep and new‐born lambs. J Physiol 351: 25‐37, 1984.
 36.Blitz DM, Ramirez JM. Long‐term modulation of respiratory network activity following anoxia in vitro. J Neurophysiol 87 (6): 2964‐2971, 2002.
 37.Blott M, Greenough A, Nicolaides KH, Campbell S. The ultrasonographic assessment of the fetal thorax and fetal breathing movements in the prediction of pulmonary hypoplasia. Early Hum Dev 21 (3): 143‐151, 1990.
 38.Boddy K, Dawes GS, Fisher R, Pinter S, Robinson JS. Foetal respiratory movements, electrocortical and cardiovascular responses to hypoxaemia and hypercapnia in sheep. J Physiol 243 (3): 599‐618, 1974.
 39.Bose C, Van Marter LJ, Laughon M, O'Shea TM, Allred EN, Karna P, Ehrenkranz RA, Boggess K, Leviton A, Extremely Low Gestational Age Newborn Study I. Fetal growth restriction and chronic lung disease among infants born before the 28th week of gestation. Pediatrics 124 (3): e450‐e458, 2009.
 40.Bouvier J, Thoby‐Brisson M, Renier N, Dubreuil V, Ericson J, Champagnat J, Pierani A, Chedotal A, Fortin G. Hindbrain interneurons and axon guidance signaling critical for breathing. Nat Neurosci 13 (9): 1066‐1074, 2010.
 41.Brady JP, Ceruti E. Chemoreceptor reflexes in the new‐born infant: Effects of varying degrees of hypoxia on heart rate and ventilation in a warm environment. J Physiol 184 (3): 631‐645, 1966.
 42.Branchfield K, Nantie L, Verheyden JM, Sui P, Wienhold MD, Sun X. Pulmonary neuroendocrine cells function as airway sensors to control lung immune response. Science 351 (6274): 707‐710, 2016.
 43.Breen S, Rees S, Walker D. Identification of brainstem neurons responding to hypoxia in fetal and newborn sheep. Brain Res 748 (1–2): 107‐121, 1997.
 44.Bright FM, Byard RW, Vink R, Paterson DS. Medullary serotonin neuron abnormalities in an Australian cohort of sudden infant death syndrome. J Neuropathol Exp Neurol 76 (10): 864‐873, 2017.
 45.Buchanan GF, Richerson GB. Central serotonin neurons are required for arousal to CO2. Proc Natl Acad Sci USA 107 (37): 16354‐16359, 2010.
 46.Calder NA, Williams BA, Smyth J, Boon AW, Kumar P, Hanson MA. Absence of ventilatory responses to alternating breaths of mild hypoxia and air in infants who have had bronchopulmonary dysplasia: Implications for the risk of sudden infant death. Pediatr Res 35 (6): 677‐681, 1994.
 47.Cardot V, Chardon K, Tourneux P, Micallef S, Stephan E, Leke A, Bach V, Libert JP, Telliez F. Ventilatory response to a hyperoxic test is related to the frequency of short apneic episodes in late preterm neonates. Pediatr Res 62 (5): 591‐596, 2007.
 48.Carroll JL. Developmental plasticity in respiratory control. J Appl Physiol (1985) 94 (1): 375‐389, 2003.
 49.Carroll JL, Agarwal A. Development of ventilatory control in infants. Paediatr Respir Rev 11 (4): 199‐207, 2010.
 50.Carroll JL, Kim I. Postnatal development of carotid body glomus cell O2 sensitivity. Respir Physiol Neurobiol 149 (1–3): 201‐215, 2005.
 51.Carter AM. Placental gas exchange and the oxygen supply to the fetus. Compr Physiol 5 (3): 1381‐1403, 2015.
 52.Cerpa VJ, Aylwin Mde L, Beltran‐Castillo S, Bravo EU, Llona IR, Richerson GB, Eugenin JL. The alteration of neonatal raphe neurons by prenatal‐perinatal nicotine. Meaning for sudden infant death syndrome. Am J Respir Cell Mol Biol 53 (4): 489‐499, 2015.
 53.Cerpa VJ, Wu Y, Bravo E, Teran FA, Flynn RS, Richerson GB. Medullary 5‐HT neurons: Switch from tonic respiratory drive to chemoreception during postnatal development. Neuroscience 344: 1‐14, 2017.
 54.Chang RB, Strochlic DE, Williams EK, Umans BD, Liberles SD. Vagal sensory neuron subtypes that differentially control breathing. Cell 161 (3): 622‐633, 2015.
 55.Clewlow F, Dawes GS, Johnston BM, Walker DW. Changes in breathing, electrocortical and muscle activity in unanaesthetized fetal lambs with age. J Physiol 341: 463‐476, 1983.
 56.Collaco JM, McGrath‐Morrow SA. Respiratory phenotypes for preterm infants, children, and adults: Bronchopulmonary dysplasia and more. Ann Am Thorac Soc 15 (5): 530‐538, 2018.
 57.Cotton EK, Grunstein MM. Effects of hypoxia on respiratory control in neonates at high altitude. J Appl Physiol Respir Environ Exerc Physiol 48 (4): 587‐595, 1980.
 58.Covello KL, Kehler J, Yu H, Gordan JD, Arsham AM, Hu C‐J, Labosky PA, Simon MC, Keith B. HIF‐2α regulates Oct‐4: Effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev 20 (5): 557‐570, 2006.
 59.Cummings KJ, Hewitt JC, Li A, Daubenspeck JA, Nattie EE. Postnatal loss of brainstem serotonin neurones compromises the ability of neonatal rats to survive episodic severe hypoxia. J Physiol 589 (Pt 21): 5247‐5256, 2011.
 60.Cummings KJ, Commons KG, Hewitt JC, Daubenspeck JA, Li A, Kinney HC, Nattie EE. Failed heart rate recovery at a critical age in 5‐HT‐deficient mice exposed to episodic anoxia: Implications for SIDS. J Appl Physiol 111 (3): 825‐833, 2011.
 61.Cutz E. Hyperplasia of pulmonary neuroendocrine cells in infancy and childhood. Semin Diagn Pathol 32 (6): 420‐437, 2015.
 62.Cutz E, Yeger H, Pan J. Pulmonary neuroendocrine cell system in pediatric lung disease‐recent advances. Pediatr Dev Pathol 10 (6): 419‐435, 2007.
 63.Cutz E, Perrin DG, Pan J, Haas EA, Krous HF. Pulmonary neuroendocrine cells and neuroepithelial bodies in sudden infant death syndrome: Potential markers of airway chemoreceptor dysfunction. Pediatr Dev Pathol 10 (2): 106‐116, 2007.
 64.Darnall RA. The carotid body and arousal in the fetus and neonate. Respir Physiol Neurobiol 185 (1): 132‐143, 2013.
 65.Davey MG, Moss TJ, McCrabb GJ, Harding R. Prematurity alters hypoxic and hypercapnic ventilatory responses in developing lambs. Respir Physiol 105 (1–2): 57‐67, 1996.
 66.Dawes GS. The central control of fetal breathing and skeletal muscle movements. J Physiol 346: 1‐18, 1984.
 67.Delpapa EH, Edelstone DI, Milley JR, Balsan M. Effects of chronic maternal anemia on systemic and uteroplacental oxygenation in near‐term pregnant sheep. Am J Obstet Gynecol 166 (3): 1007‐1012, 1992.
 68.Dempsey JA. Crossing the apnoeic threshold: Causes and consequences. Exp Physiol 90 (1): 13‐24, 2005.
 69.Deoras KS, Greenspan JS, Wolfson MR, Keklikian EN, Shaffer TH, Allen JL. Effects of inspiratory resistive loading on chest wall motion and ventilation: Differences between preterm and full‐term infants. Pediatr Res 32 (5): 589‐594, 1992.
 70.Di Fiore JM, Vento M. Intermittent hypoxemia and oxidative stress in preterm infants. Respir Physiol Neurobiol 266: 121‐129, 2019.
 71.Di Fiore JM, Bloom JN, Orge F, Schutt A, Schluchter M, Cheruvu VK, Walsh M, Finer N, Martin RJ. A higher incidence of intermittent hypoxemic episodes is associated with severe retinopathy of prematurity. J Pediatr 157 (1): 69‐73, 2010.
 72.Di Pasquale E, Tell F, Monteau R, Hilaire G. Perinatal developmental changes in respiratory activity of medullary and spinal neurons: An in vitro study on fetal and newborn rats. Brain Res Dev Brain Res 91 (1): 121‐130, 1996.
