Comprehensive Physiology Wiley Online Library

Regulation of Breathing and Autonomic Outflows by Chemoreceptors

Full Article on Wiley Online Library



Abstract

Lung ventilation fluctuates widely with behavior but arterial PCO2 remains stable. Under normal conditions, the chemoreflexes contribute to PaCO2 stability by producing small corrective cardiorespiratory adjustments mediated by lower brainstem circuits. Carotid body (CB) information reaches the respiratory pattern generator (RPG) via nucleus solitarius (NTS) glutamatergic neurons which also target rostral ventrolateral medulla (RVLM) presympathetic neurons thereby raising sympathetic nerve activity (SNA). Chemoreceptors also regulate presympathetic neurons and cardiovagal preganglionic neurons indirectly via inputs from the RPG. Secondary effects of chemoreceptors on the autonomic outflows result from changes in lung stretch afferent and baroreceptor activity. Central respiratory chemosensitivity is caused by direct effects of acid on neurons and indirect effects of CO2 via astrocytes. Central respiratory chemoreceptors are not definitively identified but the retrotrapezoid nucleus (RTN) is a particularly strong candidate. The absence of RTN likely causes severe central apneas in congenital central hypoventilation syndrome. Like other stressors, intense chemosensory stimuli produce arousal and activate circuits that are wake‐ or attention‐promoting. Such pathways (e.g., locus coeruleus, raphe, and orexin system) modulate the chemoreflexes in a state‐dependent manner and their activation by strong chemosensory stimuli intensifies these reflexes. In essential hypertension, obstructive sleep apnea and congestive heart failure, chronically elevated CB afferent activity contributes to raising SNA but breathing is unchanged or becomes periodic (severe CHF). Extreme CNS hypoxia produces a stereotyped cardiorespiratory response (gasping, increased SNA). The effects of these various pathologies on brainstem cardiorespiratory networks are discussed, special consideration being given to the interactions between central and peripheral chemoreflexes. © 2014 American Physiological Society. Compr Physiol 4:1511‐1562, 2014.

Figure 1. Figure 1. Organigram of the chemoreflexes. Cascade of cardiorespiratory responses elicited in anesthetized mammals by hypoxic stimulation of the carotid bodies or by hypercapnia. These cardiorespiratory responses are elaborated primarily within spinal and pontomedullary circuits. The same circuits are also presumably recruited by small perturbations of the blood gases in the conscious state to stabilize PCO2. Large acute perturbations of blood gases produce arousal, aversive sensations and stress, responses that involve numerous other brain regions and processes. The direct effects of hypoxia on the CNS are not considered here. Green arrows denote cell activation (e.g., carotid bodies by hypoxia) or a globally excitatory connection (e.g., effect of the carotid bodies, CBs, on the RPG), or an increase in a dependent variable [e.g., effect of CO2 on cerebral blood flow (CBF) resulting in a “washout” of brain CO2]. Red arrows have the opposite meaning. The baroreflex (BaroR) potentiates or attenuates the chemoreflexes depending on the direction of the change in arterial pressure (AP). Slowly adapting lung stretch receptors (SARs) exert a feedback regulation on the RPG and on central chemoreceptors (CCRs) and inhibit the cardiovagal outflow (CVPSN, cardiovagal parasympathetic nerve activity). The chemoreceptors, both central and peripheral, activate the sympathetic nervous system (SNS) both via the RPG and independently of it.
Figure 2. Figure 2. Pontomedullary regions responsible for eupneic breathing and for generating the autonomic outflows to the cardiovascular system: anatomy and simplified circuitry. (A1) Parasagittal section through the pons and medulla oblongata of a rodent. The regions colored in magenta contain the principal building blocks of the respiratory pattern generator. The ventral respiratory column (VRC) contains four functional compartments aligned in rostrocaudal order (Bötzinger Complex (BötC), pre‐BötC, rostral ventral respiratory group (rVRG) and caudal VRG (cVRG)). The retrotrapezoid nucleus (RTN) resides at the rostral end of the VRC under the facial motor nucleus. In this article, the term RTN refers specifically to a cluster of about 2000 CO2‐activated Phox2b‐ir glutamatergic neurons (in rats, 800 in mice). (A2) Minimal circuitry responsible for the generation of eupneic breathing [adapted, with permission, from Lindsey, Ryback & Smith (247)]. The drawing depicts some of the neuronal interconnections within and between the four compartments of the ventral respiratory column and a few of the connections of RTN neurons (for details see text). The parafacial respiratory group (pfRG) is a physiologically defined entity now believed to be specifically involved in the generation of active expiration (331). Its constituent neurons and their location are not yet defined. Bötzinger augmenting expiratory neurons have been included by some authors in the pfRG (328). Inhibitory (GABAergic or glycinergic) neurons are represented in red, glutamatergic neurons in green, motoneurons in blue, connections with both excitatory and inhibitory components in magenta (e.g., neurons transmitting information from arterial baroreceptors, pulmonary stretch afferents, the carotid bodies etc.). (B1) The regions colored in magenta are thought to contain the main components of the network that generates the autonomic outflows to the cardiovascular system. From an autonomic regulation standpoint, the ventrolateral medulla can be subdivided into three regions whose anatomical relationship with the respiratory compartments can be appreciated by comparing panels A1 and B1. (B2) Schematic of cardiovagal parasympathetic neurons, RVLM presympathetic neurons and connections responsible for their regulation by arterial baroreceptors. Abbreviations: aug‐E, augmenting expiratory neurons; aug‐I, augmenting inspiratory neurons (a.k.a. inspiratory premotor neurons); CVLM, caudal VLM; DRG, dorsal respiratory group (caudolateral portion of the NTS); early‐I, early‐inspiratory neurons, early‐I(1) and early‐I(2) are postulated to have distinct input‐output functions; IVLM, intermediate VLM; Itr, intertrigeminal region; KF, Kölliker‐Fuse nucleus; LPBN, lateral parabrachial nuclei; LRt, lateral reticular nucleus; Mo5, trigeminal motor nucleus; NTS, nucleus of the solitary tract; pfRG, parafacial respiratory group; Pn, pontine nuclei; post‐I, postinspiratory; preI/I, preinspiratory/inspiratory (putative rhythmogenic neurons); scp, superior cerebellar peduncle; SO, superior olive; SPGN, sympathetic preganglionic neurons; SPN sympathetic (post)ganglionic neuron; VRC, ventral respiratory column.
Figure 3. Figure 3. Regulation of the sympathetic vasomotor outflow by the respiratory pattern generator. (A) Plausible circuitry responsible for coupling respiration with the sympathetic nerve activity to the cardiovascular system. Sympathetic ganglionic neurons (SPNs) display stereotyped patterns of respiratory modulation in deafferented preparations (vagotomized, barodenervated). Two of the most commonly found patterns in rats (early‐I and post‐I) are depicted in D [top traces, average integrated phrenic nerve discharge, iPND; lower trace iPND‐triggered rate histogram depicting the probability of firing of a single SPN during the central respiratory cycle; redrawn, with permission, after (72)]. These SPN patterns are virtually identical to those of single RVLM presympathetic neurons [shown in B; redrawn, with permission, from (168)]. The IVLM contains GABAergic neurons that are activated by arterial baroreceptor stimulation and inhibit RVLM presympathetic neurons (panel A). The respiratory modulation of these IVLM GABAergic neurons is approximately the mirror image of that of the RVLM presympathetic neurons [C; reprinted from Mandel and Schreihofer (266) with permission from Wiley & Sons] hence the hypothesis that IVLM neurons with early‐inspiratory discharge produce post‐I modulation in RVLM presympathetic neurons and vice versa (red arrows in C). The depicted inputs from the RPG to IVLM neurons are plausible but speculative. (E) Intracellular recordings of RVLM presympathetic neurons in an arterially perfused midcollicular transected rat preparation [from Moraes et al. (294) with permission]. The early‐I (left trace) and the post‐I patterns (middle trace) are instantly recognizable. The right panel shows a presympathetic RVLM neuron that exhibits an extra peak of activity during late expiration. The latter recording was made in a rat subjected to chronic intermittent hypoxia which caused late‐expiratory activity in an abdominal nerve at rest (not shown) and an additional late‐E peak of activity in the thoracic chain (red arrow pointing down and to the right above the thoracic chain SNA trace, tSNA). An excitatory input from pfRG has been tentatively proposed as the source of this late‐expiratory activation (illustrated in A).
Figure 4. Figure 4. Gain and loss of function of RTN Phox2b neurons in conscious rodents: effect on breathing and the hypercapnic ventilatory reflex (HCVR). (A) Steady‐state ventilatory response of a conscious rat exposed to graded levels of hypercapnia. Breathing was measured by whole body plethysmography [reprinted from (236) with permission from Elsevier]. Note that, at 8% FiCO2, fR (respiratory rate) doubles and VE triples. (B) Unilateral acidification of the RTN with a dialysis probe containing a fluid equilibrated with 25% CO2 produces a small (24% average) increase in VE [adapted from Li et al. J. Appl. Physiol. (240) with permission]. (C) Unilateral optogenetic activation (20Hz) of around 35% of RTN Phox2b neurons triples VE in a conscious rat [adapted, with permission, from (4)]. (C1) Typical example (plethysmography); (C2‐C4) average responses (from top to bottom: frequency, tidal volume and minute volume) to unilateral optogenetic stimulation of RTN neurons. (C5) Selective expression of ChR2‐mCherry fusion protein by RTN (i.e., Phox2b+) neurons in one such animal (Phox2b in green; mCherry in red). (D1) Attenuation of the hypercapnic ventilatory reflex by i.c.v. allatostatin administration to a conscious rat in which an unknown fraction of RTN neurons were transduced with the allatostatin receptor. (D2) Average results from five rats [(from Marina et al. (270) with permission]. (E1) Absence of HCVR in neonatal (day 9) transgenic mice in which a mutated form of transcription factor Phox2b (Phox2b‐27ala) is expressed selectively by cells of rhombomere 3 and 5 lineage (Phox2b27Alacki; Egr2cre/0) preventing or aborting RTN neuron development. (E2) Group data. (E3) Partial recovery (one third) of the HCVR in adult Phox2b27Alacki; Egr2cre/0 mice. (E4) Absence of Phox2b‐ir neurons in the RTN of a Phox2b27Alacki; Egr2cre/0 mouse (embryonic day 14; RTN identified by white oval, Phox2b in red) [E1‐E4 reprinted from Ramanantsoa et al. (347) with permission].
Figure 5. Figure 5. Gain and loss of function of RTN Phox2b neurons in conscious rodents: effect on breathing and the hypercapnic ventilatory reflex (HCVR). (A) Steady‐state ventilatory response of a conscious rat exposed to graded levels of hypercapnia. Breathing was measured by whole body plethysmography [reprinted from (236) with permission from Elsevier]. Note that, at 8% FiCO2, fR (respiratory rate) doubles and VE triples. (B) Unilateral acidification of the RTN with a dialysis probe containing a fluid equilibrated with 25% CO2 produces a small (24% average) increase in VE [adapted from Li et al. J. Appl. Physiol. (240) with permission]. (C) Unilateral optogenetic activation (20Hz) of around 35% of RTN Phox2b neurons triples VE in a conscious rat [adapted, with permission, from (4)]. (C1) Typical example (plethysmography); (C2‐C4) average responses (from top to bottom: frequency, tidal volume and minute volume) to unilateral optogenetic stimulation of RTN neurons. (C5) Selective expression of ChR2‐mCherry fusion protein by RTN (i.e., Phox2b+) neurons in one such animal (Phox2b in green; mCherry in red). (D1) Attenuation of the hypercapnic ventilatory reflex by i.c.v. allatostatin administration to a conscious rat in which an unknown fraction of RTN neurons were transduced with the allatostatin receptor. (D2) Average results from five rats [(from Marina et al. (270) with permission]. (E1) Absence of HCVR in neonatal (day 9) transgenic mice in which a mutated form of transcription factor Phox2b (Phox2b‐27ala) is expressed selectively by cells of rhombomere 3 and 5 lineage (Phox2b27Alacki; Egr2cre/0) preventing or aborting RTN neuron development. (E2) Group data. (E3) Partial recovery (one third) of the HCVR in adult Phox2b27Alacki; Egr2cre/0 mice. (E4) Absence of Phox2b‐ir neurons in the RTN of a Phox2b27Alacki; Egr2cre/0 mouse (embryonic day 14; RTN identified by white oval, Phox2b in red) [E1‐E4 reprinted from Ramanantsoa et al. (347) with permission].
Figure 6. Figure 6. Activation of RTN neurons by CO2. (A) Location and anatomical projections of RTN neurons (definition of RTN in Fig. 2 legend; abbreviations as in Fig. 2). (B) Transverse section through the lower right quadrant of the rat medulla oblongata at the level of the dotted line in A. The facial motor nucleus is in blue (choline acetyl‐transferase immunoreactivity), the C1 presympathetic neurons are in red (tyrosine hydroxylase) and phox2b immunoreactivity (green, nuclear localization) identifies RTN neurons [adapted, with permission, from (402)]. (C) Response of an RTN neuron to graded hypercapnia in an anesthetized rat (end‐expiratory CO2 shown in top trace) [adapted, with permission, from (402)]. (D) Example of one RTN neuron excited by brief hypoxia (carotid body stimulation, bottom trace) and by hypercapnia (end‐expiratory CO2 in green). After i.c.v. administration of the broad spectrum glutamatergic blocker kynurenic acid, the cell no longer responds to hypoxia but its response to hypercapnia is unaffected [adapted, with permission, from (301)]. (E) RTN CO2‐activated neuron labeled juxtacellularly with biotinamide in vivo (green fluorescence) has a Phox2b‐ir nucleus [adapted, with permission, from (402)]. (F) Structure of an RTN neuron whose cell body was located at the ventral surface of the medulla oblongata (in vivo recording, juxtacellular labeling with biotinamide, and transverse plane projection). Note the extensive dendrites on the ventral surface [adapted, with permission, from (301)]. (G1) Two intracellularly labeled RTN neurons recorded in a Phox2b‐eGFP transgenic mouse coronal slice (green eGFP, red biotinamide). (G2) Representative acid sensitivity of one such neuron recorded at room temperature (cell attached recording, integrated rate histogram, 10 s bin) [adapted, with permission, from (231)]. (H1) RTN Phox2b+ neuron acutely isolated from a Phox2b‐eGFP mouse. (H2) Acid sensitivity of such an acutely isolated RTN neuron. [adapted, with permission, from (447)]. Note that the cell responds to a change in pH, not to CO2 per se. I1, selective expression of TASK‐2 potassium channels by Phox2b+ RTN neurons in mice [adapted from Gestreau et al. (137) with permission]. Left panel shows TASK‐2 expression monitored with LacZ reaction product. Middle panel shows that TASK‐2 expressing neurons are eliminated in a mouse line in which RTN neurons express the 27‐ala Phox2b mutation and fail to develop (Task2+/−; Phox2b27Ala/+ ). Right panel: Task‐2 expressing neurons from a Task2+/‐ mouse (ventral surface view) showing the superficial location in perfect register with the RTN. I2, reduced acid sensitivity of RTN neurons in Task‐2 knock‐out mice compared to Task‐2 +/− control mice [adapted, with permission, from (446)].
Figure 7. Figure 7. Serotonergic neurons and breathing. (A) Selective expression of ChR2‐mCherry fusion protein by raphe obscurus serotonergic neurons in an e‐PET Cre mouse [adapted, with permission, after (86)]. (B) Photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in such a mouse (ketamine/dexmedetomidine initial anesthesia with 0.3% to 0.6% isoflurane supplementation) increases diaphragm EMG amplitude (top trace) and respiratory rate (fR, second from top). Bottom traces shows a single raphe serotonergic unit being photoactivated every time the laser light was pulsed [adapted, with permission, after (86)]. (C) Selective optogenetic photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in a conscious mouse (whole body plethysmography). The response has the same slow ON‐slow OFF kinetics as in the anesthetized mouse [adapted, with permission, after (155)]. (D) Robust acid sensitivity of a serotonergic neuron in culture [reprinted from Richerson (354)] adapted with permission from Macmillan Publishers Ltd., Nature Neuroscience (5), 2004). (E1) In an arterially perfused rat preparation, serotonergic neurons are either modestly inhibited or modestly excited by CO2. (E2) In this preparation, hypercapnia had no effect on the mean activity of the serotonergic population at large (all 5‐HT) [from Iceman et al. (188) with permission]. (F) Putative serotonergic neuron recorded in conscious cats showing a modest but dose‐dependent increase in discharge rate (2.7 to 3.9 Hz; +44% at 6% FiCO2). The effect of CO2 essentially disappeared during non‐REM sleep [reproduced from Veasey et al. (441) with permission]. (G) Serotonin2A receptor antagonism dramatically slows the pre‐Bötzinger complex rhythm in a slice (top traces, mass activity of pre‐Bötzinger complex neurons, bottom traces whole cell recording of a presumed rhythmogenic neuron) [reproduced from Peña and Ramirez (338) with permission]. (H1‐2) Iontophoretic application of serotonin activates RTN neurons by a constant amount regardless of the level of end‐expiratory CO2 [H1, representative example, H2 average results; adapted, with permission, from (300)]. (I) Selective global inhibition of CNS serotonergic neurons (DREADD methodology) attenuates the hypercapnic ventilatory reflex of a conscious mouse [after Ray et al. (350) reprinted with permission from AAAS]. (J) Interpretations: serotonergic neurons activate breathing via innumerable mechanisms (e.g., direct projection to RTN, the RPG, dorsolateral pons, motoneurons, and indirect effects via changes in vigilance). The integrity of serotonergic neurons is required for full expression of the hypercapnic ventilatory reflex, possibly because a fraction of serotonergic neuron is directly responsive to acidification in vivo.
Figure 8. Figure 8. Serotonergic neurons and breathing. (A) Selective expression of ChR2‐mCherry fusion protein by raphe obscurus serotonergic neurons in an e‐PET Cre mouse [adapted, with permission, after (86)]. (B) Photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in such a mouse (ketamine/dexmedetomidine initial anesthesia with 0.3% to 0.6% isoflurane supplementation) increases diaphragm EMG amplitude (top trace) and respiratory rate (fR, second from top). Bottom traces shows a single raphe serotonergic unit being photoactivated every time the laser light was pulsed [adapted, with permission, after (86)]. (C) Selective optogenetic photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in a conscious mouse (whole body plethysmography). The response has the same slow ON‐slow OFF kinetics as in the anesthetized mouse [adapted, with permission, after (155)]. (D) Robust acid sensitivity of a serotonergic neuron in culture [reprinted from Richerson (354)] adapted with permission from Macmillan Publishers Ltd., Nature Neuroscience (5), 2004). (E1) In an arterially perfused rat preparation, serotonergic neurons are either modestly inhibited or modestly excited by CO2. (E2) In this preparation, hypercapnia had no effect on the mean activity of the serotonergic population at large (all 5‐HT) [from Iceman et al. (188) with permission]. (F) Putative serotonergic neuron recorded in conscious cats showing a modest but dose‐dependent increase in discharge rate (2.7 to 3.9 Hz; +44% at 6% FiCO2). The effect of CO2 essentially disappeared during non‐REM sleep [reproduced from Veasey et al. (441) with permission]. (G) Serotonin2A receptor antagonism dramatically slows the pre‐Bötzinger complex rhythm in a slice (top traces, mass activity of pre‐Bötzinger complex neurons, bottom traces whole cell recording of a presumed rhythmogenic neuron) [reproduced from Peña and Ramirez (338) with permission]. (H1‐2) Iontophoretic application of serotonin activates RTN neurons by a constant amount regardless of the level of end‐expiratory CO2 [H1, representative example, H2 average results; adapted, with permission, from (300)]. (I) Selective global inhibition of CNS serotonergic neurons (DREADD methodology) attenuates the hypercapnic ventilatory reflex of a conscious mouse [after Ray et al. (350) reprinted with permission from AAAS]. (J) Interpretations: serotonergic neurons activate breathing via innumerable mechanisms (e.g., direct projection to RTN, the RPG, dorsolateral pons, motoneurons, and indirect effects via changes in vigilance). The integrity of serotonergic neurons is required for full expression of the hypercapnic ventilatory reflex, possibly because a fraction of serotonergic neuron is directly responsive to acidification in vivo.
Figure 9. Figure 9. Glial cells and central respiratory chemosensitivity. (A) Acidification increases intracellular calcium fluorescence in medullary astrocytes [from Gourine et al. (146) reprinted with permission from AAAS]. (B) Optogenetic depolarization of ChR2‐expressing ventrolateral medullary astrocytes activates the phrenic outflow in an anesthetized rat. Note the very slow onset of the response to light and its persistence [adapted, with permission, from (146) reprinted with permission from AAAS]. (C) Activation of RTN neurons by CO2 in a slice of neonate rat brain is attenuated by application of the P2 receptor antagonists PPADS or suramin. Summary data (right panels): about a quarter of the overall response of RTN neurons to 15% CO2 could be mediated by ATP release in this preparation [reproduced from Wenker et al. (454) with permission]. (D) Schema of the contribution of ventral medullary surface (VMS) astrocytes to central respiratory chemosensitivity according to (145) [after Gourine and Kasparov, Exp. Phsiol. (96), 2011 reprinted with permission from John Wiley & Sons]. VMS astrocytes are thought to mediate the hypercapnic ventilatory reflex by simultaneously activating RTN neurons and undefined components of the central pattern generating (CPG). ATP release by astrocytes may be a calcium‐dependent exocytotic process triggered by intracellular acidification and/or a leak through connexin channels (Cx26 primarily) opened by molecular CO2 via carbamylation.
Figure 10. Figure 10. Sympathetic nerve activation by carotid body stimulation: lower brainstem pathways. The four experiments depicted around the interpretative schematic (A‐D, clockwise from top right) suggest that, in anesthetized rodents, carotid body stimulation increases sympathetic vasomotor tone via two pathways that converge on RVLM presympathetic neurons, only one of which depends on the respiratory pattern generator. (A) Microinjection of the GABA mimetic agonist muscimol into the commissural portion of the NTS abolishes the response to cyanide [adapted, with permission, from (295)]. (B) Injection of muscimol into the rVRG eliminates the phrenic nerve discharge but does not change the sympathetic response to carotid body activation (N2, 10 s nitrogen inhalation) [adapted, with permission, from (217)]. (C) Injection of muscimol into the pre‐Bötzinger complex, eliminates the phrenic nerve discharge and eliminates the respiratory oscillations of the sympathetic nerve discharge elicited by carotid body stimulation. However, the sympathoexcitation produced by carotid body stimulation persists. The response of a simultaneously recorded single RVLM presympathetic neuron is also shown. Note that muscimol produces the same effect on the neuron as on SNA, that is, muscimol changes the response from respiratory synchronous oscillations to a tonic activation [adapted, with permission, from (216)]. (D) Administration of kynurenic acid into the RVLM dramatically reduces SNA activation caused by carotid body stimulation [after (295) reprinted with permission from the Society for Neuroscience]. The respiratory dependent pathway of the chemoreflex is considered in greater detail in Fig. 3).
Figure 11. Figure 11. Regulation of the cardiovagal parasympathetic outflow by the respiratory pattern generator. (A1‐3) [Reproduced from McAllen et al. (278), reprinted with permission.] Intracellular recordings of cardiovagal ganglionic neurons from an arterially perfused rat preparation in which the lungs are not inflated therefore reflexes from lung stretch receptors are absent (A1). The firing properties of cardiovagal preganglionic neurons (CVPGN) were inferred from the discharge pattern of the ganglionic neurons (A2) or the EPSPs recorded from these neurons (A3). Activation of the carotid bodies with cyanide or of arterial baroreceptors by raising perfusion pressure produced the expected robust excitation of cardiovagal neurons, and so did the activation of the diving reflex (A2). A3, PND‐triggered histograms of EPSPs recorded in cardiovagal ganglionic neurons (CVGNs) indicate that, in this preparation, CVPGNs discharge preferentially during the postinspiratory phase. (B) Schematic interpretation based on A2‐3 and C. Rat cardiovagal preganglionic neurons could be receiving inhibitory inputs from early‐I and Aug‐E neurons. The existence of inhibitory input during inspiration has been documented in slices as shown in C. Pulmonary stretch receptors (inactive in an arterially perfused preparation) inhibit cardiovagal preganglionic neurons, possibly via a direct input from GABAergic “pump cells” located within the NTS. Carotid body stimulation produces opposite effects on CVPGN activity depending on the intensity of the stimulus. Mild stimulation inhibits these neurons via the RPG, presumably via inhibitory inputs from early‐I and post‐I neurons as shown in B. Acute and intense stimulation of the carotid bodies (e.g., with cyanide) activates cardiovagal preganglionic neurons as shown in A2. This effect may be mediated by polymodal NTS neurons that also respond to noxious stimulation of the airways (pathway not represented). Post‐I glutamatergic neurons are an alternative plausible source of excitation of cardiovagal preganglionic neurons and other vagal efferents with a post‐I discharge pattern but the existence of such neurons has yet to be demonstrated. (Panel C was originally published in (153), and has been reproduced by permission of Oxford University Press [http://www.oxfordscholarship.com/view/10.1093/acprof:oso/9780195306637.001.0001/acprof‐9780195306637]).
Figure 12. Figure 12. Feedback regulation of RTN neurons and interaction between central and peripheral respiratory chemoreflexes. (A1‐4) Four examples of RTN neurons showing different types of respiratory modulation which can be interpreted as post‐I and E‐aug inhibition in cell 1, early‐I and post‐I inhibition in cell 2, early‐I only in cell 3, and early‐I, post‐I and E‐AUG in cell 4 [adapted, with permission, from (158)]. (B) Typical RTN neuron devoid of respiratory modulation at low end‐expiratory CO2. This cell exhibits the early‐I/post‐I pattern when FiCO2 is increased (unpublished example from P. Guyenet). C, single RTN neuron inhibited by lung‐inflation [Tp, tracheal pressure, iPND integrated rectified phrenic nerve discharge; adapted, with permission, from (296)]. (D1) Single RTN neuron recorded in a vagally intact anesthetized rat showing the complex respiratory modulation of the cell (integrated rate histogram and rectified PND triggered on expiratory CO2 shown as bottom trace). (D2) Average steady‐state response of RTN neurons to end‐expiratory CO2 in the same preparation as D1 [D1‐D2 adapted, with permission, from (158)]. (E1 and E2) Similar recordings after i.c.v. administration of the glutamatergic antagonist kynurenic acid. (E1) PND and the respiratory modulation of the RTN neuron was abolished by kynurenic acid. (E2) The relationship between the discharge rate of RTN neurons and end‐expiratory CO2 became linear after kynurenic acid administration [adapted, with permission, from (158)]. (F) Interpretation of the results shown in A‐E: the response of RTN neurons to CO2 is saturable because of the existence of inhibitory feedback from lung stretch receptors and from the RPG. G, schematic wiring diagram based on A‐E. (H) Hypothetical scenario in which the RPG is unusually active or excitable (left). In such a case, respiratory feedback to the RTN would minimize the contribution of this nucleus to ventilation causing hypoadditivity between the peripheral and central chemoreflexes. On the other hand, additivity would result from a situation in which the RPG is moderately excitable (right panel). (I) Possible hyperadditivity scenario. RTN hyperpolarization at rest (right panel) could result in hyperadditivity between the central and peripheral chemoreflexes. In this configuration, moderate hypercapnia alone would produce a minimal stimulation of breathing because RTN depolarization would be largely subthreshold. Carotid body stimulation would depolarize RTN neurons above their discharge threshold causing them to respond vigorously to the previously ineffective hypercapnic stimulus, hence the apparent hyperadditivity of the reflexes.
Figure 13. Figure 13. Feedback regulation of RTN neurons and interaction between central and peripheral respiratory chemoreflexes. (A1‐4) Four examples of RTN neurons showing different types of respiratory modulation which can be interpreted as post‐I and E‐aug inhibition in cell 1, early‐I and post‐I inhibition in cell 2, early‐I only in cell 3, and early‐I, post‐I and E‐AUG in cell 4 [adapted, with permission, from (158)]. (B) Typical RTN neuron devoid of respiratory modulation at low end‐expiratory CO2. This cell exhibits the early‐I/post‐I pattern when FiCO2 is increased (unpublished example from P. Guyenet). C, single RTN neuron inhibited by lung‐inflation [Tp, tracheal pressure, iPND integrated rectified phrenic nerve discharge; adapted, with permission, from (296)]. (D1) Single RTN neuron recorded in a vagally intact anesthetized rat showing the complex respiratory modulation of the cell (integrated rate histogram and rectified PND triggered on expiratory CO2 shown as bottom trace). (D2) Average steady‐state response of RTN neurons to end‐expiratory CO2 in the same preparation as D1 [D1‐D2 adapted, with permission, from (158)]. (E1 and E2) Similar recordings after i.c.v. administration of the glutamatergic antagonist kynurenic acid. (E1) PND and the respiratory modulation of the RTN neuron was abolished by kynurenic acid. (E2) The relationship between the discharge rate of RTN neurons and end‐expiratory CO2 became linear after kynurenic acid administration [adapted, with permission, from (158)]. (F) Interpretation of the results shown in A‐E: the response of RTN neurons to CO2 is saturable because of the existence of inhibitory feedback from lung stretch receptors and from the RPG. G, schematic wiring diagram based on A‐E. (H) Hypothetical scenario in which the RPG is unusually active or excitable (left). In such a case, respiratory feedback to the RTN would minimize the contribution of this nucleus to ventilation causing hypoadditivity between the peripheral and central chemoreflexes. On the other hand, additivity would result from a situation in which the RPG is moderately excitable (right panel). (I) Possible hyperadditivity scenario. RTN hyperpolarization at rest (right panel) could result in hyperadditivity between the central and peripheral chemoreflexes. In this configuration, moderate hypercapnia alone would produce a minimal stimulation of breathing because RTN depolarization would be largely subthreshold. Carotid body stimulation would depolarize RTN neurons above their discharge threshold causing them to respond vigorously to the previously ineffective hypercapnic stimulus, hence the apparent hyperadditivity of the reflexes.


Figure 1. Organigram of the chemoreflexes. Cascade of cardiorespiratory responses elicited in anesthetized mammals by hypoxic stimulation of the carotid bodies or by hypercapnia. These cardiorespiratory responses are elaborated primarily within spinal and pontomedullary circuits. The same circuits are also presumably recruited by small perturbations of the blood gases in the conscious state to stabilize PCO2. Large acute perturbations of blood gases produce arousal, aversive sensations and stress, responses that involve numerous other brain regions and processes. The direct effects of hypoxia on the CNS are not considered here. Green arrows denote cell activation (e.g., carotid bodies by hypoxia) or a globally excitatory connection (e.g., effect of the carotid bodies, CBs, on the RPG), or an increase in a dependent variable [e.g., effect of CO2 on cerebral blood flow (CBF) resulting in a “washout” of brain CO2]. Red arrows have the opposite meaning. The baroreflex (BaroR) potentiates or attenuates the chemoreflexes depending on the direction of the change in arterial pressure (AP). Slowly adapting lung stretch receptors (SARs) exert a feedback regulation on the RPG and on central chemoreceptors (CCRs) and inhibit the cardiovagal outflow (CVPSN, cardiovagal parasympathetic nerve activity). The chemoreceptors, both central and peripheral, activate the sympathetic nervous system (SNS) both via the RPG and independently of it.


Figure 2. Pontomedullary regions responsible for eupneic breathing and for generating the autonomic outflows to the cardiovascular system: anatomy and simplified circuitry. (A1) Parasagittal section through the pons and medulla oblongata of a rodent. The regions colored in magenta contain the principal building blocks of the respiratory pattern generator. The ventral respiratory column (VRC) contains four functional compartments aligned in rostrocaudal order (Bötzinger Complex (BötC), pre‐BötC, rostral ventral respiratory group (rVRG) and caudal VRG (cVRG)). The retrotrapezoid nucleus (RTN) resides at the rostral end of the VRC under the facial motor nucleus. In this article, the term RTN refers specifically to a cluster of about 2000 CO2‐activated Phox2b‐ir glutamatergic neurons (in rats, 800 in mice). (A2) Minimal circuitry responsible for the generation of eupneic breathing [adapted, with permission, from Lindsey, Ryback & Smith (247)]. The drawing depicts some of the neuronal interconnections within and between the four compartments of the ventral respiratory column and a few of the connections of RTN neurons (for details see text). The parafacial respiratory group (pfRG) is a physiologically defined entity now believed to be specifically involved in the generation of active expiration (331). Its constituent neurons and their location are not yet defined. Bötzinger augmenting expiratory neurons have been included by some authors in the pfRG (328). Inhibitory (GABAergic or glycinergic) neurons are represented in red, glutamatergic neurons in green, motoneurons in blue, connections with both excitatory and inhibitory components in magenta (e.g., neurons transmitting information from arterial baroreceptors, pulmonary stretch afferents, the carotid bodies etc.). (B1) The regions colored in magenta are thought to contain the main components of the network that generates the autonomic outflows to the cardiovascular system. From an autonomic regulation standpoint, the ventrolateral medulla can be subdivided into three regions whose anatomical relationship with the respiratory compartments can be appreciated by comparing panels A1 and B1. (B2) Schematic of cardiovagal parasympathetic neurons, RVLM presympathetic neurons and connections responsible for their regulation by arterial baroreceptors. Abbreviations: aug‐E, augmenting expiratory neurons; aug‐I, augmenting inspiratory neurons (a.k.a. inspiratory premotor neurons); CVLM, caudal VLM; DRG, dorsal respiratory group (caudolateral portion of the NTS); early‐I, early‐inspiratory neurons, early‐I(1) and early‐I(2) are postulated to have distinct input‐output functions; IVLM, intermediate VLM; Itr, intertrigeminal region; KF, Kölliker‐Fuse nucleus; LPBN, lateral parabrachial nuclei; LRt, lateral reticular nucleus; Mo5, trigeminal motor nucleus; NTS, nucleus of the solitary tract; pfRG, parafacial respiratory group; Pn, pontine nuclei; post‐I, postinspiratory; preI/I, preinspiratory/inspiratory (putative rhythmogenic neurons); scp, superior cerebellar peduncle; SO, superior olive; SPGN, sympathetic preganglionic neurons; SPN sympathetic (post)ganglionic neuron; VRC, ventral respiratory column.