 73.Donnelly DF, Doyle TP. Developmental changes in hypoxia‐induced catecholamine release from rat carotid body, in vitro. J Physiol 475 (2): 267‐275, 1994.
 74.Dosumu‐Johnson RT, Cocoran AE, Chang Y, Nattie E, Dymecki SM. Acute perturbation of Pet1‐neuron activity in neonatal mice impairs cardiorespiratory homeostatic recovery. elife 7: e37857, 2018.
 75.Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational hypoxia and developmental plasticity. Physiol Rev 98 (3): 1241‐1334, 2018.
 76.Duncan JR, Paterson DS, Hoffman JM, Mokler DJ, Borenstein NS, Belliveau RA, Krous HF, Haas EA, Stanley C, Nattie EE, Trachtenberg FL, Kinney HC. Brainstem serotonergic deficiency in sudden infant death syndrome. JAMA 303 (5): 430‐437, 2010.
 77.Dunwoodie SL. The role of hypoxia in development of the mammalian embryo. Dev Cell 17 (6): 755‐773, 2009.
 78.Edelstone DI, Caine ME, Fumia FD. Relationship of fetal oxygen consumption and acid‐base balance to fetal hematocrit. Am J Obstet Gynecol 151 (7): 844‐851, 1985.
 79.Eden GJ, Hanson MA. Effects of chronic hypoxia from birth on the ventilatory response to acute hypoxia in the newborn rat. J Physiol 392: 11‐19, 1987.
 80.Eichenwald EC, Committee on Fetus and Newborn, American Academy of Pediatrics. Apnea of prematurity. Pediatrics 137 (1): e20153757, 2016.
 81.Ek M, Engblom D, Saha S, Blomqvist A, Jakobsson PJ, Ericsson‐Dahlstrand A. Inflammatory response: Pathway across the blood‐brain barrier. Nature 410 (6827): 430‐431, 2001.
 82.Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med 364 (7): 656‐665, 2011.
 83.Endo A, Minato M, Takada M, Takahashi S, Harada K, Yamada T, Takashima S. A case of pulmonary hypoplasia associated with intrauterine brainstem necrosis. Eur J Pediatr 160 (11): 675‐676, 2001.
 84.Erickson JT. Central serotonin and autoresuscitation capability in mammalian neonates. Exp Neurol 326: 113162, 2020.
 85.Erickson JT, Sposato BC. Autoresuscitation responses to hypoxia‐induced apnea are delayed in newborn 5‐HT‐deficient Pet‐1 homozygous mice. J Appl Physiol (1985) 106 (6): 1785‐1792, 2009.
 86.Ewald AC, Kiernan EA, Baker TL, Watters JJ. The transcriptomic profiles of microglia from respiratory control centers in adult offspring exposed to gestational intermittent hypoxia differ by sex and CNS region. FASEB J 33 (1_supplement): 730.7‐730.7, 2019.
 87.Fanaroff AA, Korones SB, Wright LL, Verter J, Poland RL, Bauer CR, Tyson JE, Philips JB 3rd, Edwards W, Lucey JF, Catz CS, Shankaran S, Oh W. Incidence, presenting features, risk factors and significance of late onset septicemia in very low birth weight infants. The National Institute of Child Health and Human Development Neonatal Research Network. Pediatr Infect Dis J 17 (7): 593‐598, 1998.
 88.Fewell JE, Baker SB. Arousal from sleep during rapidly developing hypoxemia in lambs. Pediatr Res 22 (4): 471‐477, 1987.
 89.Fewell JE, Baker SB. Arousal and cardiopulmonary responses to hyperoxic hypercapnia in lambs. J Dev Physiol 12 (1): 21‐26, 1989.
 90.Fewell JE, Konduri GG. Repeated exposure to rapidly developing hypoxemia influences the interaction between oxygen and carbon dioxide in initiating arousal from sleep in lambs. Pediatr Res 24 (1): 28‐33, 1988.
 91.Fewell JE, Konduri GG. Influence of repeated exposure to rapidly developing hypoxaemia on the arousal and cardiopulmonary response to rapidly developing hypoxaemia in lambs. J Dev Physiol 11 (2): 77‐82, 1989.
 92.Fewell JE, Lee CC, Kitterman JA. Effects of phrenic nerve section on the respiratory system of fetal lambs. J Appl Physiol 51 (2): 293‐297, 1981.
 93.Fewell JE, Kondo CS, Dascalu V, Filyk SC. Influence of carotid‐denervation on the arousal and cardiopulmonary responses to alveolar hypercapnia in lambs. J Dev Physiol 12 (4): 193‐199, 1989.
 94.Fewell JE, Kondo CS, Dascalu V, Filyk SC. Influence of carotid denervation on the arousal and cardiopulmonary response to rapidly developing hypoxemia in lambs. Pediatr Res 25 (5): 473‐477, 1989.
 95.Finer NN, Barrington KJ, Hayes BJ, Hugh A. Obstructive, mixed, and central apnea in the neonate: Physiologic correlates. J Pediatr 121 (6): 943‐950, 1992.
 96.Forster HV, Pan LG, Lowry TF, Serra A, Wenninger J, Martino P. Important role of carotid chemoreceptor afferents in control of breathing of adult and neonatal mammals. Respir Physiol 119 (2–3): 199‐208, 2000.
 97.Funk GD, Gourine AV. CrossTalk proposal: A central hypoxia sensor contributes to the excitatory hypoxic ventilatory response. J Physiol 596 (15): 2935‐2938, 2018.
 98.Garite TJ, Clark R, Thorp JA. Intrauterine growth restriction increases morbidity and mortality among premature neonates. Am J Obstet Gynecol 191 (2): 481‐487, 2004.
 99.Gassmann NN, van Elteren HA, Goos TG, Morales CR, Rivera‐Ch M, Martin DS, Cabala Peralta P, Passano Del Carpio A, Aranibar Machaca S, Huicho L, Reiss IK, Gassmann M, de Jonge RC. Pregnancy at high altitude in the Andes leads to increased total vessel density in healthy newborns. J Appl Physiol (1985) 121 (3): 709‐715, 2016.
 100.Gauda EB, Shirahata M, Mason A, Pichard LE, Kostuk EW, Chavez‐Valdez R. Inflammation in the carotid body during development and its contribution to apnea of prematurity. Respir Physiol Neurobiol 185 (1): 120‐131, 2013.
 101.Giussani DA. The fetal brain sparing response to hypoxia: Physiological mechanisms. J Physiol 594 (5): 1215‐1230, 2016.
 102.Giussani DA, Spencer JA, Moore PJ, Bennet L, Hanson MA. Afferent and efferent components of the cardiovascular reflex responses to acute hypoxia in term fetal sheep. J Physiol 461: 431‐449, 1993.
 103.Giussani DA, Gardner DS, Cox DT, Fletcher AJ. Purinergic contribution to circulatory, metabolic, and adrenergic responses to acute hypoxemia in fetal sheep. Am J Physiol Regul Integr Comp Physiol 280 (3): R678‐R685, 2001.
 104.Goldstein JD, Reid LM. Pulmonary hypoplasia resulting from phrenic nerve agenesis and diaphragmatic amyoplasia. J Pediatr 97 (2): 282‐287, 1980.
 105.Gortner L, Misselwitz B, Milligan D, Zeitlin J, Kollee L, Boerch K, Agostino R, Van Reempts P, Chabernaud JL, Breart G, Papiernik E, Jarreau PH, Carrapato M, Gadzinowski J, Draper E, Members of the MRG. Rates of bronchopulmonary dysplasia in very preterm neonates in Europe: Results from the MOSAIC cohort. Neonatology 99 (2): 112‐117, 2011.
 106.Gourine AV, Funk GD. On the existence of a central respiratory oxygen sensor. J Appl Physiol (1985) 123 (5): 1344‐1349, 2017.
 107.Gourine AV, Llaudet E, Dale N, Spyer KM. Release of ATP in the ventral medulla during hypoxia in rats: Role in hypoxic ventilatory response. J Neurosci 25 (5): 1211‐1218, 2005.
 108.Gozal D, Gozal E, Reeves SR, Lipton AJ. Gasping and autoresuscitation in the developing rat: Effect of antecedent intermittent hypoxia. J Appl Physiol (1985) 92 (3): 1141‐1144, 2002.
 109.Gozal D, Reeves SR, Row BW, Neville JJ, Guo SZ, Lipton AJ. Respiratory effects of gestational intermittent hypoxia in the developing rat. Am J Respir Crit Care Med 167 (11): 1540‐1547, 2003.