Figure 3. Regulation of the sympathetic vasomotor outflow by the respiratory pattern generator. (A) Plausible circuitry responsible for coupling respiration with the sympathetic nerve activity to the cardiovascular system. Sympathetic ganglionic neurons (SPNs) display stereotyped patterns of respiratory modulation in deafferented preparations (vagotomized, barodenervated). Two of the most commonly found patterns in rats (early‐I and post‐I) are depicted in D [top traces, average integrated phrenic nerve discharge, iPND; lower trace iPND‐triggered rate histogram depicting the probability of firing of a single SPN during the central respiratory cycle; redrawn, with permission, after (72)]. These SPN patterns are virtually identical to those of single RVLM presympathetic neurons [shown in B; redrawn, with permission, from (168)]. The IVLM contains GABAergic neurons that are activated by arterial baroreceptor stimulation and inhibit RVLM presympathetic neurons (panel A). The respiratory modulation of these IVLM GABAergic neurons is approximately the mirror image of that of the RVLM presympathetic neurons [C; reprinted from Mandel and Schreihofer (266) with permission from Wiley & Sons] hence the hypothesis that IVLM neurons with early‐inspiratory discharge produce post‐I modulation in RVLM presympathetic neurons and vice versa (red arrows in C). The depicted inputs from the RPG to IVLM neurons are plausible but speculative. (E) Intracellular recordings of RVLM presympathetic neurons in an arterially perfused midcollicular transected rat preparation [from Moraes et al. (294) with permission]. The early‐I (left trace) and the post‐I patterns (middle trace) are instantly recognizable. The right panel shows a presympathetic RVLM neuron that exhibits an extra peak of activity during late expiration. The latter recording was made in a rat subjected to chronic intermittent hypoxia which caused late‐expiratory activity in an abdominal nerve at rest (not shown) and an additional late‐E peak of activity in the thoracic chain (red arrow pointing down and to the right above the thoracic chain SNA trace, tSNA). An excitatory input from pfRG has been tentatively proposed as the source of this late‐expiratory activation (illustrated in A).


Figure 4. Gain and loss of function of RTN Phox2b neurons in conscious rodents: effect on breathing and the hypercapnic ventilatory reflex (HCVR). (A) Steady‐state ventilatory response of a conscious rat exposed to graded levels of hypercapnia. Breathing was measured by whole body plethysmography [reprinted from (236) with permission from Elsevier]. Note that, at 8% FiCO2, fR (respiratory rate) doubles and VE triples. (B) Unilateral acidification of the RTN with a dialysis probe containing a fluid equilibrated with 25% CO2 produces a small (24% average) increase in VE [adapted from Li et al. J. Appl. Physiol. (240) with permission]. (C) Unilateral optogenetic activation (20Hz) of around 35% of RTN Phox2b neurons triples VE in a conscious rat [adapted, with permission, from (4)]. (C1) Typical example (plethysmography); (C2‐C4) average responses (from top to bottom: frequency, tidal volume and minute volume) to unilateral optogenetic stimulation of RTN neurons. (C5) Selective expression of ChR2‐mCherry fusion protein by RTN (i.e., Phox2b+) neurons in one such animal (Phox2b in green; mCherry in red). (D1) Attenuation of the hypercapnic ventilatory reflex by i.c.v. allatostatin administration to a conscious rat in which an unknown fraction of RTN neurons were transduced with the allatostatin receptor. (D2) Average results from five rats [(from Marina et al. (270) with permission]. (E1) Absence of HCVR in neonatal (day 9) transgenic mice in which a mutated form of transcription factor Phox2b (Phox2b‐27ala) is expressed selectively by cells of rhombomere 3 and 5 lineage (Phox2b27Alacki; Egr2cre/0) preventing or aborting RTN neuron development. (E2) Group data. (E3) Partial recovery (one third) of the HCVR in adult Phox2b27Alacki; Egr2cre/0 mice. (E4) Absence of Phox2b‐ir neurons in the RTN of a Phox2b27Alacki; Egr2cre/0 mouse (embryonic day 14; RTN identified by white oval, Phox2b in red) [E1‐E4 reprinted from Ramanantsoa et al. (347) with permission].


Figure 5. Gain and loss of function of RTN Phox2b neurons in conscious rodents: effect on breathing and the hypercapnic ventilatory reflex (HCVR). (A) Steady‐state ventilatory response of a conscious rat exposed to graded levels of hypercapnia. Breathing was measured by whole body plethysmography [reprinted from (236) with permission from Elsevier]. Note that, at 8% FiCO2, fR (respiratory rate) doubles and VE triples. (B) Unilateral acidification of the RTN with a dialysis probe containing a fluid equilibrated with 25% CO2 produces a small (24% average) increase in VE [adapted from Li et al. J. Appl. Physiol. (240) with permission]. (C) Unilateral optogenetic activation (20Hz) of around 35% of RTN Phox2b neurons triples VE in a conscious rat [adapted, with permission, from (4)]. (C1) Typical example (plethysmography); (C2‐C4) average responses (from top to bottom: frequency, tidal volume and minute volume) to unilateral optogenetic stimulation of RTN neurons. (C5) Selective expression of ChR2‐mCherry fusion protein by RTN (i.e., Phox2b+) neurons in one such animal (Phox2b in green; mCherry in red). (D1) Attenuation of the hypercapnic ventilatory reflex by i.c.v. allatostatin administration to a conscious rat in which an unknown fraction of RTN neurons were transduced with the allatostatin receptor. (D2) Average results from five rats [(from Marina et al. (270) with permission]. (E1) Absence of HCVR in neonatal (day 9) transgenic mice in which a mutated form of transcription factor Phox2b (Phox2b‐27ala) is expressed selectively by cells of rhombomere 3 and 5 lineage (Phox2b27Alacki; Egr2cre/0) preventing or aborting RTN neuron development. (E2) Group data. (E3) Partial recovery (one third) of the HCVR in adult Phox2b27Alacki; Egr2cre/0 mice. (E4) Absence of Phox2b‐ir neurons in the RTN of a Phox2b27Alacki; Egr2cre/0 mouse (embryonic day 14; RTN identified by white oval, Phox2b in red) [E1‐E4 reprinted from Ramanantsoa et al. (347) with permission].


Figure 6. Activation of RTN neurons by CO2. (A) Location and anatomical projections of RTN neurons (definition of RTN in Fig. 2 legend; abbreviations as in Fig. 2). (B) Transverse section through the lower right quadrant of the rat medulla oblongata at the level of the dotted line in A. The facial motor nucleus is in blue (choline acetyl‐transferase immunoreactivity), the C1 presympathetic neurons are in red (tyrosine hydroxylase) and phox2b immunoreactivity (green, nuclear localization) identifies RTN neurons [adapted, with permission, from (402)]. (C) Response of an RTN neuron to graded hypercapnia in an anesthetized rat (end‐expiratory CO2 shown in top trace) [adapted, with permission, from (402)]. (D) Example of one RTN neuron excited by brief hypoxia (carotid body stimulation, bottom trace) and by hypercapnia (end‐expiratory CO2 in green). After i.c.v. administration of the broad spectrum glutamatergic blocker kynurenic acid, the cell no longer responds to hypoxia but its response to hypercapnia is unaffected [adapted, with permission, from (301)]. (E) RTN CO2‐activated neuron labeled juxtacellularly with biotinamide in vivo (green fluorescence) has a Phox2b‐ir nucleus [adapted, with permission, from (402)]. (F) Structure of an RTN neuron whose cell body was located at the ventral surface of the medulla oblongata (in vivo recording, juxtacellular labeling with biotinamide, and transverse plane projection). Note the extensive dendrites on the ventral surface [adapted, with permission, from (301)]. (G1) Two intracellularly labeled RTN neurons recorded in a Phox2b‐eGFP transgenic mouse coronal slice (green eGFP, red biotinamide). (G2) Representative acid sensitivity of one such neuron recorded at room temperature (cell attached recording, integrated rate histogram, 10 s bin) [adapted, with permission, from (231)]. (H1) RTN Phox2b+ neuron acutely isolated from a Phox2b‐eGFP mouse. (H2) Acid sensitivity of such an acutely isolated RTN neuron. [adapted, with permission, from (447)]. Note that the cell responds to a change in pH, not to CO2 per se. I1, selective expression of TASK‐2 potassium channels by Phox2b+ RTN neurons in mice [adapted from Gestreau et al. (137) with permission]. Left panel shows TASK‐2 expression monitored with LacZ reaction product. Middle panel shows that TASK‐2 expressing neurons are eliminated in a mouse line in which RTN neurons express the 27‐ala Phox2b mutation and fail to develop (Task2+/−; Phox2b27Ala/+ ). Right panel: Task‐2 expressing neurons from a Task2+/‐ mouse (ventral surface view) showing the superficial location in perfect register with the RTN. I2, reduced acid sensitivity of RTN neurons in Task‐2 knock‐out mice compared to Task‐2 +/− control mice [adapted, with permission, from (446)].


Figure 7. Serotonergic neurons and breathing. (A) Selective expression of ChR2‐mCherry fusion protein by raphe obscurus serotonergic neurons in an e‐PET Cre mouse [adapted, with permission, after (86)]. (B) Photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in such a mouse (ketamine/dexmedetomidine initial anesthesia with 0.3% to 0.6% isoflurane supplementation) increases diaphragm EMG amplitude (top trace) and respiratory rate (fR, second from top). Bottom traces shows a single raphe serotonergic unit being photoactivated every time the laser light was pulsed [adapted, with permission, after (86)]. (C) Selective optogenetic photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in a conscious mouse (whole body plethysmography). The response has the same slow ON‐slow OFF kinetics as in the anesthetized mouse [adapted, with permission, after (155)]. (D) Robust acid sensitivity of a serotonergic neuron in culture [reprinted from Richerson (354)] adapted with permission from Macmillan Publishers Ltd., Nature Neuroscience (5), 2004). (E1) In an arterially perfused rat preparation, serotonergic neurons are either modestly inhibited or modestly excited by CO2. (E2) In this preparation, hypercapnia had no effect on the mean activity of the serotonergic population at large (all 5‐HT) [from Iceman et al. (188) with permission]. (F) Putative serotonergic neuron recorded in conscious cats showing a modest but dose‐dependent increase in discharge rate (2.7 to 3.9 Hz; +44% at 6% FiCO2). The effect of CO2 essentially disappeared during non‐REM sleep [reproduced from Veasey et al. (441) with permission]. (G) Serotonin2A receptor antagonism dramatically slows the pre‐Bötzinger complex rhythm in a slice (top traces, mass activity of pre‐Bötzinger complex neurons, bottom traces whole cell recording of a presumed rhythmogenic neuron) [reproduced from Peña and Ramirez (338) with permission]. (H1‐2) Iontophoretic application of serotonin activates RTN neurons by a constant amount regardless of the level of end‐expiratory CO2 [H1, representative example, H2 average results; adapted, with permission, from (300)]. (I) Selective global inhibition of CNS serotonergic neurons (DREADD methodology) attenuates the hypercapnic ventilatory reflex of a conscious mouse [after Ray et al. (350) reprinted with permission from AAAS]. (J) Interpretations: serotonergic neurons activate breathing via innumerable mechanisms (e.g., direct projection to RTN, the RPG, dorsolateral pons, motoneurons, and indirect effects via changes in vigilance). The integrity of serotonergic neurons is required for full expression of the hypercapnic ventilatory reflex, possibly because a fraction of serotonergic neuron is directly responsive to acidification in vivo.


Figure 8. Serotonergic neurons and breathing. (A) Selective expression of ChR2‐mCherry fusion protein by raphe obscurus serotonergic neurons in an e‐PET Cre mouse [adapted, with permission, after (86)]. (B) Photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in such a mouse (ketamine/dexmedetomidine initial anesthesia with 0.3% to 0.6% isoflurane supplementation) increases diaphragm EMG amplitude (top trace) and respiratory rate (fR, second from top). Bottom traces shows a single raphe serotonergic unit being photoactivated every time the laser light was pulsed [adapted, with permission, after (86)]. (C) Selective optogenetic photostimulation of ChR2‐expressing raphe obscurus serotonergic neurons in a conscious mouse (whole body plethysmography). The response has the same slow ON‐slow OFF kinetics as in the anesthetized mouse [adapted, with permission, after (155)]. (D) Robust acid sensitivity of a serotonergic neuron in culture [reprinted from Richerson (354)] adapted with permission from Macmillan Publishers Ltd., Nature Neuroscience (5), 2004). (E1) In an arterially perfused rat preparation, serotonergic neurons are either modestly inhibited or modestly excited by CO2. (E2) In this preparation, hypercapnia had no effect on the mean activity of the serotonergic population at large (all 5‐HT) [from Iceman et al. (188) with permission]. (F) Putative serotonergic neuron recorded in conscious cats showing a modest but dose‐dependent increase in discharge rate (2.7 to 3.9 Hz; +44% at 6% FiCO2). The effect of CO2 essentially disappeared during non‐REM sleep [reproduced from Veasey et al. (441) with permission]. (G) Serotonin2A receptor antagonism dramatically slows the pre‐Bötzinger complex rhythm in a slice (top traces, mass activity of pre‐Bötzinger complex neurons, bottom traces whole cell recording of a presumed rhythmogenic neuron) [reproduced from Peña and Ramirez (338) with permission]. (H1‐2) Iontophoretic application of serotonin activates RTN neurons by a constant amount regardless of the level of end‐expiratory CO2 [H1, representative example, H2 average results; adapted, with permission, from (300)]. (I) Selective global inhibition of CNS serotonergic neurons (DREADD methodology) attenuates the hypercapnic ventilatory reflex of a conscious mouse [after Ray et al. (350) reprinted with permission from AAAS]. (J) Interpretations: serotonergic neurons activate breathing via innumerable mechanisms (e.g., direct projection to RTN, the RPG, dorsolateral pons, motoneurons, and indirect effects via changes in vigilance). The integrity of serotonergic neurons is required for full expression of the hypercapnic ventilatory reflex, possibly because a fraction of serotonergic neuron is directly responsive to acidification in vivo.


Figure 9. Glial cells and central respiratory chemosensitivity. (A) Acidification increases intracellular calcium fluorescence in medullary astrocytes [from Gourine et al. (146) reprinted with permission from AAAS]. (B) Optogenetic depolarization of ChR2‐expressing ventrolateral medullary astrocytes activates the phrenic outflow in an anesthetized rat. Note the very slow onset of the response to light and its persistence [adapted, with permission, from (146) reprinted with permission from AAAS]. (C) Activation of RTN neurons by CO2 in a slice of neonate rat brain is attenuated by application of the P2 receptor antagonists PPADS or suramin. Summary data (right panels): about a quarter of the overall response of RTN neurons to 15% CO2 could be mediated by ATP release in this preparation [reproduced from Wenker et al. (454) with permission]. (D) Schema of the contribution of ventral medullary surface (VMS) astrocytes to central respiratory chemosensitivity according to (145) [after Gourine and Kasparov, Exp. Phsiol. (96), 2011 reprinted with permission from John Wiley & Sons]. VMS astrocytes are thought to mediate the hypercapnic ventilatory reflex by simultaneously activating RTN neurons and undefined components of the central pattern generating (CPG). ATP release by astrocytes may be a calcium‐dependent exocytotic process triggered by intracellular acidification and/or a leak through connexin channels (Cx26 primarily) opened by molecular CO2 via carbamylation.


Figure 10. Sympathetic nerve activation by carotid body stimulation: lower brainstem pathways. The four experiments depicted around the interpretative schematic (A‐D, clockwise from top right) suggest that, in anesthetized rodents, carotid body stimulation increases sympathetic vasomotor tone via two pathways that converge on RVLM presympathetic neurons, only one of which depends on the respiratory pattern generator. (A) Microinjection of the GABA mimetic agonist muscimol into the commissural portion of the NTS abolishes the response to cyanide [adapted, with permission, from (295)]. (B) Injection of muscimol into the rVRG eliminates the phrenic nerve discharge but does not change the sympathetic response to carotid body activation (N2, 10 s nitrogen inhalation) [adapted, with permission, from (217)]. (C) Injection of muscimol into the pre‐Bötzinger complex, eliminates the phrenic nerve discharge and eliminates the respiratory oscillations of the sympathetic nerve discharge elicited by carotid body stimulation. However, the sympathoexcitation produced by carotid body stimulation persists. The response of a simultaneously recorded single RVLM presympathetic neuron is also shown. Note that muscimol produces the same effect on the neuron as on SNA, that is, muscimol changes the response from respiratory synchronous oscillations to a tonic activation [adapted, with permission, from (216)]. (D) Administration of kynurenic acid into the RVLM dramatically reduces SNA activation caused by carotid body stimulation [after (295) reprinted with permission from the Society for Neuroscience]. The respiratory dependent pathway of the chemoreflex is considered in greater detail in Fig. 3).


Figure 11. Regulation of the cardiovagal parasympathetic outflow by the respiratory pattern generator. (A1‐3) [Reproduced from McAllen et al. (278), reprinted with permission.] Intracellular recordings of cardiovagal ganglionic neurons from an arterially perfused rat preparation in which the lungs are not inflated therefore reflexes from lung stretch receptors are absent (A1). The firing properties of cardiovagal preganglionic neurons (CVPGN) were inferred from the discharge pattern of the ganglionic neurons (A2) or the EPSPs recorded from these neurons (A3). Activation of the carotid bodies with cyanide or of arterial baroreceptors by raising perfusion pressure produced the expected robust excitation of cardiovagal neurons, and so did the activation of the diving reflex (A2). A3, PND‐triggered histograms of EPSPs recorded in cardiovagal ganglionic neurons (CVGNs) indicate that, in this preparation, CVPGNs discharge preferentially during the postinspiratory phase. (B) Schematic interpretation based on A2‐3 and C. Rat cardiovagal preganglionic neurons could be receiving inhibitory inputs from early‐I and Aug‐E neurons. The existence of inhibitory input during inspiration has been documented in slices as shown in C. Pulmonary stretch receptors (inactive in an arterially perfused preparation) inhibit cardiovagal preganglionic neurons, possibly via a direct input from GABAergic “pump cells” located within the NTS. Carotid body stimulation produces opposite effects on CVPGN activity depending on the intensity of the stimulus. Mild stimulation inhibits these neurons via the RPG, presumably via inhibitory inputs from early‐I and post‐I neurons as shown in B. Acute and intense stimulation of the carotid bodies (e.g., with cyanide) activates cardiovagal preganglionic neurons as shown in A2. This effect may be mediated by polymodal NTS neurons that also respond to noxious stimulation of the airways (pathway not represented). Post‐I glutamatergic neurons are an alternative plausible source of excitation of cardiovagal preganglionic neurons and other vagal efferents with a post‐I discharge pattern but the existence of such neurons has yet to be demonstrated. (Panel C was originally published in (153), and has been reproduced by permission of Oxford University Press [http://www.oxfordscholarship.com/view/10.1093/acprof:oso/9780195306637.001.0001/acprof‐9780195306637]).