 110.Gray PA, Hayes JA, Ling GY, Llona I, Tupal S, Picardo MC, Ross SE, Hirata T, Corbin JG, Eugenin J, Del Negro CA. Developmental origin of preBotzinger complex respiratory neurons. J Neurosci 30 (44): 14883‐14895, 2010.
 111.Greenspan JS, Wolfson MR, Locke RG, Allen JL, Shaffer TH. Increased respiratory drive and limited adaptation to loaded breathing in bronchopulmonary dysplasia. Pediatr Res 32 (3): 356‐359, 1992.
 112.Greer JJ. Control of breathing activity in the fetus and newborn. Compr Physiol 2 (3): 1873‐1888, 2012.
 113.Greer JJ, Smith JC, Feldman JL. Respiratory and locomotor patterns generated in the fetal rat brain stem‐spinal cord in vitro. J Neurophysiol 67 (4): 996‐999, 1992.
 114.Hannhart B, Pickett CK, Moore LG. Effects of estrogen and progesterone on carotid body neural output responsiveness to hypoxia. J Appl Physiol (1985) 68 (5): 1909‐1916, 1990.
 115.Hanson MA, Eden GJ, Nijhuis JG, Moore PJ. Peripheral chemoreceptors and other oxygen sensors in the fetus and newborn. In: Lahiri S, Forster RE, Davies RO, Pack AI, editors. Chemoreceptors and Reflexes in Breathing: Cellular and Molecular Aspects. New York: Oxford University Press, 1989, p. 113‐120.
 116.Hanson MA, Kumar P, Williams BA. The effect of chronic hypoxia upon the development of respiratory chemoreflexes in the newborn kitten. J Physiol 411: 563‐574, 1989.
 117.Harding R, Hooper SB, Han VK. Abolition of fetal breathing movements by spinal cord transection leads to reductions in fetal lung liquid volume, lung growth, and IGF‐II gene expression. Pediatr Res 34 (2): 148‐153, 1993.
 118.Harding R, Tester ML, Moss TJ, Davey MG, Louey S, Joyce B, Hooper SB, Maritz G. Effects of intra‐uterine growth restriction on the control of breathing and lung development after birth. Clin Exp Pharmacol Physiol 27 (1–2): 114‐119, 2000.
 119.Herlenius E. An inflammatory pathway to apnea and autonomic dysregulation. Respir Physiol Neurobiol 178 (3): 449‐457, 2011.
 120.Hoch B, Bernhard M. Central apnoea and endogenous prostaglandins in neonates. Acta Paediatr 89 (11): 1364‐1368, 2000.
 121.Hocker AD, Beyeler SA, Gardner AN, Johnson SM, Watters JJ, Huxtable AG. One bout of neonatal inflammation impairs adult respiratory motor plasticity in male and female rats. elife 8: e45399, 2019.
 122.Hodges MR, Richerson GB. Contributions of 5‐HT neurons to respiratory control: Neuromodulatory and trophic effects. Respir Physiol Neurobiol 164: 222‐232, 2008.
 123.Hofstetter AO, Saha S, Siljehav V, Jakobsson PJ, Herlenius E. The induced prostaglandin E2 pathway is a key regulator of the respiratory response to infection and hypoxia in neonates. Proc Natl Acad Sci USA 104 (23): 9894‐9899, 2007.
 124.Holton P, Wood JB. The effects of bilateral removal of the carotid bodies and denervation of the carotid sinuses in two human subjects. J Physiol 181 (2): 365‐378, 1965.
 125.Hu CJ, Poth JM, Zhang H, Flockton A, Laux A, Kumar S, McKeon B, Frid MG, Mouradian G, Li M, Riddle S, Pugliese SC, Brown RD, Wallace EM, Graham BB, Stenmark KR. Suppression of HIF2 signalling attenuates the initiation of hypoxia‐induced pulmonary hypertension. Eur Respir J, 2019.
 126.Huxtable AG, Vinit S, Windelborn JA, Crader SM, Guenther CH, Watters JJ, Mitchell GS. Systemic inflammation impairs respiratory chemoreflexes and plasticity. Respir Physiol Neurobiol 178 (3): 482‐489, 2011.
 127.Inanlou MR, Kablar B. Contractile activity of skeletal musculature involved in breathing is essential for normal lung cell differentiation, as revealed in Myf5−/−:MyoD−/− embryos. Dev Dyn 233 (3): 772‐782, 2005.
 128.Inanlou MR, Kablar B. Abnormal development of the intercostal muscles and the rib cage in Myf5−/− embryos leads to pulmonary hypoplasia. Dev Dyn 232 (1): 43‐54, 2005.
 129.Itskovitz J, LaGamma EF, Rudolph AM. The effect of reducing umbilical blood flow on fetal oxygenation. Am J Obstet Gynecol 145 (7): 813‐818, 1983.
 130.Itskovitz J, LaGamma EF, Bristow J, Rudolph AM. Cardiovascular responses to hypoxemia in sinoaortic‐denervated fetal sheep. Pediatr Res 30 (4): 381‐385, 1991.
 131.Iyer NV, Kotch LE, Agani F, Leung SW, Laughner E, Wenger RH, Gassmann M, Gearhart JD, Lawler AM, Yu AY, Semenza GL. Cellular and developmental control of O2 homeostasis by hypoxia‐inducible factor 1 alpha. Genes Dev 12 (2): 149‐162, 1998.
 132.Jacono FJ, Mayer CA, Hsieh YH, Wilson CG, Dick TE. Lung and brainstem cytokine levels are associated with breathing pattern changes in a rodent model of acute lung injury. Respir Physiol Neurobiol 178 (3): 429‐438, 2011.
 133.Jawdeh EGA. Intermittent hypoxemia in preterm infants: Etiology and clinical relevance. NeoReviews 18 (11): e637‐e646, 2017.
 134.Jensen GM, Moore LG. The effect of high altitude and other risk factors on birthweight: Independent or interactive effects? Am J Public Health 87 (6): 1003‐1007, 1997.
 135.Johnson SM, Randhawa KS, Epstein JJ, Gustafson E, Hocker AD, Huxtable AG, Baker TL, Watters JJ. Gestational intermittent hypoxia increases susceptibility to neuroinflammation and alters respiratory motor control in neonatal rats. Respir Physiol Neurobiol 256: 128‐142, 2018.
 136.Johnston BM, Gluckman PD. Lateral pontine lesions affect central chemosensitivity in unanesthetized fetal lambs. J Appl Physiol (1985) 67 (3): 1113‐1118, 1989.
 137.Johnston BM, Gluckman PD. Peripheral chemoreceptors respond to hypoxia in pontine‐lesioned fetal lambs in utero. J Appl Physiol (1985) 75 (3): 1027‐1034, 1993.
 138.Julian CG. High altitude during pregnancy. Clin Chest Med 32 (1): 21‐31, 2011.
 139.Julian CG, Moore LG. Human genetic adaptation to high altitude: Evidence from the Andes. Genes (Basel) 10 (2): 150, 2019.
 140.Julian CG, Vargas E, Armaza JF, Wilson MJ, Niermeyer S, Moore LG. High‐altitude ancestry protects against hypoxia‐associated reductions in fetal growth. Arch Dis Child Fetal Neonatal Ed 92 (5): F372‐F377, 2007.
 141.Julien C, Bairam A, Joseph V. Chronic intermittent hypoxia reduces ventilatory long‐term facilitation and enhances apnea frequency in newborn rats. Am J Physiol Regul Integr Comp Physiol 294 (4): R1356‐R1366, 2008.
 142.Kaelin WG Jr, Ratcliffe PJ. Oxygen sensing by metazoans: The central role of the HIF hydroxylase pathway. Mol Cell 30 (4): 393‐402, 2008.
 143.Katz‐Salamon M, Jonsson B, Lagercrantz H. Blunted peripheral chemoreceptor response to hyperoxia in a group of infants with bronchopulmonary dysplasia. Pediatr Pulmonol 20 (2): 101‐106, 1995.
 144.Kc A, Singhal N, Gautam J, Rana N, Andersson O. Effect of early versus delayed cord clamping in neonate on heart rate, breathing and oxygen saturation during first 10 minutes of birth ‐ randomized clinical trial. Matern Health Neonatol Perinatol 5: 7, 2019.
 145.Kiernan EA, Wang T, Vanderplow AM, Cherukuri S, Cahill ME, Watters JJ. Neonatal intermittent hypoxia induces lasting sex‐specific augmentation of rat microglial cytokine expression. Front Immunol 10: 1479, 2019.
 146.Kingdom JC, Kaufmann P. Oxygen and placental villous development: Origins of fetal hypoxia. Placenta 18 (8): 613‐621, 1997. discussion 623–626.
 147.Kinney HC, Myers MM, Belliveau RA, Randall LL, Trachtenberg FL, Fingers ST, Youngman M, Habbe D, Fifer WP. Subtle autonomic and respiratory dysfunction in sudden infant death syndrome associated with serotonergic brainstem abnormalities: A case report. J Neuropathol Exp Neurol 64 (8): 689‐694, 2005.
 148.Kinney HC, Richerson GB, Dymecki SM, Darnall RA, Nattie EE. The brainstem and serotonin in the sudden infant death syndrome. Annu Rev Pathol 4: 517‐550, 2009.
 149.Kline DD, Peng YJ, Manalo DJ, Semenza GL, Prabhakar NR. Defective carotid body function and impaired ventilatory responses to chronic hypoxia in mice partially deficient for hypoxia‐inducible factor 1 alpha. Proc Natl Acad Sci USA 99 (2): 821‐826, 2002.
 150.Kobayashi K, Lemke RP, Greer JJ. Ultrasound measurements of fetal breathing movements in the rat. J Appl Physiol (1985) 91 (1): 316‐320, 2001.
 151.Koch H, Caughie C, Elsen FP, Doi A, Garcia AJ 3rd, Zanella S, Ramirez JM. Prostaglandin E2 differentially modulates the central control of eupnoea, sighs and gasping in mice. J Physiol 593 (1): 305‐319, 2015.
 152.Koos BJ. Adenosine A(2)a receptors and O(2) sensing in development. Am J Physiol Regul Integr Comp Physiol 301 (3): R601‐R622, 2011.
 153.Koos BJ, Chau A. Fetal cardiovascular and breathing responses to an adenosine A2a receptor agonist in sheep. Am J Physiol 274 (1): R152‐R159, 1998.
 154.Koos BJ, Chau A, Ogunyemi D. Adenosine mediates metabolic and cardiovascular responses to hypoxia in fetal sheep. J Physiol 488 (Pt 3): 761‐766, 1995.
 155.Koos BJ, Maeda T. Adenosine A(2A) receptors mediate cardiovascular responses to hypoxia in fetal sheep. Am J Physiol Heart Circ Physiol 280 (1): H83‐H89, 2001.
 156.Koos BJ, Matsuda K. Fetal breathing, sleep state, and cardiovascular responses to adenosine in sheep. J Appl Physiol (1985) 68 (2): 489‐495, 1990.
 157.Koos BJ, Rajaee A. Fetal breathing movements and changes at birth. Adv Exp Med Biol 814: 89‐101, 2014.
 158.Koos BJ, Sameshima H, Power GG. Fetal breathing movement, sleep state and cardiovascular responses to an inhibitor of mitochondrial ATPase in sheep. J Dev Physiol 8 (1): 67‐75, 1986.
 159.Koos BJ, Sameshima H, Power GG. Fetal breathing, sleep state, and cardiovascular responses to graded anemia in sheep. J Appl Physiol (1985) 63 (4): 1463‐1468, 1987.
 160.Koos BJ, Mason BA, Punla O, Adinolfi AM. Hypoxic inhibition of breathing in fetal sheep: Relationship to brain adenosine concentrations. J Appl Physiol (1985) 77 (6): 2734‐2739, 1994.
 161.Koos BJ, Chau A, Matsuura M, Punla O, Kruger L. Thalamic locus mediates hypoxic inhibition of breathing in fetal sheep. J Neurophysiol 79 (5): 2383‐2393, 1998.
 162.Koos BJ, Chau A, Matsuura M, Punla O, Kruger L. Thalamic lesions dissociate breathing inhibition by hypoxia and adenosine in fetal sheep. Am J Physiol Regul Integr Comp Physiol 278 (4): R831‐R837, 2000.
 163.Koos BJ, Maeda T, Jan C, Lopez G. Adenosine A(2A) receptors mediate hypoxic inhibition of fetal breathing in sheep. Am J Obstet Gynecol 186 (4): 663‐668, 2002.
 164.Koos BJ, Rajaee A, Ibe B, Guerra C, Kruger L. Thalamic mediation of hypoxic respiratory depression in lambs. Am J Physiol Regul Integr Comp Physiol 310 (7): R586‐R595, 2016.
 165.Kumar P, Prabhakar NR. Peripheral chemoreceptors: Function and plasticity of the carotid body. Compr Physiol 2 (1): 141‐219, 2012.
 166.Lahiri S, Brody JS, Motoyama EK, Velasquez TM. Regulation of breathing in newborns at high altitude. J Appl Physiol Respir Environ Exerc Physiol 44 (5): 673‐678, 1978.
 167.Lahiri S, Roy A, Baby SM, Hoshi T, Semenza GL, Prabhakar NR. Oxygen sensing in the body. Prog Biophys Mol Biol 91 (3): 249‐286, 2006.
 168.Lakshminrusimha S, Russell JA, Steinhorn RH, Swartz DD, Ryan RM, Gugino SF, Wynn KA, Kumar VH, Mathew B, Kirmani K, Morin FC 3rd. Pulmonary hemodynamics in neonatal lambs resuscitated with 21%, 50%, and 100% oxygen. Pediatr Res 62 (3): 313‐318, 2007.
 169.Lal MK, Manktelow BN, Draper ES, Field DJ. Chronic lung disease of prematurity and intrauterine growth retardation: A population‐based study. Pediatrics 111 (3): 483‐487, 2003.
 170.Lee D, Caces R, Kwiatkowski K, Cates D, Rigatto H. A developmental study on types and frequency distribution of short apneas (3 to 15 seconds) in term and preterm infants. Pediatr Res 22 (3): 344‐349, 1987.
 171.Lee SK, Ye XY, Singhal N, De La Rue S, Lodha A, Shah PS, Canadian Neonatal N. Higher altitude and risk of bronchopulmonary dysplasia among preterm infants. Am J Perinatol 30 (7): 601‐606, 2013.
 172.Lee SY, Sirieix CM, Nattie E, Li A. Pre‐ and early postnatal nicotine exposure exacerbates autoresuscitation failure in serotonin‐deficient rat neonates. J Physiol 596 (23): 5977‐5991, 2018.
 173.Liggins GC, Vilos GA, Campos GA, Kitterman JA, Lee CH. The effect of spinal cord transection on lung development in fetal sheep. J Dev Physiol 3 (4): 267‐274, 1981.
 174.Lijowska AS, Reed NW, Chiodini BA, Thach BT. Sequential arousal and airway‐defensive behavior of infants in asphyxial sleep environments. J Appl Physiol (1985) 83 (1): 219‐228, 1997.
 175.Ling L, Olson EB Jr, Vidruk EH, Mitchell GS. Attenuation of the hypoxic ventilatory response in adult rats following one month of perinatal hyperoxia. J Physiol 495 (Pt 2): 561‐571, 1996.
 176.Liu J, Boujedaini N, Cazin L, Mallet E, Clabaut M. Developmental changes in cardio‐respiratory responses to hypoxia and hypercapnia in anesthetized low‐birth‐weight rats. Respir Physiol 123 (3): 189‐199, 2000.
 177.Liu J, Dong C, Cazin L, Clabaut M, Dubuc I, Costentin J, Coquerel A. Developmental changes of (3)H‐labelled mu‐opioid receptors in brainstems of intra‐uterine growth‐restricted rats. Brain Res Dev Brain Res 126 (2): 211‐215, 2001.
 178.Liu Q, Lowry TF, Wong‐Riley MT. Postnatal changes in ventilation during normoxia and acute hypoxia in the rat: Implication for a sensitive period. J Physiol 577 (Pt 3): 957‐970, 2006.
 179.Livermore S, Zhou Y, Pan J, Yeger H, Nurse CA, Cutz E. Pulmonary neuroepithelial bodies are polymodal airway sensors: Evidence for CO2/H+ sensing. Am J Physiol Lung Cell Mol Physiol 308 (8): L807‐L815, 2015.
 180.Livermore S, Pan J, Yeger H, Ratcliffe P, Bishop T, Cutz E. Augmented 5‐HT secretion in pulmonary neuroepithelial bodies from PHD1 null mice. Adv Exp Med Biol 860: 309‐313, 2015.
 181.Loboda A, Jozkowicz A, Dulak J. HIF‐1 and HIF‐2 transcription factors—similar but not identical. Mol Cells 29 (5): 435‐442, 2010.