Figure 12. Feedback regulation of RTN neurons and interaction between central and peripheral respiratory chemoreflexes. (A1‐4) Four examples of RTN neurons showing different types of respiratory modulation which can be interpreted as post‐I and E‐aug inhibition in cell 1, early‐I and post‐I inhibition in cell 2, early‐I only in cell 3, and early‐I, post‐I and E‐AUG in cell 4 [adapted, with permission, from (158)]. (B) Typical RTN neuron devoid of respiratory modulation at low end‐expiratory CO2. This cell exhibits the early‐I/post‐I pattern when FiCO2 is increased (unpublished example from P. Guyenet). C, single RTN neuron inhibited by lung‐inflation [Tp, tracheal pressure, iPND integrated rectified phrenic nerve discharge; adapted, with permission, from (296)]. (D1) Single RTN neuron recorded in a vagally intact anesthetized rat showing the complex respiratory modulation of the cell (integrated rate histogram and rectified PND triggered on expiratory CO2 shown as bottom trace). (D2) Average steady‐state response of RTN neurons to end‐expiratory CO2 in the same preparation as D1 [D1‐D2 adapted, with permission, from (158)]. (E1 and E2) Similar recordings after i.c.v. administration of the glutamatergic antagonist kynurenic acid. (E1) PND and the respiratory modulation of the RTN neuron was abolished by kynurenic acid. (E2) The relationship between the discharge rate of RTN neurons and end‐expiratory CO2 became linear after kynurenic acid administration [adapted, with permission, from (158)]. (F) Interpretation of the results shown in A‐E: the response of RTN neurons to CO2 is saturable because of the existence of inhibitory feedback from lung stretch receptors and from the RPG. G, schematic wiring diagram based on A‐E. (H) Hypothetical scenario in which the RPG is unusually active or excitable (left). In such a case, respiratory feedback to the RTN would minimize the contribution of this nucleus to ventilation causing hypoadditivity between the peripheral and central chemoreflexes. On the other hand, additivity would result from a situation in which the RPG is moderately excitable (right panel). (I) Possible hyperadditivity scenario. RTN hyperpolarization at rest (right panel) could result in hyperadditivity between the central and peripheral chemoreflexes. In this configuration, moderate hypercapnia alone would produce a minimal stimulation of breathing because RTN depolarization would be largely subthreshold. Carotid body stimulation would depolarize RTN neurons above their discharge threshold causing them to respond vigorously to the previously ineffective hypercapnic stimulus, hence the apparent hyperadditivity of the reflexes.