 182.LoMauro A, Aliverti A. Respiratory physiology of pregnancy: Physiology masterclass. Breathe (Sheff) 11 (4): 297‐301, 2015.
 183.Lorea‐Hernandez JJ, Morales T, Rivera‐Angulo AJ, Alcantara‐Gonzalez D, Pena‐Ortega F. Microglia modulate respiratory rhythm generation and autoresuscitation. Glia 64 (4): 603‐619, 2016.
 184.Lorenzo FR, Huff C, Myllymaki M, Olenchock B, Swierczek S, Tashi T, Gordeuk V, Wuren T, Ri‐Li G, McClain DA, Khan TM, Koul PA, Guchhait P, Salama ME, Xing J, Semenza GL, Liberzon E, Wilson A, Simonson TS, Jorde LB, Kaelin WG Jr, Koivunen P, Prchal JT. A genetic mechanism for Tibetan high‐altitude adaptation. Nat Genet 46 (9): 951‐956, 2014.
 185.Lovering AT, Elliott JE, Laurie SS, Beasley KM, Gust CE, Mangum TS, Gladstone IM, Duke JW. Ventilatory and sensory responses in adult survivors of preterm birth and bronchopulmonary dysplasia with reduced exercise capacity. Ann Am Thorac Soc 11 (10): 1528‐1537, 2014.
 186.Lucius H, Gahlenbeck H, Kleine HO, Fabel H, Bartels H. Respiratory functions, buffer system, and electrolyte concentrations of blood during human pregnancy. Respir Physiol 9 (3): 311‐317, 1970.
 187.Lun R, Zhang C, Fewell JE. Carotid chemoreceptors do not mediate hypoxic‐induced gasping and autoresuscitation in newborn rats. Respir Physiol Neurobiol 212‐214: 33‐38, 2015.
 188.MacFarlane PM, Mayer CA, Litvin DG. Microglia modulate brainstem serotonergic expression following neonatal sustained hypoxia exposure: Implications for sudden infant death syndrome. J Physiol 594 (11): 3079‐3094, 2016.
 189.MacLean JE, DeHaan K, Fuhr D, Hariharan S, Kamstra B, Hendson L, Adatia I, Majaesic C, Lovering AT, Thompson RB, Nicholas D, Thebaud B, Stickland MK. Altered breathing mechanics and ventilatory response during exercise in children born extremely preterm. Thorax 71 (11): 1012‐1019, 2016.
 190.Majmundar AJ, Wong WJ, Simon MC. Hypoxia‐inducible factors and the response to hypoxic stress. Mol Cell 40 (2): 294‐309, 2010.
 191.Malloy MH, Hoffman HJ. Prematurity, sudden infant death syndrome, and age of death. Pediatrics 96 (3 Pt 1): 464‐471, 1995.
 192.Mariani G, Dik PB, Ezquer A, Aguirre A, Esteban ML, Perez C, Fernandez Jonusas S, Fustinana C. Pre‐ductal and post‐ductal O2 saturation in healthy term neonates after birth. J Pediatr 150 (4): 418‐421, 2007.
 193.Martin RJ, DiFiore JM, Jana L, Davis RL, Miller MJ, Coles SK, Dick TE. Persistence of the biphasic ventilatory response to hypoxia in preterm infants. J Pediatr 132 (6): 960‐964, 1998.
 194.Martin RJ, Di Fiore JM, Macfarlane PM, Wilson CG. Physiologic basis for intermittent hypoxic episodes in preterm infants. Adv Exp Med Biol 758: 351‐358, 2012.
 195.Martin‐Body RL, Johnston BM. Central origin of the hypoxic depression of breathing in the newborn. Respir Physiol 71 (1): 25‐32, 1988.
 196.Martinez‐Garcia MA, Campos‐Rodriguez F, Nagore E, Martorell A, Rodriguez‐Peralto JL, Riveiro‐Falkenbach E, Hernandez L, Banuls J, Arias E, Ortiz P, Cabriada V, Gardeazabal J, Montserrat JM, Carrera C, Corral J, Masa JF, de Terreros JG, Abad J, Boada A, Mediano O, de Eusebio E, Chiner E, Landete P, Mayos M, Fortuno A, Barbe F, Sanchez de la Torre M, Sanchez de la Torre A, Cano I, Gonzalez C, Perez‐Gil A, Gomez‐Garcia T, Cullen D, Somoza M, Formigon M, Aizpuru F, Navarro C, Selma‐Ferrer MJ, Garcia‐Ortega A, de Unamuno B, Almendros I, Farre R, Gozal D, Spanish Sleep N. Sleep‐disordered breathing is independently associated with increased aggressiveness of cutaneous melanoma: A multicenter observational study in 443 patients. Chest 154 (6): 1348‐1358, 2018.
 197.Masoumi Z, Familari M, Kallen K, Ranstam J, Olofsson P, Hansson SR. Fetal hemoglobin in umbilical cord blood in preeclamptic and normotensive pregnancies: A cross‐sectional comparative study. PLoS One 12 (4): e0176697, 2017.
 198.Matoth Y, Zaizov R, Varsano I. Postnatal changes in some red cell parameters. Acta Paediatr Scand 60 (3): 317‐323, 1971.
 199.Mayer CA, Wilson CG, MacFarlane PM. Changes in carotid body and nTS neuronal excitability following neonatal sustained and chronic intermittent hypoxia exposure. Respir Physiol Neurobiol 205: 28‐36, 2015.
 200.Mayer CA, Ao J, Di Fiore JM, Martin RJ, MacFarlane PM. Impaired hypoxic ventilatory response following neonatal sustained and subsequent chronic intermittent hypoxia in rats. Respir Physiol Neurobiol 187 (2): 167‐175, 2013.
 201.Mayer CA, Di Fiore JM, Martin RJ, MacFarlane PM. Vulnerability of neonatal respiratory neural control to sustained hypoxia during a uniquely sensitive window of development. J Appl Physiol (1985) 116 (5): 514‐521, 2014.
 202.Milross MA, Piper AJ, Dobbin CJ, Bye PT, Grunstein RR. Sleep disordered breathing in cystic fibrosis. Sleep Med Rev 8 (4): 295‐308, 2004.
 203.Monneret D, Desmurs L, Zaepfel S, Chardon L, Doret‐Dion M, Cartier R. Reference percentiles for paired arterial and venous umbilical cord blood gases: An indirect nonparametric approach. Clin Biochem 67: 40‐47, 2019.
 204.Moore LG. Maternal O2 transport and fetal growth in Colorado, Peru, and Tibet high‐altitude residents. Am J Hum Biol 2 (6): 627‐637, 1990.
 205.Moore LG. Comparative human ventilatory adaptation to high altitude. Respir Physiol 121 (2–3): 257‐276, 2000.
 206.Moore LG, Rounds SS, Jahnigen D, Grover RF, Reeves JT. Infant birth weight is related to maternal arterial oxygenation at high altitude. J Appl Physiol Respir Environ Exerc Physiol 52 (3): 695‐699, 1982.
 207.Moore LG, Jahnigen D, Rounds SS, Reeves JT, Grover RF. Maternal hyperventilation helps preserve arterial oxygenation during high‐altitude pregnancy. J Appl Physiol Respir Environ Exerc Physiol 52 (3): 690‐694, 1982.
 208.Moore PJ, Parkes MJ, Nijhuis JG, Hanson MA. The incidence of breathing movements of fetal sheep in normoxia and hypoxia after peripheral chemodenervation and brain‐stem transection. J Dev Physiol 11 (3): 147‐151, 1989.
 209.Mortola JP. How newborn mammals cope with hypoxia. Respir Physiol 116 (2–3): 95‐103, 1999.
 210.Mortola JP. Implications of hypoxic hypometabolism during mammalian ontogenesis. Respir Physiol Neurobiol 141 (3): 345‐356, 2004.
 211.Mortola JP. Influence of temperature on metabolism and breathing during mammalian ontogenesis. Respir Physiol Neurobiol 149 (1–3): 155‐164, 2005.
 212.Mortola JP, Morgan CA, Virgona V. Respiratory adaptation to chronic hypoxia in newborn rats. J Appl Physiol (1985) 61 (4): 1329‐1336, 1986.
 213.Mortola JP, Rezzonico R, Lanthier C. Ventilation and oxygen consumption during acute hypoxia in newborn mammals: A comparative analysis. Respir Physiol 78 (1): 31‐43, 1989.
 214.Mortola JP, Saiki C. Ventilatory response to hypoxia in rats: Gender differences. Respir Physiol 106 (1): 21‐34, 1996.
 215.Mortola JP, Frappell PB, Frappell DE, Villena‐Cabrera N, Villena‐Cabrera M, Pena F. Ventilation and gaseous metabolism in infants born at high altitude, and their responses to hyperoxia. Am Rev Respir Dis 146 (5 Pt 1): 1206‐1209, 1992.
 216.Mortola JP, Trippenbach T, Rezzonico R, Fisher JT, Diaz M, Villena‐Cabrera N, Pena F. Hering‐Breuer reflexes in high‐altitude infants. Clin Sci (Lond) 88 (3): 345‐350, 1995.
 217.Moss TJ. Respiratory consequences of preterm birth. Clin Exp Pharmacol Physiol 33 (3): 280‐284, 2006.
 218.Moss TJ, Davey MG, McCrabb GJ, Harding R. Development of ventilatory responsiveness to progressive hypoxia and hypercapnia in low‐birth‐weight lambs. J Appl Physiol (1985) 81 (4): 1555‐1561, 1996.
 219.Mostello D, Chalk C, Khoury J, Mack CE, Siddiqi TA, Clark KE. Chronic anemia in pregnant ewes: Maternal and fetal effects. Am J Physiol 261 (5 Pt 2): R1075‐R1083, 1991.
 220.Mouradian GC, Forster HV, Hodges MR. Acute and chronic effects of carotid body denervation (CBD) on ventilation and chemoreflexes in three rat strains. J Physiol 590 (14): 3335‐3347, 2012.
 221.Mouradian GC, Forster HV, Hodges MR. Acute and chronic effects of carotid body denervation on ventilation and chemoreflexes in three rat strains. J Physiol 590 (14): 3335‐3347, 2012.
 222.Mouradian GC Jr, Liu P, Hodges MR. Raphe gene expression changes implicate immune‐related functions in ventilatory plasticity following carotid body denervation in rats. Exp Neurol 287 (Pt 2): 102‐112, 2017.
 223.Mouradian GC Jr, Alvarez‐Argote S, Gorzek R, Thuku G, Michkalkiewicz T, Wong‐Riley MTT, Konduri GG, Hodges MR. Acute and chronic changes in the control of breathing in a rat model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 316 (3): L506‐L518, 2019.
 224.Mouradian GC Jr, Alvarez‐Argote S, Gorzek R, Thuku G, Michalkiewicz T, Wong‐Riley MTT, Konduri GG, Hodges MR. Acute and chronic changes in the control of breathing in a hyperoxia‐induced model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 316 (3): L506‐L518, 2019.
 225.Mu L, Xia DD, Michalkiewicz T, Hodges M, Mouradian G, Konduri GG, Wong‐Riley MTT. Effects of neonatal hyperoxia on the critical period of postnatal development of neurochemical expressions in brain stem respiratory‐related nuclei in the rat. Physiol Rep 6 (5): e13627, 2018.
 226.Mu L, Xia DD, Michalkiewicz T, Konduri GG, Hodges M, Mouradian G, Wong‐Riley MTT. Effects of neonatal hyperoxia on the critical period of postnatal development of neurochemical expressions in brain stem respiratory‐related nuclei in the rat. Physiol Rep 6 (5): e13627, 2018.
 227.Natale R, Nasello‐Paterson C, Connors G. Patterns of fetal breathing activity in the human fetus at 24 to 28 weeks of gestation. Am J Obstet Gynecol 158 (2): 317‐321, 1988.
 228.Niblock MM, Perez A, Broitman S, Jacoby B, Aviv E, Gilkey S. In utero development of fetal breathing movements in C57BL6 mice. Respir Physiol Neurobiol 271: 103288, 2020.
 229.Niermeyer S. Cardiopulmonary transition in the high altitude infant. High Alt Med Biol 4 (2): 225‐239, 2003.
 230.Niermeyer S, Shaffer E, Moore LG. Impaired cardiopulmonary transition at high altitude † 1710. Pediatr Res 43 (4): 292, 1998.
 231.Nock ML, Difiore JM, Arko MK, Martin RJ. Relationship of the ventilatory response to hypoxia with neonatal apnea in preterm infants. J Pediatr 144 (3): 291‐295, 2004.
 232.Okubo S, Mortola JP. Long‐term respiratory effects of neonatal hypoxia in the rat. J Appl Physiol (1985) 64 (3): 952‐958, 1988.
 233.Okubo S, Mortola JP. Control of ventilation in adult rats hypoxic in the neonatal period. Am J Physiol 259 (4 Pt 2): R836‐R841, 1990.
 234.Onimaru H, Dutschmann M. Calcium imaging of neuronal activity in the most rostral parafacial respiratory group of the newborn rat. J Physiol Sci 62 (1): 71‐77, 2012.
 235.Onimaru H, Homma I. Development of the rat respiratory neuron network during the late fetal period. Neurosci Res 42 (3): 209‐218, 2002.
 236.Onimaru H, Homma I. A novel functional neuron group for respiratory rhythm generation in the ventral medulla. J Neurosci 23 (4): 1478‐1486, 2003.
 237.Onimaru H, Homma I. Developmental changes in the spatio‐temporal pattern of respiratory neuron activity in the medulla of late fetal rat. Neuroscience 131 (4): 969‐977, 2005.
 238.Pagliardini S, Ren J, Greer JJ. Ontogeny of the pre‐Botzinger complex in perinatal rats. J Neurosci 23 (29): 9575‐9584, 2003.
 239.Pan J, Yeger H, Cutz E. Innervation of pulmonary neuroendocrine cells and neuroepithelial bodies in developing rabbit lung. J Histochem Cytochem 52 (3): 379‐389, 2004.
 240.Pan J, Bishop T, Ratcliffe PJ, Yeger H, Cutz E. Hyperplasia and hypertrophy of pulmonary neuroepithelial bodies, presumed airway hypoxia sensors, in hypoxia‐inducible factor prolyl hydroxylase‐deficient mice. Hypoxia (Auckl) 4: 69‐80, 2016.
 241.Paterson DS, Trachtenberg FL, Thompson EG, Belliveau RA, Beggs AH, Darnall R, Chadwick AE, Krous HF, Kinney HC. Multiple serotonergic brainstem abnormalities in sudden infant death syndrome. JAMA 296 (17): 2124‐2132, 2006.
 242.Patrick J, Campbell K, Carmichael L, Natale R, Richardson B. Patterns of human fetal breathing during the last 10 weeks of pregnancy. Obstet Gynecol 56 (1): 24‐30, 1980.
 243.Pawar A, Peng YJ, Jacono FJ, Prabhakar NR. Comparative analysis of neonatal and adult rat carotid body responses to chronic intermittent hypoxia. J Appl Physiol (1985) 104 (5): 1287‐1294, 2008.
 244.Peng Y, Cui C, He Y, Ouzhuluobu, Zhang H, Yang D, Zhang Q, Bianbazhuoma, Yang L, He Y, Xiang K, Zhang X, Bhandari S, Shi P, Yangla, Dejiquzong, Baimakangzhuo, Duojizhuoma, Pan Y, Cirenyangji, Baimayangji, Gonggalanzi, Bai C, Bianba, Basang, Ciwangsangbu, Xu S, Chen H, Liu S, Wu T, Qi X, Su B. Down‐regulation of EPAS1 transcription and genetic adaptation of Tibetans to high‐altitude hypoxia. Mol Biol Evol 34 (4): 818‐830, 2017.
 245.Peng YJ, Rennison J, Prabhakar NR. Intermittent hypoxia augments carotid body and ventilatory response to hypoxia in neonatal rat pups. J Appl Physiol (1985) 97 (5): 2020‐2025, 2004.
 246.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 USA 100 (17): 10073‐10078, 2003.
 247.Peng YJ, Yuan G, Jacono FJ, Kumar GK, Prabhakar NR. 5‐HT evokes sensory long‐term facilitation of rodent carotid body via activation of NADPH oxidase. J Physiol 576 (Pt 1): 289‐295, 2006.
 248.Peng YJ, Yuan G, Ramakrishnan D, Sharma SD, Bosch‐Marce M, Kumar GK, Semenza GL, Prabhakar NR. Heterozygous HIF‐1alpha deficiency impairs carotid body‐mediated systemic responses and reactive oxygen species generation in mice exposed to intermittent hypoxia. J Physiol 577 (Pt 2): 705‐716, 2006.