Figure 13. Feedback regulation of RTN neurons and interaction between central and peripheral respiratory chemoreflexes. (A1‐4) Four examples of RTN neurons showing different types of respiratory modulation which can be interpreted as post‐I and E‐aug inhibition in cell 1, early‐I and post‐I inhibition in cell 2, early‐I only in cell 3, and early‐I, post‐I and E‐AUG in cell 4 [adapted, with permission, from (158)]. (B) Typical RTN neuron devoid of respiratory modulation at low end‐expiratory CO2. This cell exhibits the early‐I/post‐I pattern when FiCO2 is increased (unpublished example from P. Guyenet). C, single RTN neuron inhibited by lung‐inflation [Tp, tracheal pressure, iPND integrated rectified phrenic nerve discharge; adapted, with permission, from (296)]. (D1) Single RTN neuron recorded in a vagally intact anesthetized rat showing the complex respiratory modulation of the cell (integrated rate histogram and rectified PND triggered on expiratory CO2 shown as bottom trace). (D2) Average steady‐state response of RTN neurons to end‐expiratory CO2 in the same preparation as D1 [D1‐D2 adapted, with permission, from (158)]. (E1 and E2) Similar recordings after i.c.v. administration of the glutamatergic antagonist kynurenic acid. (E1) PND and the respiratory modulation of the RTN neuron was abolished by kynurenic acid. (E2) The relationship between the discharge rate of RTN neurons and end‐expiratory CO2 became linear after kynurenic acid administration [adapted, with permission, from (158)]. (F) Interpretation of the results shown in A‐E: the response of RTN neurons to CO2 is saturable because of the existence of inhibitory feedback from lung stretch receptors and from the RPG. G, schematic wiring diagram based on A‐E. (H) Hypothetical scenario in which the RPG is unusually active or excitable (left). In such a case, respiratory feedback to the RTN would minimize the contribution of this nucleus to ventilation causing hypoadditivity between the peripheral and central chemoreflexes. On the other hand, additivity would result from a situation in which the RPG is moderately excitable (right panel). (I) Possible hyperadditivity scenario. RTN hyperpolarization at rest (right panel) could result in hyperadditivity between the central and peripheral chemoreflexes. In this configuration, moderate hypercapnia alone would produce a minimal stimulation of breathing because RTN depolarization would be largely subthreshold. Carotid body stimulation would depolarize RTN neurons above their discharge threshold causing them to respond vigorously to the previously ineffective hypercapnic stimulus, hence the apparent hyperadditivity of the reflexes.
References
 1. Abbott SB , Coates MB , Stornetta RL , Guyenet PG . Optogenetic stimulation of C1 and retrotrapezoid nucleus neurons causes sleep state‐dependent cardiorespiratory stimulation and arousal in rats. Hypertension 61:835‐841, 2013.
 2. Abbott SB , Depuy SD , Nguyen T , Coates MB , Stornetta RL , Guyenet PG . Selective optogenetic activation of rostral ventrolateral medullary catecholaminergic neurons produces cardiorespiratory stimulation in conscious mice. J Neurosci 33:3164‐3177, 2013.
 3. Abbott SB , Kanbar R , Bochorishvili G , Coates MB , Stornetta RL , Guyenet PG . C1 neurons excite locus coeruleus and A5 noradrenergic neurons along with sympathetic outflow in rats. J Physiol 590:2897‐2915, 2012.
 4. Abbott SB , Stornetta RL , Coates MB , Guyenet PG . Phox2b‐expressing neurons of the parafacial region regulate breathing rate, inspiration, and expiration in conscious rats. J Neurosci 31:16410‐16422, 2011.
 5. Abbott SB , Stornetta RL , Fortuna MG , Depuy SD , West GH , Harris TE , Guyenet PG . Photostimulation of retrotrapezoid nucleus phox2b‐expressing neurons in vivo produces long‐lasting activation of breathing in rats. J Neurosci 29:5806‐5819, 2009.
 6. Abdala AP , McBryde FD , Marina N , Hendy EB , Engelman ZJ , Fudim M , Sobotka PA , Gourine AV , Paton JF . Hypertension is critically dependent on the carotid body input in the spontaneously hypertensive rat. J Physiol 590:4269‐4277, 2012.
 7. 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.
 8. Abdala AP , Rybak IA , Smith JC , Zoccal DB , Machado BH , St‐John WM , Paton JF . Multiple pontomedullary mechanisms of respiratory rhythmogenesis. Respir Physiol Neurobiol 168:19‐25, 2009.
 9. Aicher SA , Saravay RH , Cravo S , Jeske I , Morrison SF , Reis DJ , Milner TA . Monosynaptic projections from the nucleus tractus solitarii to C1 adrenergic neurons in the rostral ventrolateral medulla: Comparison with input from the caudal ventrolateral medulla. J Comp Neurol 373:62‐75, 1996.
 10. Ainslie PN , Duffin J . Integration of cerebrovascular CO2 reactivity and chemoreflex control of breathing: Mechanisms of regulation, measurement, and interpretation. Am J Physiol Regul Integr Comp Physiol 296:R1473‐R1495, 2009.
 11. Alheid GF , Jiao W , McCrimmon DR . Caudal nuclei of the rat nucleus of the solitary tract differentially innervate respiratory compartments within the ventrolateral medulla. Neurosci 190:207‐227, 2011.
 12. Alheid GF , McCrimmon DR . The chemical neuroanatomy of breathing. Respir Physiol Neurobiol 164:3‐11, 2008.
 13. Allen AM . Inhibition of the hypothalamic paraventricular nucleus in spontaneously hypertensive rats dramatically reduces sympathetic vasomotor tone. Hypertension 39:275‐280, 2002.
 14. Allen AM , Dosanjh JK , Erac M , Dassanayake S , Hannan RD , Thomas WG . Expression of constitutively active angiotensin receptors in the rostral ventrolateral medulla increases blood pressure. Hypertension 47:1054‐1061, 2006.
 15. Aller MI , Wisden W . Changes in expression of some two‐pore domain potassium channel genes (KCNK) in selected brain regions of developing mice. Neurosci 151:1154‐1172, 2008.
 16. Amiel J , Dubreuil V , Ramanantsoa N , Fortin G , Gallego J , Brunet JF , Goridis C . PHOX2B in respiratory control: Lessons from congenital central hypoventilation syndrome and its mouse models. Respir Physiol Neurobiol 168:125‐132, 2009.
 17. Amiel J , Laudier B , Attie‐Bitach T , Trang H , de PL, Gener B , Trochet D , Etchevers H , Ray P , Simonneau M , Vekemans M , Munnich A , Gaultier C , Lyonnet S . Polyalanine expansion and frameshift mutations of the paired‐like homeobox gene PHOX2B in congenital central hypoventilation syndrome. Nat Genet 33:459‐461, 2003.
 18. Anazco C , Pena‐Munzenmayer G , Araya C , Cid LP , Sepulveda FV , Niemeyer MI . G protein modulation of K potassium channel TASK‐2: A role of basic residues in the C terminus domain. Pflugers Arch 465:1715‐1726, 2013.
 19. Anders K , Ballantyne D , Bischoff AM , Lalley PM , Richter DW . Inhibition of caudal medullary expiratory neurones by retrofacial inspiratory neurones in the cat. J Physiol 437:1‐25, 1991.
 20. Appleyard SM , Marks D , Kobayashi K , Okano H , Low MJ , Andresen MC . Visceral afferents directly activate catecholamine neurons in the solitary tract nucleus. J Neurosci 27:13292‐13302, 2007.
 21. Arita H , Kogo N , Koshiya N . Morphological and physiological properties of caudal medullary expiratory neurons of the cat. Brain Res 401(2):258‐266, 1987.
 22. Armbruster BN , Li X , Pausch MH , Herlitze S , Roth BL . Evolving the lock to fit the key to create a family of G protein‐coupled receptors potently activated by an inert ligand. Proc Natl Acad Sci U S A 104:5163‐5168, 2007.
 23. Aston‐Jones G , Bloom FE . Activity of norepinephrine‐containing locus coeruleus neurons in behaving rats anticipates fluctuations in the sleep‐waking cycle. J Neurosci 1:876‐886, 1981.
 24. Aston‐Jones G , Cohen JD . An integrative theory of locus coeruleus‐norepinephrine function: Adaptive gain and optimal performance. Annu Rev Neurosci 28:403‐450, 2005.
 25. Aston‐Jones G , Ennis M , Pieribone VA , Nickell WT , Shipley MT . The brain nucleus locus coeruleus: Restricted afferent control of a broad efferent network. Science 234:734‐737, 1986.
 26. Atasoy D , Betley JN , Su HH , Sternson SM . Deconstruction of a neural circuit for hunger. Nature 488:172‐177, 2012.
 27. Bailey TW , Hermes SM , Andresen MC , Aicher SA . Cranial visceral afferent pathways through the nucleus of the solitary tract to caudal ventrolateral medulla or paraventricular hypothalamus: Target‐specific synaptic reliability and convergence patterns. J Neurosci 26:11893‐11902, 2006.
 28. Bainton CR , Kirkwood PA . The effect of carbon dioxide on the tonic and the rhythmic discharges of expiratory bulbospinal neurones. J Physiol 296:291‐314, 1979.
 29. Banzett RB , Lansing RW , Brown R , Topulos GP , Yager D , Steele SM , Londono B , Loring SH , Reid MB , Adams L . ‘Air hunger’ from increased PCO2 persists after complete neuromuscular block in humans. Respir Physiol 81:1‐17, 1990.
 30. Barna BF , Takakura AC , Moreira TS . Pontomedullary and hypothalamic distribution of Fos‐like immunoreactive neurons after acute exercise in rats. Neurosci 212:120‐130, 2012.
 31. Bayliss DA , Barrett PQ . Emerging roles for two‐pore‐domain potassium channels and their potential therapeutic impact. Trends Pharmacol Sci 29:566‐575, 2008.
 32. Becker LE . Neural maturational delay as a link in the chain of events leading to SIDS 176. Can J Neurol Sci 17:361‐371, 1990.
 33. Ben‐Tal A , Shamailov SS , Paton JF . Evaluating the physiological significance of respiratory sinus arrhythmia: Looking beyond ventilation‐perfusion efficiency. J Physiol 590:1989‐2008, 2012.
 34. Berger AJ , Krasney JA , Dutton RE . Respiratory recovery from CO2 breathing in intact and chemodenervated awake dogs. J Appl Physiol 35:35‐41, 1973.
 35. Berquin P , Bodineau L , Gros F , Larnicol N . Brainstem and hypothalamic areas involved in respiratory chemoreflexes: A Fos study in adult rats. Brain Res 857:30‐40, 2000.
 36. Berridge CW , Waterhouse BD . The locus coeruleus‐noradrenergic system: Modulation of behavioral state and state‐dependent cognitive processes. Brain Res Brain Res Rev 42:33‐84, 2003.
 37. Bevan AT , Honour AJ , Stott FH . Direct arterial pressure recording in unrestricted man. Clin Sci 36:329‐344, 1969.
 38. Biancardi V , Bicego KC , Almeida MC , Gargaglioni LH . Locus coeruleus noradrenergic neurons and CO2 drive to breathing. Pflugers Arch 455:1119‐1128, 2008.
 39. Biancardi V , da Silva LT , Bicego KC , Gargaglioni LH . Role of Locus coeruleus noradrenergic neurons in cardiorespiratory and thermal control during hypoxia. Respir Physiol Neurobiol 170:150‐156, 2010.
 40. Blain GM , Smith CA , Henderson KS , Dempsey JA . Contribution of the carotid body chemoreceptors to eupneic ventilation in the intact, unanesthetized dog. J Appl Physiol 106:1564‐1573, 2009.
 41. Blain GM , Smith CA , Henderson KS , Dempsey JA . Peripheral chemoreceptors determine the respiratory sensitivity of central chemoreceptors to CO(2). J Physiol 588:2455‐2471, 2010.
 42. Bochorishvili G , Stornetta RL , Coates MB , Guyenet PG . Pre‐Botzinger complex receives glutamatergic innervation from galaninergic and other retrotrapezoid nucleus neurons. J Comp Neurol 520:1047‐1061, 2012.
 43. Bodineau L , Frugière A , Marlot D , Wallois F . Connections between retrotrapezoid nucleus and nucleus tractus solitarii in cat. Neurosci Lett 280:111‐114, 2000a.
 44. Bodineau L , Frugiere A , Marlot D , Wallois F . Effect of hypoxia on the activity of respiratory and non‐respiratory modulated retrotrapezoid neurons of the cat. Auton Neurosci 86:70‐77, 2000b.
 45. Bowes G , Townsend ER , Kozar LF , Bromley SM , Phillipson EA . Effect of carotid body denervation on arousal response to hypoxia in sleeping dogs. J Appl Physiol 51:40‐45, 1981.
 46. Bradley SR , Pieribone VA , Wang WG , Severson CA , Jacobs RA , Richerson GB . Chemosensitive serotonergic neurons are closely associated with large medullary arteries. Nat Neurosci 5:401‐402, 2002.
 47. Braga VA , Soriano RN , Braccialli AL , De Paula PM , Bonagamba LG , Paton JF , Machado BH . Involvement of L‐glutamate and ATP in the neurotransmission of the sympathoexcitatory component of the chemoreflex in the commissural nucleus tractus solitarii of awake rats and in the working heart‐brainstem preparation. J Physiol 581:1129‐1145, 2007.
 48. Brown RE , Sergeeva O , Eriksson KS , Haas HL . Orexin A excites serotonergic neurons in the dorsal raphe nucleus of the rat. Neuropharmacology 40:457‐459, 2001.
 49. Buchanan GF , Richerson GB . Central serotonin neurons are required for arousal to CO2 . Proc Natl Acad Sci U S A 107:16354‐16359, 2010.
 50. Campos RR , McAllen RM . Tonic drive to sympathetic premotor neurons of rostral ventrolateral medulla from caudal pressor area neurons. Am J Physiol Regul Integr Comp Physiol 276:R1209‐R1213, 1999.
 51. Card JP , Sved JC , Craig B , Raizada M , Vazquez J , Sved AF . Efferent projections of rat rostroventrolateral medulla C1 catecholamine neurons: Implications for the central control of cardiovascular regulation. J Comp Neurol 499:840‐859, 2006.
 52. Chamberlin NL . Functional organization of the parabrachial complex and intertrigeminal region in the control of breathing. Respir Physiol Neurobiol 143:115‐125, 2004.
 53. Chamberlin NL , Saper CB . Topographic organization of cardiovascular responses to electrical and glutamate microstimulation of the parabrachial nucleus in the rat. J Comp Neurol 326:245‐262, 1992.
 54. Chamberlin NL , Saper CB . Topographic organization of respiratory responses to glutamate microstimulation of the parabrachial nucleus in the rat. J Neurosci 14:6500‐6510, 1994.
 55. Chan RKW , Sawchenko PE . Organization and transmitter specificity of medullary neurons activated by sustained hypertension: Implications for understanding baroreceptor reflex circuitry. J Neurosci 18:371‐387, 1998.
 56. Chonan T , Mulholland MB , Leitner J , Altose MD , Cherniack NS . Sensation of dyspnea during hypercapnia, exercise, and voluntary hyperventilation. J Appl Physiol 68:2100‐2106, 1990.
 57. Cid LP , Roa‐Rojas HA , Niemeyer MI , Gonzalez W , Araki M , Araki K , Sepulveda FV . TASK‐2: a K2P K(+) channel with complex regulation and diverse physiological functions. Front Physiol 4:198, 2013.
 58. Coates EL , Li A , Nattie EE . Widespread sites of brain stem ventilatory chemoreceptors. J Appl Physiol 75:5‐14, 1993.
 59. Coates EL , Li AH , Nattie EE . Acetazolamide on the ventral medulla of the cat increases phrenic output and delays the ventilatory response to CO2 . J Physiol 441:433‐451, 1991.
 60. Connelly CA , Ellenberger HH , Feldman JL . Respiratory activity in retrotrapezoid nucleus in cat. Am J Physiol 258:L33‐L44, 1990.
 61. Coote JH . Spinal and supraspinal reflex pathways of cardio‐cardiac sympathetic reflexes. Neurosci Lett 46:243‐247, 1984.
 62. Coote JH . A role for the paraventricular nucleus of the hypothalamus in the autonomic control of heart and kidney. Exp Physiol 90:169‐173, 2005.
 63. Cui Z , Fisher JA , Duffin J . Central‐peripheral respiratory chemoreflex interaction in humans. Respir Physiol Neurobiol 180:126‐131, 2012.
 64. Cummings KJ , Hewitt JC , Li A , Daubenspeck JA , Nattie EE . Postnatal loss of brainstem serotonin neurons compromises the ability of neonatal rats to survive episodic severe hypoxia. J Physiol 589:5247‐5256, 2011.
 65. Cushing H . Concerning a definitive regulatory mechanism of the vaso‐motor centre which controls blood pressure during cerebral compression. B Johns Hopkins Hosp 12:290‐292, 1901.
 66. da Silva GS , Li A , Nattie E . High CO2/H +dialysis in the caudal ventrolateral medulla (Loeschcke's area) increases ventilation in wakefulness. Respir Physiol Neurobiol 171:46‐53, 2010.
 67. Dampney RA , Horiuchi J , McDowall LM . Hypothalamic mechanisms coordinating cardiorespiratory function during exercise and defensive behaviour. Auton Neurosci 142:3‐10, 2008.
 68. Dampney RA , McAllen RM . Differential control of sympathetic fibres supplying hindlimb skin and muscle by subretrofacial neurones in the cat. J Physiol 395:41‐56, 1988.
 69. Dampney RAL . Functional organization of central pathways regulating the cardiovascular system. Physiol Rev 74:323‐364, 1994.
 70. Dampney RAL . The subretrofacial vasomotor nucleus: Anatomical, chemical and pharmacological properties and role in cardiovascular regulation. Prog in Neurobiol 42:197‐228, 1994.
 71. Darnall RA . The carotid body and arousal in the fetus and neonate. Respir Physiol Neurobiol 185:132‐143, 2013.
 72. Darnall RA , Guyenet P . Respiratory modulation of pre‐ and postganglionic lumbar vasomotor sympathetic neurons in the rat. Neurosci Lett 119:148‐152, 1990.
 73. Dauger S , Pattyn A , Lofaso F , Gaultier C , Goridis C , Gallego J , Brunet JF . Phox2b controls the development of peripheral chemoreceptors and afferent visceral pathways. Development 130:6635‐6642, 2003.
 74. Davis SE , Solhied G , Castillo M , Dwinell MR , Brozoski D , Forster HV . Postnatal developmental changes in CO2 sensitivity in rats. J Appl Physiol 101: 1097‐103, 2006.
 75. Day TA , Wilson RJ . A negative interaction between brainstem and peripheral respiratory chemoreceptors modulates peripheral chemoreflex magnitude. J Physiol 587:883‐896, 2009.
 76. Dean JB , Bayliss DA , Erickson JT , Lawing WL , Millhorn DE . Depolarization and stimulation of neurons in nucleus tractus solitarii by carbon dioxide does not require chemical synaptic input. Neurosci 36:207‐216, 1990.
 77. Dean JB , Lawing WL , Millhorn DE . CO2 decreases membrane conductance and depolarizes neurons in the nucleus tractus solitarii. Exp Brain Res 76:656‐661, 1989.
 78. Dejours P , Raynoud J , CUENOD CL , LABROUSSE Y . Instantaneous modifications of ventilation at the beginning and at the cessation of muscular exercise; interpretation. J Physiol (Paris) 47:155‐159, 1955.
 79. Del Negro CA , Koshiya N , Butera RJ, Jr. , Smith JC . Persistent sodium current, membrane properties and bursting behavior of pre‐Botzinger complex inspiratory neurons in vitro. J Neurophysiol 88:2242‐2250, 2002.
 80. Del Rio R , Marcus NJ , Schultz HD . Carotid chemoreceptor ablation improves survival in heart failure: Rescuing autonomic control of cardiorespiratory function. J Am Coll Cardiol 62:2422‐2430, 2013.
 81. Dembowsky K , Czachurski J , Amendt K , Seller H . Tonic descending inhibition of the spinal somato‐sympathetic reflex from the lower brain stem. J Auton Nerv Syst 2:157‐182, 1980.
 82. Dembowsky K , Lackner K , Czachurski J , Seller H . Tonic catecholaminergic inhibition of the spinal somato‐sympathetic reflexes originating in the ventrolateral medulla oblongata. J Auton Nerv Syst 3:277‐290, 1981.
 83. Dempsey JA . New perspectives concerning feedback influences on cardiorespiratory control during rhythmic exercise and on exercise performance. J Physiol 590:4129‐4144, 2012.
 84. Dempsey JA , Sheel AW , St Croix CM , Morgan BJ . Respiratory influences on sympathetic vasomotor outflow in humans. Respir Physiol Neurobiol 130:3‐20, 2002.
 85. Dempsey JA , Veasey SC , Morgan BJ , O'Donnell CP . Pathophysiology of sleep apnea. Physiol Rev 90:47‐112, 2010.
 86. Depuy SD , Kanbar R , Coates MB , Stornetta RL , Guyenet PG . Control of breathing by raphe obscurus serotonergic neurons in mice. J Neurosci 31:1981‐1990, 2011.
 87. Deuchars SA . Multi‐tasking in the spinal cord–do ‘sympathetic’ interneurones work harder than we give them credit for? J Physiol 580:723‐729, 2007.
 88. Deuchars SA , Milligan CJ , Stornetta RL , Deuchars J . GABAergic neurons in the central region of the spinal cord: A novel substrate for sympathetic inhibition. J Neurosci 25:1063‐1070, 2005.
 89. Dias MB , Li A , Nattie EE . Focal CO2 dialysis in raphe obscurus does not stimulate ventilation but enhances the response to focal CO2 dialysis in the retrotrapezoid nucleus (RTN). J Appl Physiol 105:83‐90, 2008.
 90. Dias MB , Li A , Nattie EE . Antagonism of orexin receptor 1 (OX1R) in the retrotrapezoid nucleus (RTN) inhibits the ventilatory response to hypercapnia predominantly in wakefulness. J Physiol 587:2059‐2067, 2009.
 91. Dias MB , Nucci TB , Margatho LO , ntunes‐Rodrigues J , Gargaglioni LH , Branco LGS . Raphe Magnus Nucleus is involved in ventilatory but not hypothermic response to CO2 . J Appl Physiol 103:1780‐1788, 2007.
 92. Dobbins EG , Feldman JL . Brainstem network controlling descending drive to phrenic motoneurons in rat. J Comp Neurol 347:64‐86, 1994.
 93. Doi A , Ramirez JM . Neuromodulation and the orchestration of the respiratory rhythm. Respir Physiol Neurobiol 164:96‐104, 2008.
 94. Doi A , Ramirez JM . State‐dependent interactions between excitatory neuromodulators in the neuronal control of breathing. J Neurosci 30:8251‐8262, 2010.
 95. Donoghue S , Felder RB , Gilbey MP , Jordan D , Spyer KM . Post‐synaptic activity evoked in the nucleus tractus solitarius by carotid sinus and aortic nerve afferents in the cat. J Physiol 360:261‐273, 1985.
 96. Donoghue S , Felder RB , Jordan D , Spyer KM . The central projections of carotid baroreceptors and chemoreceptors in the cat: A neurophysiological study. J Physiol 347:397‐409, 1984.
 97. Dubreuil V , Ramanantsoa N , Trochet D , Vaubourg V , Amiel J , Gallego J , Brunet JF , Goridis C . A human mutation in Phox2b causes lack of CO2 chemosensitivity, fatal central apnoea and specific loss of parafacial neurons. Proc Natl Acad Sci U S A 105:1067‐1072, 2008.
 98. Dubreuil V , Thoby‐Brisson M , Rallu M , Persson K , Pattyn A , Birchmeier C , Brunet JF , Fortin G , Goridis C . Defective respiratory rhythmogenesis and loss of central chemosensitivity in phox2b mutants targeting retrotrapezoid nucleus neurons. J Neurosci 29:14836‐14846, 2009.
 99. Duffin J . The role of the central chemoreceptors: A modeling perspective. Respir Physiol Neurobiol 173:230‐243, 2010.
 100. Duffin J , Mateika JH . Cross‐Talk opposing view: Peripheral and central chemoreflexes have additive effects on ventilation in humans. J Physiol 591:4351‐4353, 2013.
 101. Dun NJ , Mo N . Inhibitory postsynaptic potentials in neonatal rat sympathetic preganglionic neurones in vitro. J Physiol 410:267‐281, 1989.
 102. 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 303:430‐437, 2010.
 103. Duprat F , Lauritzen I , Patel A , Honore E . The TASK background K2P channels: Chemo‐ and nutrient sensors. Trends Neurosci 30:573‐580, 2007.
 104. Elam M , Yao T , Thoren P , Svensson TH . Hypercapnia and Hypoxia: Chemoreceptor‐mediated control of locus coeruleus neurons and splanchnic sympathetic nerve. Brain Res 222:373‐381, 1981.
 105. Eldridge FL . Central integration of mechanisms in exercise hyperpnea. Med Sci Sports Exerc 26:319‐327, 1994.
 106. Eldridge FL , Millhorn DE , Kiley JP , Waldrop TG . Stimulation by central command of locomotion, respiration and circulation during exercise. Respir Physiol 59:313‐337, 1985.
 107. Ellenberger HH , Feldman JL . Brainstem connections of the rostral ventral respiratory group of the rat. Brain Res 513:35‐42, 1990.
 108. Ennis M , Aston‐Jones G . Potent inhibitory input to locus coeruleus from the nucleus prepositus hypoglossi. Brain Res Bull 22:793‐803, 1989.
 109. Erlichman JS , Leiter JC . Glia modulation of the extracellular milieu as a factor in central CO2 chemosensitivity and respiratory control. J Appl Physiol (1985) 108:1803‐1811, 2010.
 110. Erlichman JS , Leiter JC , Gourine AV . ATP, glia and central respiratory control. Respir Physiol Neurobiol 173:305‐311, 2010.
 111. Erlichman JS , Li A , Nattie EE . Ventilatory effects of glial dysfunction in a rat brain stem chemoreceptor region. J Appl Physiol 85:1599‐1604, 1998.
 112. Esler M . The sympathetic system and hypertension. Am J Hypertens 13:99S‐105S, 2000.
 113. Esler M , Kaye D . Sympathetic nervous system activation in essential hypertension, cardiac failure and psychosomatic heart disease. J Cardiovasc Pharmacol 35:S1‐S7, 2000.
 114. Estabrooke IV , McCarthy MT , Ko E , Chou TC , Chemelli RM , Yanagisawa M , Saper CB , Scammell TE . Fos expression in orexin neurons varies with behavioral state. J Neurosci 21:1656‐1662, 2001.
 115. Fatouleh R , Macefield VG . Respiratory modulation of muscle sympathetic nerve activity is not increased in essential hypertension or chronic obstructive pulmonary disease. J Physiol 589:4997‐5006, 2011.
 116. Feldman JL , Del Negro CA , Gray PA . Understanding the rhythm of breathing: So near, yet so far. Annu Rev Physiol 75:423‐452, 2013.
 117. Feldman JL , Mitchell GS , Nattie EE . Breathing: Rhythmicity, plasticity, chemosensitivity. Annu Rev Neurosci 26:239‐266, 2003.
 118. Finley JCW , Katz DM . The central organization of carotid body afferent projections to the brainstem of the rat. Brain Res 571(2):108‐116, 1992.
 119. Fletcher EC . Effect of episodic hypoxia on sympathetic activity and blood pressure. Resp Physiol 119:189‐197, 2000.
 120. Fletcher EC . Invited review: Physiological consequences of intermittent hypoxia: Systemic blood pressure. J Appl Physiol (1985) 90:1600‐1605, 2001.
 121. Fletcher EC , Bao G , Miller CC, III . Effect of recurrent episodic hypocapnic, eucapnic, and hypercapnic hypoxia on systemic blood pressure. J Appl Physiol (1985) 78:1516‐1521, 1995.
 122. Fletcher EC , Lesske J , Qian W , Miller CC, III , Unger T . Repetitive, episodic hypoxia causes diurnal elevation of blood pressure in rats. Hypertension 19:555‐561, 1992.
 123. Forster HV , Haouzi P , Dempsey JA . Control of breathing during exercise. Compr Physiol 2:743‐777, 2012.
 124. Forster HV , Smith CA . Contributions of central and peripheral chemoreceptors to the ventilatory response to CO2/H+. J Appl Physiol 108:989‐994, 2010.
 125. Fortuna MG , Stornetta RL , West GH , Guyenet PG . Activation of the retrotrapezoid nucleus by posterior hypothalamic stimulation. J Physiol 587:5121‐5138, 2009.
 126. Fukuda Y , Honda Y . pH‐sensitive cells at ventro–lateral surface of rat medulla oblongata. Nature 256:317‐318, 1975.
 127. Fukuda Y , Honda Y , Schlafke ME , Loeschcke HH . Effect of H+ on the membrane potential of silent cells in the ventral and dorsal surface layers of the rat medulla in vitro. Pflugers Arch 376:229‐235, 1978.
 128. Fukuda Y , Sato A , Trzebski A . Carotid chemoreceptor discharge responses to hypoxia and hypercapnia in normotensive and spontaneously hypertensive rats. J Auton Nerv Syst 19:1‐11, 1987.
 129. Gao L , Schultz HD , Patel KP , Zucker IH , Wang W . Augmented input from cardiac sympathetic afferents inhibits baroreflex in rats with heart failure. Hypertension 45:1173‐1181, 2005.
 130. Garcia AJ, III , Koschnitzky JE , Dashevskiy T , Ramirez JM . Cardiorespiratory coupling in health and disease. Auton Neurosci 175:26‐37, 2013.
 131. Garcia AJ, III , Koschnitzky JE , Ramirez JM . The physiological determinants of Sudden Infant Death Syndrome. Respir Physiol Neurobiol 189:288‐300, 2013.
 132. Gargaglioni LH , Hartzler LK , Putnam RW . The locus coeruleus and central chemosensitivity. Respir Physiol Neurobiol 173:264‐273, 2010.
 133. Gatti PJ , Johnson TA , Massari VJ . Can neurons in the nucleus ambiguus selectively regulate cardiac rate and atrio‐ventricular conduction? J Auton Nerv Syst 57:123‐127, 1996.
 134. Gautier H . Interactions among metabolic rate, hypoxia, and control of breathing. J Appl Physiol (1985) 81:521‐527, 1996.
 135. Gebber GL , Barman SM . Studies on the origin and generation of sympathetic nerve activity. Clin Exp Hypertens A 10(Suppl 1):33‐44, 1988.
 136. Geerling JC , Shin JW , Chimenti PC , Loewy AD . Paraventricular hypothalamic nucleus: Axonal projections to the brainstem. J Comp Neurol 518:1460‐1499, 2010.
 137. Gestreau C , Heitzmann D , Thomas J , Dubreuil V , Bandulik S , Reichold M , Bendahhou S , Pierson P , Sterner C , Peyronnet‐Roux J , Benfriha C , Tegtmeier I , Ehnes A , Georgieff M , Lesage F , Brunet JF , Goridis C , Warth R , Barhanin J . Task2 potassium channels set central respiratory CO2 and O2 sensitivity. Proc Natl Acad Sci U S A 107:2325‐2330, 2010.
 138. Giaume C , Leybaert L , Naus CC , Saez JC . Connexin and pannexin hemichannels in brain glial cells: Properties, pharmacology, and roles. Front Pharmacol 4:88, 2013.
 139. Gleeson K , Zwillich CW , White DP . The influence of increasing ventilatory effort on arousal from sleep. Am Rev Respir Dis 142:295‐300, 1990.
 140. Gonczi M , Szentandrassy N , Johnson IT , Heagerty AM , Weston AH . Investigation of the role of TASK‐2 channels in rat pulmonary arteries; pharmacological and functional studies following RNA interference procedures. Br J Pharmacol 147:496‐505, 2006.
 141. Gonzalez D , Gomez‐Hernandez JM , Barrio LC . Species specificity of mammalian connexin‐26 to form open voltage‐gated hemichannels. Faseb J 20:2329‐2338, 2006.
 142. Gonzalez D , Gomez‐Hernandez JM , Barrio LC . Molecular basis of voltage dependence of connexin channels: An integrative appraisal. Prog Biophys Mol Biol 94:66‐106, 2007.
 143. Gonzalez JA , Jensen LT , Doyle SE , Miranda‐Anaya M , Menaker M , Fugger L , Bayliss DA , Burdakov D . Deletion of TASK1 and TASK3 channels disrupts intrinsic excitability but does not abolish glucose or pH responses of orexin/hypocretin neurons. Eur J Neurosci 30:57‐64, 2009.
 144. Goridis C , Brunet JF . Central chemoreception: Lessons from mouse and human genetics. Respir Physiol Neurobiol 173:312‐321, 2010.
 145. Gourine AV , Kasparov S . Astrocytes as brain interoceptors. Exp Physiol 96:411‐416, 2011.
 146. Gourine AV , Kasymov V , Marina N , Tang F , Figueiredo MF , Lane S , Teschemacher AG , Spyer KM , Deisseroth K , Kasparov S . Astrocytes control breathing through pH‐dependent release of ATP. Science 329:571‐575, 2010.
 147. Grace KP , Hughes SW , Horner RL . Identification of the mechanism mediating genioglossus muscle suppression in REM sleep. Am J Respir Crit Care Med 187:311‐319, 2013.
 148. Granjeiro EM , Scopinho AA , Correa FM , Resstel LB . Prelimbic but not infralimbic cortex is involved in the pressor response to chemoreflex activation in awake rats. Exp Physiol 96:518‐527, 2011.
 149. Gray PA . Transcription factors and the genetic organization of brainstem respiratory neurons. J Appl Physiol 104:1513‐1521, 2008.
 150. Grkovic I , Fernandez K , McAllen RM , Anderson CR . Misidentification of cardiac vagal pre‐ganglionic neurons after injections of retrograde tracer into the pericardial space in the rat. Cell Tissue Res 321:335‐340, 2005.
 151. Guo L , Bi X , Li Z , Zhang C . Effect of methylmercuric chloride on apoptosis and c‐fos expression of brain glial cells. Wei Sheng Yan Jiu 31:7‐9, 2002.
 152. Guyenet PG . The 2008 Carl Ludwig Lecture: Retrotrapezoid nucleus, CO2 homeostasis, and breathing automaticity. J Appl Physiol 105:404‐416, 2008.
 153. Guyenet PG. Cardiorespiratory Integration. In Central Regulation of Autonomic Functions. Edited by Llewellyn‐Smith IJ, Verberne AJ. New‐York: Oxford University Press, 2011, pp. 180‐201.
 154. Guyenet PG , Abbott SB . Chemoreception and asphyxia‐induced arousal. Respir Physiol Neurobiol 188:333‐343, 2013.
 155. Guyenet PG , Abbott SB , Stornetta RL . The respiratory chemoreception conundrum: Light at the end of the tunnel? Brain Res 1511: 126‐137, 2012.
 156. Guyenet PG , Brown DL . Unit activity in nucleus paragigantocellularis lateralis during cerebral ischemia in the rat. Brain Res 364:301‐314, 1986.
 157. Guyenet PG , Filtz TM , Donaldson SR . Role of excitatory amino acids in rat vagal and sympathetic baroreflexes. Brain Res 407:272‐284, 1987.
 158. Guyenet PG , Mulkey DK , Stornetta RL , Bayliss DA . Regulation of ventral surface chemoreceptors by the central respiratory pattern generator. J Neurosci 25:8938‐8947, 2005.
 159. Guyenet PG , Stornetta RL , Abbott SB , Depuy SD , Fortuna MG , Kanbar R . Central CO2‐chemoreception and integrated neural mechanisms of cardiovascular and respiratory control. J Appl Physiol 108:995‐1002, 2010.
 160. Guyenet PG , Stornetta RL , Bochorishvili G , Depuy SD , Burke PG , Abbott SB . Invited Review EB 2012: C1 neurons: The body's EMTs. Am J Physiol Regul Integr Comp Physiol 305:R187‐R204, 2013.
 161. Habler HJ , Janig W , Michaelis M . Respiratory modulation in the activity of sympathetic neurones. Prog Neurobiol 43:567‐606, 1994.
 162. Haddad GG , Jiang C . Mechanisms of anoxia‐induced depolarization in brainstem neurons ‐ invitro current and voltage clamp studies in the adult rat. Brain Res 625:261‐268, 1993.
 163. Haldane JS , Priestley JG . The regulation of the lung‐ventilation. J Physiol 32:225‐266, 1905.
 164. Hammarstrom AK , Gage PW . Inhibition of oxidative metabolism increases persistent sodium current in rat CA1 hippocampal neurons. J Physiol 510:735‐741, 1998.
 165. Hammarstrom AK , Gage PW . Hypoxia and persistent sodium current. Eur Biophys J 31:323‐330, 2002.
 166. Haouzi P . Theories on the nature of the coupling between ventilation and gas exchange during exercise. Respir Physiol Neurobiol 151:267‐279, 2006.
 167. Hartzler LK , Dean JB , Putnam RW . The chemosensitive response of neurons from the locus coeruleus (LC) to hypercapnic acidosis with clamped intracellular pH. Adv Exp Med Biol 605:333‐337, 2008.
 168. Haselton JR , Guyenet PG . Central respiratory modulation of medullary sympathoexcitatory neurons in rat. Am J Physiol Regul Integr Comp Physiol 256:R739‐R750, 1989.
 169. Hassani OK , Henny P , Lee MG , Jones BE . GABAergic neurons intermingled with orexin and MCH neurons in the lateral hypothalamus discharge maximally during sleep. Eur J Neurosci 32:448‐457, 2010.
 170. Hawryluk JM , Moreira TS , Takakura AC , Wenker IC , Tzingounis AV , Mulkey DK . KCNQ channels determine serotonergic modulation of ventral surface chemoreceptors and respiratory drive. J Neurosci 32:16943‐16952, 2012.
 171. Haxhiu MA , Yung K , Erokwu B , Cherniack NS . CO2‐induced c‐fos expression in the CNS catecholaminergic neurons. Resp Physiol 105:35‐45, 1996.
 172. Herndon RM , Coyle JT . Selective destruction of neurons by a transmitter agonist. Science 198:71‐72, 1977.
 173. Hilton SM , Marshall JM . The pattern of cardiovascular response to carotid chemoreceptor stimulation in the cat. J Physiol 326:495‐513, 1982.
 174. Hodges MR , Tattersall GJ , Harris MB , McEvoy SD , Richerson DN , Deneris ES , Johnson RL , Chen ZF , Richerson GB . Defects in breathing and thermoregulation in mice with near‐complete absence of central serotonin neurons. J Neurosci 28:2495‐2505, 2008.
 175. Honda Y . Respiratory and circulatory activities in carotid body‐resected humans. J Appl Physiol 73:1‐8, 1992.
 176. Hori T , Roth GI , Yamamoto WS . Respiratory sensitivity of rat brain‐stem surface to chemical stimuli. J Appl Physiol 28:721‐724, 1970.
 177. Horiuchi J , Dampney RA . Evidence for tonic disinhibition of RVLM sympathoexcitatory neurons from the caudal pressor area. Auton Neurosci 99:102‐110, 2002.
 178. Horiuchi J , McDowall LM , Dampney RA . Vasomotor and respiratory responses evoked from the dorsolateral periaqueductal grey are mediated by the dorsomedial hypothalamus. J Physiol 587:5149‐5162, 2009.
 179. Horn EM , Waldrop TG . Hypoxic augmentation of fast‐inactivating and persistent sodium currents in rat caudal hypothalamic neurons. J Neurophysiol 84:2572‐2581, 2000.
 180. Horner RL . Emerging principles and neural substrates underlying tonic sleep‐state‐dependent influences on respiratory motor activity. Philos Trans R Soc Lond B Biol Sci 364:2553‐2564, 2009.
 181. Horvath TL , Peyron C , Diano S , Ivanov A , Aston‐Jones G , Kilduff TS , Van den Pol AN . Hypocretin (Orexin) activation and synaptic innervation of the locus coeruleus noradrenergic system. J Comp Neurol 415:145‐159, 1999.
 182. Hu J , Zhong C , Ding C , Chi Q , Walz A , Mombaerts P , Matsunami H , Luo M . Detection of near‐atmospheric concentrations of CO2 by an olfactory subsystem in the mouse. Science 317:953‐957, 2007.
 183. Huckstepp RT , Dale N . Redefining the components of central CO2 chemosensitivity–towards a better understanding of mechanism. J Physiol 589:5561‐5579, 2011.
 184. Huckstepp RT , Eason R , Sachdev A , Dale N . CO2‐dependent opening of connexin 26 and related beta connexins. J Physiol 588:3921‐3931, 2010.
 185. Huckstepp RT , Id BR , Eason R , Spyer KM , Dicke N , Willecke K , Marina N , Gourine AV , Dale N . Connexin hemichannel‐mediated CO2‐dependent release of ATP in the medulla oblongata contributes to central respiratory chemosensitivity. J Physiol 588:3901‐3920, 2010.
 186. Huda R , McCrimmon DR , Martina M . pH modulation of glial glutamate transporters regulates synaptic transmission in the nucleus of the solitary tract. J Neurophysiol 110:368‐377, 2013.
 187. Hwang DY , Carlezon WA, Jr. , Isacson O , Kim KS . A high‐efficiency synthetic promoter that drives transgene expression selectively in noradrenergic neurons. Hum Gene Ther 12:1731‐1740, 2001.
 188. Iceman KE , Richerson GB , Harris MB . Medullary serotonin neurons are CO2‐sensitive in situ. J Neurophysiol 110:2536‐2544, 2013.
 189. Jacobs BL . Single unit activity of locus coeruleus neurons in behaving animals. Prog in Neurobiol 27:183‐194, 1986.
 190. Jacobs BL , Martín‐Cora FJ , Fornal CA . Activity of medullary serotonergic neurons in freely moving animals. Brain Res Rev 40:45‐52, 2002.