 249.Peng YJ, Nanduri J, Khan SA, Yuan G, Wang N, Kinsman B, Vaddi DR, Kumar GK, Garcia JA, Semenza GL, Prabhakar NR. Hypoxia‐inducible factor 2alpha (HIF‐2alpha) heterozygous‐null mice exhibit exaggerated carotid body sensitivity to hypoxia, breathing instability, and hypertension. Proc Natl Acad Sci USA 108 (7): 3065‐3070, 2011.
 250.Peyronnet J, Roux JC, Geloen A, Tang LQ, Pequignot JM, Lagercrantz H, Dalmaz Y. Prenatal hypoxia impairs the postnatal development of neural and functional chemoafferent pathway in rat. J Physiol 524 (Pt 2): 525‐537, 2000.
 251.Peyronnet J, Roux JC, Mamet J, Perrin D, Lachuer J, Pequignot JM, Dalmaz Y. Developmental plasticity of the carotid chemoafferent pathway in rats that are hypoxic during the prenatal period. Eur J Neurosci 26 (10): 2865‐2872, 2007.
 252.Pike K, Jane Pillow J, Lucas JS. Long term respiratory consequences of intrauterine growth restriction. Semin Fetal Neonatal Med 17 (2): 92‐98, 2012.
 253.Poets CF, Samuels MP, Southall DP. Epidemiology and pathophysiology of apnoea of prematurity. Biol Neonate 65 (3–4): 211‐219, 1994.
 254.Prabhakar NR. O2 sensing at the mammalian carotid body: Why multiple O2 sensors and multiple transmitters? Exp Physiol 91 (1): 17‐23, 2006.
 255.Prabhakar NR, Semenza GL. Oxygen sensing and homeostasis. Physiology 30 (5): 340‐348, 2015.
 256.Prandota J. Possible pathomechanisms of sudden infant death syndrome: Key role of chronic hypoxia, infection/inflammation states, cytokine irregularities, and metabolic trauma in genetically predisposed infants. Am J Ther 11 (6): 517‐546, 2004.
 257.Prsa M, Sun L, van Amerom J, Yoo SJ, Grosse‐Wortmann L, Jaeggi E, Macgowan C, Seed M. Reference ranges of blood flow in the major vessels of the normal human fetal circulation at term by phase‐contrast magnetic resonance imaging. Circ Cardiovasc Imaging 7 (4): 663‐670, 2014.
 258.Rajani V, Zhang Y, Jalubula V, Rancic V, SheikhBahaei S, Zwicker JD, Pagliardini S, Dickson CT, Ballanyi K, Kasparov S, Gourine AV, Funk GD. Release of ATP by pre‐Botzinger complex astrocytes contributes to the hypoxic ventilatory response via a Ca(2+) ‐dependent P2Y1 receptor mechanism. J Physiol 596 (15): 3245‐3269, 2018.
 259.Reeves SR, Gozal D. Changes in ventilatory adaptations associated with long‐term intermittent hypoxia across the age spectrum in the rat. Respir Physiol Neurobiol 150 (2–3): 135‐143, 2006.
 260.Reeves SR, Mitchell GS, Gozal D. Early postnatal chronic intermittent hypoxia modifies hypoxic respiratory responses and long‐term phrenic facilitation in adult rats. Am J Physiol Regul Integr Comp Physiol 290 (6): R1664‐R1671, 2006.
 261.Ren J, Greer JJ. Modulation of respiratory rhythmogenesis by chloride‐mediated conductances during the perinatal period. J Neurosci 26 (14): 3721‐3730, 2006.
 262.Ribeiro AP, Mayer CA, Wilson CG, Martin RJ, MacFarlane PM. Intratracheal LPS administration attenuates the acute hypoxic ventilatory response: Role of brainstem IL‐1beta receptors. Respir Physiol Neurobiol 242: 45‐51, 2017.
 263.Richardson BS, Bocking AD. Metabolic and circulatory adaptations to chronic hypoxia in the fetus. Comp Biochem Physiol A Mol Integr Physiol 119 (3): 717‐723, 1998.
 264.Richardson HL, Parslow PM, Walker AM, Harding R, Horne RS. Maturation of the initial ventilatory response to hypoxia in sleeping infants. J Sleep Res 16 (1): 117‐127, 2007.
 265.Rojas DA, Perez‐Munizaga DA, Centanin L, Antonelli M, Wappner P, Allende ML, Reyes AE. Cloning of hif‐1alpha and hif‐2alpha and mRNA expression pattern during development in zebrafish. Gene Expr Patterns 7 (3): 339‐345, 2007.
 266.Rosenfeld M, Snyder FF. Direct observation of intrauterine respiratory movements of the fetus and the role of carbon dioxide and oxygen in their regulation. Am J Physiol 119 (1): 153‐166, 1937.
 267.Rourke KS, Mayer CA, MacFarlane PM. A critical postnatal period of heightened vulnerability to lipopolysaccharide. Respir Physiol Neurobiol 232: 26‐34, 2016.
 268.Rudolph AM. Congenital Diseases of the Heart: Clinical‐Physiological Considerations (3rd ed). Chichester, UK; Hoboken, NJ: Wiley‐Blackwell, 2009. 538 p.
 269.Ryan HE, Lo J, Johnson RS. HIF‐1α is required for solid tumor formation and embryonic vascularization. EMBO J 17 (11): 3005‐3015, 1998.
 270.Rytkonen KT, Williams TA, Renshaw GM, Primmer CR, Nikinmaa M. Molecular evolution of the metazoan PHD‐HIF oxygen‐sensing system. Mol Biol Evol 28 (6): 1913‐1926, 2011.
 271.Sankaran VG, Orkin SH. The switch from fetal to adult hemoglobin. Cold Spring Harb Perspect Med 3 (1): a011643, 2013.
 272.Sawnani H, Jackson T, Murphy T, Beckerman R, Simakajornboon N. The effect of maternal smoking on respiratory and arousal patterns in preterm infants during sleep. Am J Respir Crit Care Med 169 (6): 733‐738, 2004.
 273.Schechter AN. Hemoglobin research and the origins of molecular medicine. Blood 112 (10): 3927‐3938, 2008.
 274.Schmalisch G, Wilitzki S, Wauer RR. Differences in tidal breathing between infants with chronic lung diseases and healthy controls. BMC Pediatr 5: 36, 2005.
 275.Schneider‐Maunoury S, Topilko P, Seitandou T, Levi G, Cohen‐Tannoudji M, Pournin S, Babinet C, Charnay P. Disruption of Krox‐20 results in alteration of rhombomeres 3 and 5 in the developing hindbrain. Cell 75 (6): 1199‐1214, 1993.
 276.Serra A, Brozoski D, Hedin N, Franciosi R, Forster HV. Mortality after carotid body denervation in rats. J Appl Physiol 91 (3): 1298‐1306, 2001.
 277.Sheikhbahaei S, Turovsky EA, Hosford PS, Hadjihambi A, Theparambil SM, Liu B, Marina N, Teschemacher AG, Kasparov S, Smith JC, Gourine AV. Astrocytes modulate brainstem respiratory rhythm‐generating circuits and determine exercise capacity. Nat Commun 9 (1): 370, 2018.
 278.Siljehav V, Shvarev Y, Herlenius E. Il‐1beta and prostaglandin E2 attenuate the hypercapnic as well as the hypoxic respiratory response via prostaglandin E receptor type 3 in neonatal mice. J Appl Physiol (1985) 117 (9): 1027‐1036, 2014.
 279.Sladek M, Parker RA, Grogaard JB, Sundell HW. Long‐lasting effect of prolonged hypoxemia after birth on the immediate ventilatory response to changes in arterial partial pressure of oxygen in young lambs. Pediatr Res 34 (6): 821‐828, 1993.
 280.Smith CA, Chenuel BJ, Henderson KS, Dempsey JA. The apneic threshold during non‐REM sleep in dogs: Sensitivity of carotid body vs. central chemoreceptors. J Appl Physiol (1985) 103 (2): 578‐586, 2007.
 281.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 (5032): 726‐729, 1991.
 282.Smith JC, Abdala AP, Borgmann A, Rybak IA, Paton JF. Brainstem respiratory networks: Building blocks and microcircuits. Trends Neurosci 36 (3): 152‐162, 2013.
 283.Soothill PW, Nicolaides KH, Rodeck CH, Campbell S. Effect of gestational age on fetal and intervillous blood gas and acid‐base values in human pregnancy. Fetal Ther 1 (4): 168‐175, 1986.
 284.St‐John WM, Leiter JC. Maintenance of gasping and restoration of eupnea after hypoxia is impaired following blockers of alpha1‐adrenergic receptors and serotonin 5‐HT2 receptors. J Appl Physiol (1985) 104 (3): 665‐673, 2008.