 191. Janczewski WA , Feldman JL . Distinct rhythm generators for inspiration and expiration in the juvenile rat. J Physiol 570:407‐420, 2006.
 192. Janczewski WA , Tashima A , Hsu P , Cui Y , Feldman JL . Role of inhibition in respiratory pattern generation. J Neurosci 33:5454‐5465, 2013.
 193. Janig W , Habler HJ . Neurophysiological analysis of target‐related sympathetic pathways–from animal to human: Similarities and differences. Acta Physiol Scand 177:255‐274, 2003.
 194. Jansen ASP , Nguyen XV , Karpitskiy V , Mettenleiter TC , Loewy AD . Central command neurons of the sympathetic nervous system:basis of the fight‐or flight response. Science 270:644‐646, 1995.
 195. Jensen P , Farago AF , Awatramani RB , Scott MM , Deneris ES , Dymecki SM . Redefining the serotonergic system by genetic lineage. Nat Neurosci 11:417‐419, 2008.
 196. Jin YH , Bailey TW , Li BY , Schild JH , Andresen MC . Purinergic and vanilloid receptor activation releases glutamate from separate cranial afferent terminals in nucleus tractus solitarius. J Neurosci 24:4709‐4717, 2004.
 197. Ju YK , Saint DA , Gage PW . Hypoxia increases persistent sodium current in rat ventricular myocytes. J Physiol 497:337‐347, 1996.
 198. Kam K , Worrell JW , Janczewski WA , Cui Y , Feldman JL . Distinct inspiratory rhythm and pattern generating mechanisms in the preBotzinger complex. J Neurosci 33:9235‐9245, 2013.
 199. Kang BJ , Chang DA , Mackay DD , West GH , Moreira TS , Takakura AC , Gwilt JM , Guyenet PG , Stornetta RL . Central nervous system distribution of the transcription factor Phox2b in the adult rat. J Comp Neurol 503:627‐641, 2007.
 200. Kangrga IM , Loewy AD . Whole‐cell recordings from visualized C1 adrenergic bulbospinal neurons: Ionic mechanisms underlying vasomotor tone. Brain Res 670:215‐232, 1995.
 201. Kasymov V , Larina O , Castaldo C , Marina N , Patrushev M , Kasparov S , Gourine AV . Differential sensitivity of brainstem versus cortical astrocytes to changes in pH reveals functional regional specialization of astroglia. J Neurosci 33:435‐441, 2013.
 202. Kaur S , Pedersen NP , Yokota S , Hur EE , Fuller PM , Lazarus M , Chamberlin NL , Saper CB . Glutamatergic signaling from the parabrachial nucleus plays a critical role in hypercapnic arousal. J Neurosci 33:7627‐7640, 2013.
 203. Kawai A , Ballantyne D , Muckenhoff K , Scheid P . Chemosensitive medullary neurones in the brainstem–spinal cord preparation of the neonatal rat. J Physiol 492:277‐292, 1996.
 204. Kawai A , Onimaru H , Homma I . Mechanisms of CO2/H+ chemoreception by respiratory rhythm generator neurons in the medulla from newborn rats in vitro. J Physiol 572:525‐537, 2006.
 205. Kc P , Haxhiu MA , Trouth CO , Balan KV , Anderson WA , Mack SO . CO2‐induced c‐Fos expression in hypothalamic vasopressin containing neurons. Resp Physiol 129:289‐296, 2002.
 206. Kim JE , Kwak SE , Kang TC . Upregulated TWIK‐related acid‐sensitive K+ channel‐2 in neurons and perivascular astrocytes in the hippocampus of experimental temporal lobe epilepsy. Epilepsia 50:654‐663, 2009.
 207. Kimura Y , Hirooka Y , Sagara Y , Ito K , Kishi T , Shimokawa H , Takeshita A , Sunagawa K . Overexpression of inducible nitric oxide synthase in rostral ventrolateral medulla causes hypertension and sympathoexcitation via an increase in oxidative stress. Circ Res 96:252‐260, 2005.
 208. Kinney HC , Thach BT . The sudden infant death syndrome. N Engl J Med 361:795‐805, 2009.
 209. Kirkwood PA . Synaptic excitation in the thoracic spinal cord from expiratory bulbospinal neurones in the cat. J Physiol 484:201‐225, 1995.
 210. Kishi T , Hirooka Y , Sakai K , Shigematsu H , Shimokawa H , Takeshita A . Overexpression of eNOS in the RVLM causes hypotension and bradycardia via GABA release. Hypertension 38:896‐901, 2001.
 211. Kline DD . Chronic intermittent hypoxia affects integration of sensory input by neurons in the nucleus tractus solitarii. Respir Physiol Neurobiol 174:29‐36, 2010.
 212. Kline DD , Ramirez‐Navarro A , Kunze DL . Adaptive depression in synaptic transmission in the nucleus of the solitary tract after in vivo chronic intermittent hypoxia: Evidence for homeostatic plasticity. J Neurosci 27:4663‐4673, 2007.
 213. Koizumi H , Smerin SE , Yamanishi T , Moorjani BR , Zhang R , Smith JC . TASK channels contribute to the K+‐dominated leak current regulating respiratory rhythm generation in vitro. J Neurosci 30:4273‐4284, 2010.
 214. Koizumi H , Smith JC . Persistent Na+ and K+‐dominated leak currents contribute to respiratory rhythm generation in the pre‐Botzinger complex in vitro. J Neurosci 28:1773‐1785, 2008.
 215. Koshiya N , Guyenet PG . NTS neurons with carotid chemoreceptor inputs arborize in the rostral ventrolateral medulla. Am J Physiol Regul Integr Comp Physiol 270:R1273‐R1278, 1996a.
 216. Koshiya N , Guyenet PG . Tonic sympathetic chemoreflex after blockade of respiratory rhythmogenesis in the rat. J Physiol 491:859‐869, 1996b.
 217. Koshiya N , Huangfu D , Guyenet PG . Ventrolateral medulla and sympathetic chemoreflex in the rat. Brain Res 609:174‐184, 1993.
 218. Koshiya N , Numao Y . The central respiratory drive‐related discharge patterns in the sympathetic nerves of the rat in unanesthetized and anesthetized states. Neurosci Res 7:S33, 1988.
 219. Koshiya N , Smith JC . Neuronal pacemaker for breathing visualized in vitro . Nature 400:360‐363, 1999.
 220. Krupp J , Feltz P . Excitatory postsynaptic currents and glutamate receptors in neonatal rat sympathetic preganglionic neurons in vitro. J Neurophysiol 73:1503‐1512, 1995.
 221. Krupp J , Larmet Y , Feltz P . Postnatal change of glycinergic IPSC decay in sympathetic preganglionic neurons. NeuroReport 5:2437‐2440, 1994.
 222. Kubin L , Alheid GF , Zuperku EJ , McCrimmon DR . Central pathways of pulmonary and lower airway vagal afferents. J Appl Physiol 101:618‐627, 2006.
 223. Kukkonen JP , Holmqvist T , Ammoun S , Åkerman KEO . Functions of the orexinergic/hypocretinergic system. Am J Physiol Cell Physiol 283:C1567‐C1591, 2002.
 224. Kumar P , Prabhakar NR . Peripheral chemoreceptors: Function and plasticity of the carotid body. Compr Physiol 2:141‐219, 2012.
 225. Kumar R , Ahdout R , Macey PM , Woo MA , Avedissian C , Thompson PM , Harper RM . Reduced caudate nuclei volumes in patients with congenital central hypoventilation syndrome. Neurosci 163:1373‐1379, 2009.
 226. Kuwaki T . Hypothalamic modulation of breathing. Adv Exp Med Biol 669:243‐247, 2010a.
 227. Kuwaki T . Orexin links emotional stress to autonomic functions. Auton Neurosci 161:20‐27, 2010b.
 228. Kuwaki T , Zhang W . Orexin neurons as arousal‐associated modulators of central cardiorespiratory regulation. Respir Physiol Neurobiol 174:43‐54, 2010.
 229. Lambert GW , Kaye DM , Lefkovits J , Jennings GL , Turner AG , Cox HS , Esler MD . Increased central nervous system monoamine neurotransmitter turnover and its association with sympathetic nervous activity in treated heart failure patients. Circulation 92:1813‐1818, 1995.
 230. Lavezzi AM , Weese‐Mayer DE , Yu MY , Jennings LJ , Corna MF , Casale V , Oneda R , Matturri L . Developmental alterations of the respiratory human retrotrapezoid nucleus in sudden unexplained fetal and infant death. Auton Neurosci 170:12‐19, 2012.
 231. Lazarenko RM , Milner TA , Depuy SD , Stornetta RL , West GH , Kievits JA , Bayliss DA , Guyenet PG . Acid sensitivity and ultrastructure of the retrotrapezoid nucleus in Phox2b‐EGFP transgenic mice. J Comp Neurol 517:69‐86, 2009.
 232. Lazarenko RM , Stornetta RL , Bayliss DA , Guyenet PG . Orexin A activates retrotrapezoid neurons in mice. Respir Physiol Neurobiol 175:283‐287, 2011.
 233. Lee MG , Hassani OK , Jones BE . Discharge of identified orexin/hypocretin neurons across the sleep‐waking cycle. J Neurosci 25:6716‐6720, 2005.
 234. Lein ES , Hawrylycz MJ , Ao N , Ayres M , Bensinger A , Bernard A , Boe AF , Boguski MS , Brockway KS , Byrnes EJ , Chen L , Chen L , Chen TM , Chin MC , Chong J , Crook BE , Czaplinska A , Dang CN , Datta S , Dee NR , Desaki AL , Desta T , Diep E , Dolbeare TA , Donelan MJ , Dong HW , Dougherty JG , Duncan BJ , Ebbert AJ , Eichele G , Estin LK , Faber C , Facer BA , Fields R , Fischer SR , Fliss TP , Frensley C , Gates SN , Glattfelder KJ , Halverson KR , Hart MR , Hohmann JG , Howell MP , Jeung DP , Johnson RA , Karr PT , Kawal R , Kidney JM , Knapik RH , Kuan CL , Lake JH , Laramee AR , Larsen KD , Lau C , Lemon TA , Liang AJ , Liu Y , Luong LT , Michaels J , Morgan JJ , Morgan RJ , Mortrud MT , Mosqueda NF , Ng LL , Ng R , Orta GJ , Overly CC , Pak TH , Parry SE , Pathak SD , Pearson OC , Puchalski RB , Riley ZL , Rockett HR , Rowland SA , Royall JJ , Ruiz MJ , Sarno NR , Schaffnit K , Shapovalova NV , Sivisay T , Slaughterbeck CR , Smith SC , Smith KA , Smith BI , Sodt AJ , Stewart NN , Stumpf KR , Sunkin SM , Sutram M , Tam A , Teemer CD , Thaller C , Thompson CL , Varnam LR , Visel A , Whitlock RM , Wohnoutka PE , Wolkey CK , Wong VY , Wood M , Yaylaoglu MB , Young RC , Youngstrom BL , Yuan XF , Zhang B , Zwingman TA , Jones AR . Genome‐wide atlas of gene expression in the adult mouse brain. Nature 445:168‐176, 2007.
 235. Lesage F , Barhanin J . Molecular physiology of pH‐sensitive background K(2P) channels. Physiology (Bethesda) 26:424‐437, 2011.
 236. 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.
 237. Li A , Nattie E . Catecholamine neurones in rats modulate sleep, breathing, central chemoreception and breathing variability. J Physiol 570:385‐396, 2006.
 238. Li A , Nattie E . Antagonism of rat orexin receptors by almorexant attenuates central chemoreception in wakefulness in the active period of the diurnal cycle. J Physiol 588:2935‐2944, 2010.
 239. Li A , Nattie EE . Focal central chemoreceptor sensitivity in the RTN studied with a CO2 diffusion pipette in vivo. J Appl Physiol 83:420‐428, 1997.
 240. Li A , Randall M , Nattie EE . CO(2) microdialysis in retrotrapezoid nucleus of the rat increases breathing in wakefulness but not in sleep. J Appl Physiol 87:910‐919, 1999.
 241. Li N , Li A , Nattie E . Focal microdialysis of CO(2) in the perifornical‐hypothalamic area increases ventilation during wakefulness but not NREM sleep. Respir Physiol Neurobiol 185:349‐355, 2013.
 242. 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.
 243. Li YW , Bayliss DA , Guyenet PG . C1 neurons of neonatal rats: Intrinsic beating properties and α2‐adrenergic receptors. Am J Physiol Regul Integr Comp Physiol 269:R1356‐R1369, 1995.
 244. Lindqvist N , Liu Q , Zajadacz J , Franze K , Reichenbach A . Retinal glial (Muller) cells: Sensing and responding to tissue stretch. Invest Ophthalmol Vis Sci 51:1683‐1690, 2010.
 245. Lindsay AD , Feldman JL . Modulation of respiratory activity of neonatal rat phrenic motoneurones by serotonin. J Physiol 461:213‐233, 1993.
 246. Lindsey BG , Rybak IA , Smith JC . Computational models and emergent properties of respiratory neural networks. Compr Physiol 2:1619‐1670, 2012.
 247. Lipscomb WT , Boyarsky LL . Neurophysiological investigations of medullary chemosensitive areas of respiration. Respir Physiol 16:362‐376, 1972.
 248. Lipski J , Kanjhan R , Kruszewska B , Rong WF . Properties of presympathetic neurones in the rostral ventrolateral medulla in the rat: An intracellular study ‘in vivo’. J Physiol 490:729‐744, 1996.
 249. Lipski J , Zhang X , Kruszewska B , Kanjhan R . Morphological study of long axonal projections of ventral medullary inspiratory neurons in the rat. Brain Res 640:171‐184, 1994.
 250. Llewellyn‐Smith IJ , Martin CL , Marcus JN , Yanagisawa M , Minson JB , Scammell TE . Orexin‐immunoreactive inputs to rat sympathetic preganglionic neurons. Neurosci Lett 351:115‐119, 2003.
 251. Loeschcke HH , Koepchen HP , GERTZ KH . Effect of hydrogen ion concentration and carbon dioxide pressure in the cerebrospinal fluid on respiration. Pflugers Arch 266:569‐585, 1958.
 252. Loewy AD , Spyer KM. Central Autonomic Pathways. In Central Regulation of Autonomic Functions. New York: Oxford University Press, 1990, pp. 88‐103.
 253. Lonergan T , Teschemacher AG , Hwang DY , Kim KS , Pickering AE , Kasparov S . Targeting brain stem centers of cardiovascular control using adenoviral vectors: Impact of promoters on transgene expression. Physiol Genomics 20:165‐172, 2005.
 254. Lugliani R , Whipp BJ , Seard C , Wasserman K . Effect of bilateral carotid‐body resection on ventilatory control at rest and during exercise in man. N Engl J Med 285:1105‐1111, 1971.
 255. Luiten PGM , Ter Horst GJ , Karst H , Steffens AB . The course of paraventricular hypothalamic efferents to autonomic structures in medulla and spinal cord. Brain Res 329:374‐378, 1985.
 256. Macey PM , Macey KE , Woo MA , Keens TG , Harper RM . Aberrant neural responses to cold pressor challenges in congenital central hypoventilation syndrome. Pediatr Res 57:500‐509, 2005.
 257. Macey PM , Richard CA , Kumar R , Woo MA , Ogren JA , Avedissian C , Thompson PM , Harper RM . Hippocampal volume reduction in congenital central hypoventilation syndrome. PLoS One 4:e6436, 2009.
 258. 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.
 259. Mack SO , Wu M , Kc P , Haxhiu MA . Stimulation of the hypothalamic paraventricular nucleus modulates cardiorespiratory responses via oxytocinergic innervation of neurons in pre‐Botzinger complex. J Appl Physiol 102:189‐199, 2007.
 260. Madden CJ , Morrison SF . Hypoxic activation of arterial chemoreceptors inhibits sympathetic outflow to brown adipose tissue in rats. J Physiol 566:559‐573, 2005.
 261. Madden CJ , Stocker SD , Sved AF . Attenuation of homeostatic responses to hypotension and glucoprivation after destruction of catecholaminergic rostral ventrolateral medulla (RVLM) neurons. Am J Physiol Regul Integr Comp Physiol 291:R751‐R759, 2006.
 262. Madden CJ , Sved AF . Cardiovascular regulation after destruction of the C1 cell group of the rostral ventrolateral medulla in rats. Am J Physiol Heart Circ Physiol 285:H2734‐H2748, 2003.
 263. Madden CJ , Tupone D , Cano G , Morrison SF . alpha2 adrenergic receptor‐mediated inhibition of thermogenesis. J Neurosci 33:2017‐2028, 2013.
 264. Magosso E , Ursino M . A mathematical model of CO2 effect on cardiovascular regulation. Am J Physiol Heart Circ Physiol 281:H2036‐H2052, 2001.
 265. Mandel DA , Schreihofer AM . Central respiratory modulation of barosensitive neurones in rat caudal ventrolateral medulla. J Physiol 572:881‐896, 2006.
 266. Mantyh PW , DeMaster E , Malhotra A , Ghilardi JR , Rogers SD , Mantyh CR , Liu H , Basbaum AI , Vigna SR , Maggio JE , Simone DA . Receptor endocytosis and dendrite reshaping in spinal neurons after somatosensory stimulation. Science 268:1629‐1632, 1995.
 267. Marcus NJ , Del Rio R ., Schultz EP, Xia XH, Schultz HD. Carotid body denervation improves autonomic and cardiac function and attenuates disordered breathing in congestive heart failure. J Physiol 592:391‐408, 2014.
 268. Marina N , Abdala AP , Korsak A , Simms AE , Allen AM , Paton JF , Gourine AV . Control of sympathetic vasomotor tone by catecholaminergic C1 neurones of the rostral ventrolateral medulla oblongata. Cardiovasc Res 91:703‐710, 2011.
 269. Marina N , Abdala AP , Trapp S , Li A , Nattie EE , Hewinson J , Smith JC , Paton JF , Gourine AV . Essential role of Phox2b‐expressing ventrolateral brainstem neurons in the chemosensory control of inspiration and expiration. J Neurosci 30:12466‐12473, 2010.
 270. Marina N , Tang F , Figueiredo M , Mastitskaya S , Kasimov V , Mohamed‐Ali V , Roloff E , Teschemacher AG , Gourine AV , Kasparov S . Purinergic signalling in the rostral ventro‐lateral medulla controls sympathetic drive and contributes to the progression of heart failure following myocardial infarction in rats. Basic Res Cardiol 108:317, 2013.
 271. Marshall JM . Peripheral chemoreceptors and cardiovascular regulation. Physiol Rev 74:543‐594, 1994.
 272. Marshall JM . The Joan Mott Prize Lecture. The integrated response to hypoxia: From circulation to cells. Exp Physiol 84:449‐470, 1999.
 273. Mateika JH , Duffin J . A review of the control of breathing during exercise. Eur J Appl Physiol Occup Physiol 71:1‐27, 1995.
 274. McAllen RM . Actions of carotid chemoreceptors on subretrofacial bulbospinal neurons in the cat. J Auton Nerv Syst 40:181‐188, 1992.
 275. McAllen RM , Dampney RA . Vasomotor neurons in the rostral ventrolateral medulla are organized topographically with respect to type of vascular bed but not body region. Neurosci Lett 110:91‐96, 1990.
 276. McAllen RM , May CN , Shafton AD . Functional anatomy of sympathetic premotor cell groups in the medulla. Clin Exp Hypertens 17:209‐221, 1995.
 277. McAllen RM , Salo LM , Paton JF , Pickering AE . Processing of central and reflex vagal drives by rat cardiac ganglion neurones: An intracellular analysis. J Physiol 589:5801‐5818, 2011.
 278. McAllen RM , Spyer KM . The location of cardiac vagal preganglionic motoneurones in the medulla of the cat. J Physiol 258:187‐204, 1976.
 279. McAllen RM , Spyer KM . The baroreceptor input to cardiac vagal motoneurones. J Physiol 282:365‐374, 1978.
 280. McBryde FD , Abdala AP , Hendy EB , Pijacka W , Marvar P , Moraes DJ , Sobotka PA , Paton JF . The carotid body as a putative therapeutic target for the treatment of neurogenic hypertension. Nat Commun 4:2395, 2013.
 281. McCall RB , Aghajanian GK . Serotonergic facilitation of facial motoneuron excitation. Brain Res 169:11‐27, 1979.
 282. McDougall SJ , Peters JH , Andresen MC . Convergence of cranial visceral afferents within the solitary tract nucleus. J Neurosci 29:12886‐12895, 2009.
 283. McEwen BS . Stress, adaptation, and disease. Allostasis and allostatic load. Ann N Y Acad Sci 840:33‐44, 1998.
 284. Meigh L , Greenhalgh SA , Rodgers TL , Cann MJ , Roper DI , Dale N . CO2 directly modulates connexin 26 by formation of carbamate bridges between subunits. Elife 22:e01213, 2013.
 285. Miller JR , Neumueller S , Muere C , Olesiak S , Pan L , Hodges MR , Forster HV . Changes in neurochemicals within the ventrolateral medullary respiratory column in awake goats after carotid body denervation. J Appl Physiol (1985) 115:1088‐1098, 2013.
 286. Milner TA , Pickel VM , Morrison SF , Reis DJ . Adrenergic neurons in the rostral ventrolateral medulla: Ultrastructure and synaptic relations with other transmitter‐ identified neurons. Prog Brain Res 81:29‐47, 1989.
 287. Mironov SL , Langohr K , Haller M , Richter DW . Hypoxia activates ATP‐dependent potassium channels in inspiratory neurones of neonatal mice. J Physiol 509:755‐766, 1998.