 285.St‐John WM, Li A, Leiter JC. Genesis of gasping is independent of levels of serotonin in the Pet‐1 knockout mouse. J Appl Physiol (1985) 107 (3): 679‐685, 2009.
 286.Stojanovska V, Miller SL, Hooper SB, Polglase GR. The consequences of preterm birth and chorioamnionitis on brainstem respiratory centers: Implications for neurochemical development and altered functions by inflammation and prostaglandins. Front Cell Neurosci 12: 26, 2018.
 287.Sui P, Wiesner DL, Xu J, Zhang Y, Lee J, Van Dyken S, Lashua A, Yu C, Klein BS, Locksley RM, Deutsch G, Sun X. Pulmonary neuroendocrine cells amplify allergic asthma responses. Science 360 (6393): eaan8546, 2018.
 288.SUPPORT Study Group of the Eunice Kennedy Shriver NICHD Neonatal Research Network. Target ranges of oxygen saturation in extremely preterm infants. N Engl J Med 362 (21): 1959‐1969, 2010.
 289.Tadjalli A, Mitchell GS. Cervical spinal 5‐HT2A and 5‐HT2B receptors are both necessary for moderate acute intermittent hypoxia‐induced phrenic long‐term facilitation. J Appl Physiol (1985) 127 (2): 432‐443, 2019.
 290.Taweevisit M, Theerasantipong B, Taothong K, Thorner PS. Pulmonary neuroendocrine cell hyperplasia in hemoglobin Bart‐induced hydrops fetalis: A model for chronic intrauterine hypoxia. Pediatr Dev Pathol 20 (4): 298‐307, 2017.
 291.Teitel DF, Iwamoto HS, Rudolph AM. Changes in the pulmonary circulation during birth‐related events. Pediatr Res 27 (4 Pt 1): 372‐378, 1990.
 292.Teppema LJ. CrossTalk opposing view: The hypoxic ventilatory response does not include a central, excitatory hypoxia sensing component. J Physiol 596 (15): 2939‐2941, 2018.
 293.Teppema LJ, Dahan A. The ventilatory response to hypoxia in mammals: Mechanisms, measurement, and analysis. Physiol Rev 90 (2): 675‐754, 2010.
 294.Tester NJ, Fuller DD, Fromm JS, Spiess MR, Behrman AL, Mateika JH. Long‐term facilitation of ventilation in humans with chronic spinal cord injury. Am J Respir Crit Care Med 189 (1): 57‐65, 2014.
 295.Thoby‐Brisson M, Karlen M, Wu N, Charnay P, Champagnat J, Fortin G. Genetic identification of an embryonic parafacial oscillator coupling to the preBotzinger complex. Nat Neurosci 12 (8): 1028‐1035, 2009.
 296.Tolcos M, Rees S. Chronic placental insufficiency in the fetal guinea pig affects neurochemical and neuroglial development but not neuronal numbers in the brainstem: A new method for combined stereology and immunohistochemistry. J Comp Neurol 379 (1): 99‐112, 1997.
 297.Tolcos M, Harding R, Loeliger M, Breen S, Cock M, Duncan J, Rees S. The fetal brainstem is relatively spared from injury following intrauterine hypoxemia. Brain Res Dev Brain Res 143 (1): 73‐81, 2003.
 298.Toppin VA, Harris MB, Kober AM, Leiter JC, St‐John WM. Persistence of eupnea and gasping following blockade of both serotonin type 1 and 2 receptors in the in situ juvenile rat preparation. J Appl Physiol (1985) 103 (1): 220‐227, 2007.
 299.Tree K, Viemari JC, Cayetanot F, Peyronnet J. Growth restriction induced by chronic prenatal hypoxia affects breathing rhythm and its pontine catecholaminergic modulation. J Neurophysiol 116 (4): 1654‐1662, 2016.
 300.Trudinger BJ, Knight PC. Fetal age and patterns of human fetal breathing movements. Am J Obstet Gynecol 137 (6): 724‐728, 1980.
 301.Tryba AK, Pena F, Ramirez JM. Gasping activity in vitro: A rhythm dependent on 5‐HT2A receptors. J Neurosci 26 (10): 2623‐2634, 2006.
 302.Unger C, Weiser JK, McCullough RE, Keefer S, Moore LG. Altitude, low birth weight, and infant mortality in Colorado. JAMA 259 (23): 3427‐3432, 1988.
 303.Vargas M, Vargas E, Julian CG, Armaza JF, Rodriguez A, Tellez W, Niermeyer S, Wilson M, Parra E, Shriver M, Moore LG. Determinants of blood oxygenation during pregnancy in Andean and European residents of high altitude. Am J Physiol Regul Integr Comp Physiol 293 (3): R1303‐R1312, 2007.
 304.Villa MP, Pagani J, Lucidi V, Palamides S, Ronchetti R. Nocturnal oximetry in infants with cystic fibrosis. Arch Dis Child 84 (1): 50‐54, 2001.
 305.Waites BA, Ackland GL, Noble R, Hanson MA. Red nucleus lesions abolish the biphasic respiratory response to isocapnic hypoxia in decerebrate young rabbits. J Physiol 495 (Pt 1): 217‐225, 1996.
 306.Watson CS, Schaefer R, White SE, Homan JH, Fraher L, Harding R, Bocking AD. Effect of intermittent umbilical cord occlusion on fetal respiratory activity and brain adenosine in late‐gestation sheep. Reprod Fertil Dev 14 (1–2): 35‐42, 2002.
 307.Weaver DR. A2a adenosine receptor gene expression in developing rat brain. Brain Res Mol Brain Res 20 (4): 313‐327, 1993.
 308.Wigglesworth JS, Desai R. Effect on lung growth of cervical cord section in the rabbit fetus. Early Hum Dev 3 (1): 51‐65, 1979.
 309.Williams PA, Wilson CG. Effects of inflammation on the developing respiratory system: Focus on hypoglossal (XII) neuron morphology, brainstem neurochemistry, and control of breathing. Respir Physiol Neurobiol 275: 103389, 2020.
 310.Wilson SL, Thach BT, Brouillette RT, Abu‐Osba YK. Upper airway patency in the human infant: Influence of airway pressure and posture. J Appl Physiol Respir Environ Exerc Physiol 48 (3): 500‐504, 1980.
 311.Wong‐Riley MT, Liu Q. Neurochemical and physiological correlates of a critical period of respiratory development in the rat. Respir Physiol Neurobiol 164 (1–2): 28‐37, 2008.
 312.Wong‐Riley MTT, Liu Q, Gao X. Mechanisms underlying a critical period of respiratory development in the rat. Respir Physiol Neurobiol 264: 40‐50, 2019.
 313.Wyatt CN, Wright C, Bee D, Peers C. O2‐sensitive K+ currents in carotid body chemoreceptor cells from normoxic and chronically hypoxic rats and their roles in hypoxic chemotransduction. Proc Natl Acad Sci USA 92 (1): 295‐299, 1995.
 314.Yan X, Koos BJ, Kruger L, Linden J, Murray TF. Characterization of [125I]ZM 241385 binding to adenosine A2A receptors in the pineal of sheep brain. Brain Res 1096 (1): 30‐39, 2006.
 315.Yang HT, Cummings KJ. Brain stem serotonin protects blood pressure in neonatal rats exposed to episodic anoxia. J Appl Physiol (1985) 115 (12): 1733‐1741, 2013.
 316.Young JO, Geurts A, Hodges MR, Cummings KJ. Active sleep unmasks apnea and delayed arousal in infant rat pups lacking central serotonin. J Appl Physiol (1985) 123 (4): 825‐834, 2017.
 317.Youngson C, Nurse C, Yeger H, Cutz E. Oxygen sensing in airway chemoreceptors. Nature 365 (6442): 153‐155, 1993.
 318.Zamudio S, Postigo L, Illsley NP, Rodriguez C, Heredia G, Brimacombe M, Echalar L, Torricos T, Tellez W, Maldonado I, Balanza E, Alvarez T, Ameller J, Vargas E. Maternal oxygen delivery is not related to altitude‐ and ancestry‐associated differences in human fetal growth. J Physiol 582 (Pt 2): 883‐895, 2007.

Contact Editor

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

* Required Field

How to Cite

Gary C. Mouradian, Satyan Lakshminrusimha, Girija G. Konduri. Perinatal Hypoxemia and Oxygen Sensing. Compr Physiol 2021, 11: 1653-1677. doi: 10.1002/cphy.c190046