 288. Mitchell RA , Loeschcke HH , Massion WH , Severinghaus JW . Respiratory responses mediated through superficial chemosensitive areas on the medulla. J Appl Physiol 18:523‐533, 1963.
 289. Mo N , Dun NJ . Excitatory postsynaptic potentials in neonatal rat sympathetic preganglionic neurons: Possible mediation by NMDA receptors. Neurosci Lett 77:327‐332, 1987.
 290. Molkov YI , Zoccal DB , Moraes DJ , Paton JF , Machado BH , Rybak IA . Intermittent hypoxia‐induced sensitization of central chemoreceptors contributes to sympathetic nerve activity during late expiration in rats. J Neurophysiol 105:3080‐3091, 2011.
 291. Monnier A , Alheid GF , McCrimmon DR . Defining ventral medullary respiratory compartments with a glutamate receptor agonist in the rat. J Physiol 548:859‐874, 2003.
 292. Moosavi SH , Golestanian E , Binks AP , Lansing RW , Brown R , Banzett RB . Hypoxic and hypercapnic drives to breathe generate equivalent levels of air hunger in humans. J Appl Physiol 94:141‐154, 2003.
 293. Moraes DJ , da Silva MP , Bonagamba LG , Mecawi AS , Zoccal DB , Autunes‐Rodrigues J , Varanda WA , Machado BH . Electrophysiological properties of rostral ventrolateral medulla presympathetic neurons modulated by the respiratory network in rats. J Neurosci 33:19223‐19237, 2013.
 294. Moreira TS , Takakura AC , Colombari E , Guyenet PG . Central chemoreceptors and sympathetic vasomotor outflow. J Physiol 577:369‐386, 2006.
 295. Moreira TS , Takakura AC , Colombari E , West GH , Guyenet PG . Inhibitory input from slowly adapting lung stretch receptors to retrotrapezoid nucleus chemoreceptors. J Physiol 580:285‐300, 2007.
 296. Morgado‐Valle C , Baca SM , Feldman JL . Glycinergic pacemaker neurons in preBotzinger complex of neonatal mouse. J Neurosci 30:3634‐3639, 2010.
 297. Morrison SF. 2010 Carl Ludwig distinguished lectureship of the APS neural control and autonomic regulation section: Central neural pathways for thermoregulatory cold defense. J Appl Physiol 110:1137‐1149, 2011.
 298. Mouradian GC , Forster HV , Hodges MR . Acute and chronic effects of carotid body denervation on ventilation and chemoreflexes in three rat strains. J Physiol 590:3335‐3347, 2012.
 299. Mulkey DK , Rosin DL , West G , Takakura AC , Moreira TS , Bayliss DA , Guyenet PG . Serotonergic neurons activate chemosensitive retrotrapezoid nucleus neurons by a pH‐independent mechanism. J Neurosci 27:14128‐14138, 2007.
 300. Mulkey DK , Stornetta RL , Weston MC , Simmons JR , Parker A , Bayliss DA , Guyenet PG . Respiratory control by ventral surface chemoreceptor neurons in rats. Nat Neurosci 7:1360‐1369, 2004.
 301. Mulkey DK , Talley EM , Stornetta RL , Siegel AR , West GH , Chen X , Sen N , Mistry AM , Guyenet PG , Bayliss DA . TASK channels determine pH sensitivity in select respiratory neurons but do not contribute to central respiratory chemosensitivity. J Neurosci 27:14049‐14058, 2007.
 302. Mulkey DK , Wenker IC . Astrocyte chemoreceptors: Mechanisms of H+ sensing by astrocytes in the retrotrapezoid nucleus and their possible contribution to respiratory drive. Exp Physiol 96:400‐406, 2011.
 303. Mulkey DK , Wenker IC , Kreneisz O . Current ideas on central chemoreception by neurons and glial cells in the retrotrapezoid nucleus. J Appl Physiol 108:1433‐1439, 2010.
 304. Muller UW , Dembowsky K , Czachurski J , Seller H . Tonic descending inhibition of the spinal cardio‐sympathetic reflex in the cat. J Auton Nerv Syst 23:111‐123, 1988.
 305. Nakamura K , Morrison SF . A thermosensory pathway that controls body temperature. Nat Neurosci 11:62‐71, 2008.
 306. Narkiewicz K , Pesek CA , van de Borne PJ , Kato M , Somers VK . Enhanced sympathetic and ventilatory responses to central chemoreflex activation in heart failure. Circulation 100:262‐267, 1999.
 307. Narkiewicz K , Somers VK . The sympathetic nervous system and obstructive sleep apnea: Implications for hypertension. J Hypertens 15:1613‐1619, 1997.
 308. Natarajan M , Morrison SF . Sympathoexcitatory CVLM neurons mediate responses to caudal pressor area stimulation. Am J Physiol Regul Integr Comp Physiol 279:R364‐R374, 2000.
 309. Nattie E . Julius H. Comroe, Jr., distinguished lecture: Central chemoreception: Then … and now. J Appl Physiol 110:1‐8, 2011.
 310. Nattie E , Li A . Muscimol dialysis in the retrotrapezoid nucleus region inhibits breathing in the awake rat. J Appl Physiol 89:153‐162, 2000.
 311. Nattie E , Li A . Central chemoreception is a complex system function that involves multiple brain stem sites. J Appl Physiol 106:1464‐1466, 2009.
 312. Nattie E , Li A . Central chemoreception in wakefulness and sleep: Evidence for a distributed network and a role for orexin. J Appl Physiol 108:1417‐1424, 2010.
 313. Nattie E , Shi J , Li A . Bicuculline dialysis in the retrotrapezoid nucleus (RTN) region stimulates breathing in the awake rat. Resp Physiol 124:179‐193, 2001.
 314. Nattie EE . Chemoreception and tonic drive in the retrotrapezoid nucleus (RTN) region of the awake rat: Bicuculline and muscimol dialysis in the RTN. Adv Exp Med Biol 499:27‐32, 2001.
 315. Nattie EE , Gdovin M , Li A . Retrotrapezoid nucleus glutamate receptors: Control of CO2‐sensitive phrenic and sympathetic output. J Appl Physiol 74:2958‐2968, 1993.
 316. Nattie EE , Li A . Substance P‐saporin lesion of neurons with NK1 receptors in one chemoreceptor site in rats decreases ventilation and chemosensitivity. J Physiol 544:603‐616, 2002.
 317. Nattie EE , Li A . Multiple central chemoreceptor sites: Cell types and function in vivo. Adv Exp Med Biol 605:343‐347, 2008.
 318. Nattie EE , Li AH . Fluorescence location of RVLM kainate microinjections that alter the control of breathing. J Appl Physiol 68:1157‐1166, 1990.
 319. Nattie EE , Li AH , St John WM . Lesions in retrotrapezoid nucleus decrease ventilatory output in anesthetized or decerebrate cats. J Appl Physiol 71:1364‐1375, 1991.
 320. Neff RA , Simmens SJ , Evans C , Mendelowitz D . Prenatal nicotine exposure alters central cardiorespiratory responses to hypoxia in rats: Implications for sudden infant death syndrome. J Neurosci 24:9261‐9268, 2004.
 321. Neff RA , Wang J , Baxi S , Evans C , Mendelowitz D . Respiratory sinus arrhythmia: Endogenous activation of nicotinic receptors mediates respiratory modulation of brainstem cardioinhibitory parasympathetic neurons. Circulation Research 93:565‐572, 2003.
 322. Neverova NV , Saywell SA , Nashold LJ , Mitchell GS , Feldman JL . Episodic stimulation of alpha1‐adrenoreceptors induces protein kinase C‐dependent persistent changes in motoneuronal excitability. J Neurosci 27:4435‐4442, 2007.
 323. Numao Y , Koshiya N , Gilbey MP , Spyer KM . Central respiratory drive‐related activity in sympathetic nerves of the rat: The regional differences. Neurosci Lett 81:279‐284, 1987.
 324. Nurse CA . Neurotransmitter and neuromodulatory mechanisms at peripheral arterial chemoreceptors. Exp Physiol 95:657‐667, 2010.
 325. Okada Y , Chen Z , Jiang W , Kuwana S , Eldridge FL . Anatomical arrangement of hypercapnia‐activated cells in the superficial ventral medulla of rats. J Appl Physiol 93:427‐439, 2002.
 326. Onimaru H , Homma I . A novel functional neuron group for respiratory rhythm generation in the ventral medulla. J Neurosci 23:1478‐1486, 2003.
 327. Onimaru H , Ikeda K , Kawakami K . CO2‐Sensitive Preinspiratory Neurons of the Parafacial Respiratory Group Express Phox2b in the Neonatal Rat. J Neurosci 28:12845‐12850, 2008.
 328. Onimaru H , Ikeda K , Kawakami K . Postsynaptic mechanisms of CO2 responses in parafacial respiratory neurons of newborn rats. J Physiol 590:1615‐1624, 2012.
 329. Onimaru H , Ikeda K , Kawakami K . Relationship between the distribution of Phox2b expressing cells and blood vessels in the parafacial region of the ventral medulla of neonatal rats. Neurosci 212:131‐139, 2012.
 330. Pagliardini S , Janczewski WA , Tan W , Dickson CT , Deisseroth K , Feldman JL . Active expiration induced by excitation of ventral medulla in adult anesthetized rats. J Neurosci 31:2895‐2905, 2011.
 331. Paton JF , Abdala AP , Koizumi H , Smith JC , St‐John WM . Respiratory rhythm generation during gasping depends on persistent sodium current. Nat Neurosci 9:311‐313, 2006.
 332. Paton JF , Dickinson CJ , Mitchell G . Harvey Cushing and the regulation of blood pressure in giraffe, rat and man: Introducing ‘Cushing's mechanism’. Exp Physiol 94:11‐17, 2009.
 333. Paton JFR . Convergence properties of solitary tract neurones driven synaptically by cardiac vagal afferents in the mouse. J Physiol 508:237‐252, 1998.
 334. Paton JFR , Deuchars J , Li YW , Kasparov S . Properties of solitary tract neurones responding to peripheral arterial chemoreceptors. Neurosci 105:231‐248, 2001.
 335. 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 104:1287‐1294, 2008.
 336. Pena F , Parkis MA , Tryba AK , Ramirez JM . Differential contribution of pacemaker properties to the generation of respiratory rhythms during normoxia and hypoxia. Neuron 43:105‐117, 2004.
 337. Pena F , Ramirez JM . Endogenous activation of serotonin‐2A receptors is required for respiratory rhythm generation in vitro. J Neurosci 22:11055‐11064, 2002.
 338. Phillipson EA , Kozar LF , Rebuck AS , Murphy E . Ventilatory and waking responses to CO2 in sleeping dogs. Am Rev Respir Dis 115:251‐259, 1977.
 339. Pinault D . A novel single‐cell staining procedure performed in vivo under electrophysiological control: Morpho‐functional features of juxtacellularly labeled thalamic cells and other central neurons with biocytin or Neurobiotin. J Neurosci Methods 65:113‐136, 1996.
 340. Pineda J , Aghajanian GK . Carbon dioxide regulates the tonic activity of locus coeruleus neurons by modulating a proton‐ and polyamine‐sensitive inward rectifier potassium current. Neurosci 77:723‐743, 1997.
 341. Porzionato A , Macchi V , Stecco C , De CR . The carotid body in Sudden Infant Death Syndrome. Respir Physiol Neurobiol 185:194‐201, 2013.
 342. Prabhakar NR . Sensing hypoxia: Physiology, genetics and epigenetics. J Physiol 591:2245‐2257, 2013.
 343. 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.
 344. Ptak K , Yamanishi T , Aungst J , Milescu LS , Zhang R , Richerson GB , Smith JC . Raphe neurons stimulate respiratory circuit activity by multiple mechanisms via endogenously released serotonin and substance P. J Neurosci 29:3720‐3737, 2009.
 345. Queiroz EA , Okada MN , Fumega U , Fontes MA , Moraes MF , Haibara AS . Excitatory amino acid receptors in the dorsomedial hypothalamus are involved in the cardiovascular and behavioural chemoreflex responses. Exp Physiol 96:73‐84, 2011.
 346. Ramanantsoa N , Hirsch MR , Thoby‐Brisson M , Dubreuil V , Bouvier J , Ruffault PL , Matrot B , Fortin G , Brunet JF , Gallego J , Goridis C . Breathing without CO2 chemosensitivity in conditional Phox2b mutants. J Neurosci 31:12880‐12888, 2011.
 347. Ramirez JM , Doi A , Garcia AJ , III, Elsen FP , Koch H , Wei AD . The cellular building blocks of breathing. Compr Physiol 2:2683‐2731, 2012.
 348. Ramirez JM , Quellmalz UJ , Wilken B , Richter DW . The hypoxic response of neurones within the in vitro mammalian respiratory network. J Physiol 507:571‐582, 1998.
 349. Ray RS , Corcoran AE , Brust RD , Kim JC , Richerson GB , Nattie E , Dymecki SM . Impaired respiratory and body temperature control upon acute serotonergic neuron inhibition. Science 333:637‐642, 2011.
 350. Reis DJ , Golanov EV , Galea E , Feinstein DL . Central neurogenic neuroprotection: Central neural systems that protect the brain from hypoxia and ischemia. Ann N Y Acad Sci 835:168‐186, 1997.
 351. Reis DJ , Golanov EV , Ruggiero DA , Sun M‐K . Sympatho‐excitatory neurons of the rostral ventrolateral medulla are oxygen sensors and essential elements in the tonic and reflex control of the systemic and cerebral circulations. J Hypertens 12 Suppl. 10:S159‐S180, 1994.
 352. Reyes R , Duprat F , Lesage F , Fink M , Salinas M , Farman N , Lazdunski M . Cloning and expression of a novel pH‐sensitive two pore domain K+ channel from human kidney. J Biol Chem 273:30863‐30869, 1998.
 353. Richerson GB . Serotonergic neurons as carbon dioxide sensors that maintain pH homeostasis. Nat Rev Neurosci 5:449‐461, 2004.
 354. Richter DW . Commentary on eupneic breathing patterns and gasping. Resp Physiol 139:121‐130, 2003.
 355. Ritucci NA , Erlichman JS , Leiter JC , Putnam RW . Response of membrane potential and intracellular pH to hypercapnia in neurons and astrocytes from rat retrotrapezoid nucleus. Am J Physiol Regul Integr Comp Physiol 289:R851‐R861, 2005.
 356. 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.
 357. Ross CA , Ruggiero DA , Park DH , Joh TH , Sved AF , Fernandez‐Pardal J , Saavedra JM , Reis DJ . Tonic vasomotor control by the rostral ventrolateral medulla: Effect of electrical or chemical stimulation of the area containing C1 adrenaline neurons on arterial pressure, heart rate, and plasma catecholamines and vasopressin. J Neurosci 4:474‐494, 1984.
 358. Rudzinski E , Kapur RP . PHOX2B immunolocalization of the candidate human retrotrapezoid nucleus. Pediatr Dev Pathol 13:291‐299, 2010.
 359. Sagar SM , Sharp FR , Curran T . Expression of c‐fos protein in brain: Metabolic mapping at the cellular level. Science 240:1328‐1330, 1988.
 360. Sakurai T , Mieda M , Tsujino N . The orexin system: Roles in sleep/wake regulation. Ann N Y Acad Sci 1200:149‐161, 2010.
 361. Santin JM , Watters KC , Putnam RW , Hartzler LK . Temperature influences neuronal activity and CO2/pH sensitivity of locus coeruleus neurons in the bullfrog, Lithobates catesbeianus. Am J Physiol Regul Integr Comp Physiol 305:R1451‐R1464, 2013.
 362. Saper CB , Fuller PM , Pedersen NP , Lu J , Scammell TE . Sleep state switching. Neuron 68:1023‐1042, 2010.
 363. Sartor DM , Verberne AJ . The sympathoinhibitory effects of systemic cholecystokinin are dependent on neurons in the caudal ventrolateral medulla in the rat. Am J Physiol Regul Integr Comp Physiol 291:R1390‐R1398, 2006.
 364. Sato A , Fidone S , Eyzaguirre C . Presence of chemoreceptor and baroreceptor C‐fibers in the carotid nerve of the cat. Brain Res 11:459‐463, 1968.
 365. Sato M , Severinghaus JW , Basbaum AI . Medullary CO2 chemoreceptor neuron identification by c‐fos immunocytochemistry. J Appl Physiol 73:96‐100, 1992.
 366. Sawchenko PE , Li HY , Ericsson A . Circuits and mechanisms governing hypothalamic responses to stress: A tale of two paradigms. Prog Brain Res 122:61‐78, 2000.
 367. Saywell SA , Anissimova NP , Ford TW , Meehan CF , Kirkwood PA . The respiratory drive to thoracic motoneurones in the cat and its relation to the connections from expiratory bulbospinal neurones. J Physiol 579:765‐782, 2007.
 368. Schlaefke ME , See WR , Loeschcke HH . Ventilatory response to alterations of H+ ion concentration in small areas of the ventral medullary surface. Respir Physiol 10:198‐212, 1970.
 369. Schreihofer AM , Guyenet PG . Identification of C1 presympathetic neurons in rat rostral ventrolateral medulla by juxtacellular labeling in vivo. J Comp Neurol 387:524‐536, 1997.
 370. Schreihofer AM , Guyenet PG . Baroactivated neurons with pulse‐modulated activity in the rat caudal ventrolateral medulla express GAD67 mRNA. J Neurophysiol 89:1265‐1277, 2003.
 371. Schreihofer AM , Sved AF . The Ventrolateral Medulla and Sympathetic Regulation of Arterial Pressure. In Central Regulation of Autonomic Functions. Llewellyn‐Smith IJ, Verberne AJ. New York: Oxford University Press, 2011, pp. 78‐97.
 372. Schultz HD , Del Rio R , Ding Y , Marcus NJ . Role of neurotransmitter gases in the control of the carotid body in heart failure. Respir Physiol Neurobiol 184:197‐203, 2012.
 373. Schultz HD , Marcus NJ , Del Rio R . Role of the carotid body in the pathophysiology of heart failure. Curr Hypertens Rep 15:356‐362, 2013.
 374. Severinghaus JW . Hans Loeschcke, Robert Mitchell and the medullary CO2 chemoreceptors: A brief historical review. Respir Physiol 114:17‐24, 1998.
 375. Severson CA , Wang W , Pieribone VA , Dohle CI , Richerson GB . Midbrain serotonergic neurons are central pH chemoreceptors. Nat Neurosci 6:1139‐1140, 2003.
 376. Seyedabadi M , Li Q , Padley JR , Pilowsky PM , Goodchild AK . A novel pressor area at the medullo‐cervical junction that is not dependent on the RVLM: Efferent pathways and chemical mediators. J Neurosci 26:5420‐5427, 2006.
 377. Shea SA , Andres LP , Shannon DC , Banzett RB . Ventilatory responses to exercise in humans lacking ventilatory chemosensitivity. J Physiol 468:623‐640, 1993.
 378. Sica AL , Greenberg HE , Ruggiero DA , Scharf SM . Chronic‐intermittent hypoxia: A model of sympathetic activation in the rat. Respir Physiol 121:173‐184, 2000.
 379. Simms AE , Paton JF , Pickering AE , Allen AM . Amplified respiratory‐sympathetic coupling in the spontaneously hypertensive rat: Does it contribute to hypertension? J Physiol 587:597‐610, 2009.
 380. Smith CA , Forster HV , Blain GM , Dempsey JA . An interdependent model of central/peripheral chemoreception: Evidence and implications for ventilatory control. Respir Physiol Neurobiol 173:288‐297, 2010.
 381. Smith JC , Abdala AP , Borgmann A , Rybak IA , Paton JF . Brainstem respiratory networks: Building blocks and microcircuits. Trends Neurosci 36:152‐162, 2013.
 382. 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.
 383. Smith JC , Abdala AP , Rybak IA , Paton JF . Structural and functional architecture of respiratory networks in the mammalian brainstem. Philos Trans R Soc Lond B Biol Sci 364:2577‐2587, 2009.
 384. 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.
 385. Smith JC , Morrison DE , Ellenberger HH , Otto MR , Feldman JL . Brainstem projections to the major respiratory neuron populations in the medulla of the cat. J Comp Neurol 281:69‐96, 1989.
 386. Smith PM , Connolly BC , Ferguson AV . Microinjection of orexin into the rat nucleus tractus solitarius causes increases in blood pressure. Brain Res 950:261‐267, 2002.
 387. Soffin EM , Evans ML , Gill CH , Harries MH , Benham CD , davies CH . SB‐334867‐A antagonises orexin mediated excitation in the locus coeruleus. Neuropharmacology 42:127‐133, 2002.
 388. Solomon IC , Edelman NH , O'Neill MH . CO2/H+ chemoreception in the cat pre‐Botzinger complex in vivo. J Appl Physiol 88:1996‐2007, 2000.
 389. Song G , Poon CS . Lateral parabrachial nucleus mediates shortening of expiration during hypoxia. Respir Physiol Neurobiol 165:1‐8, 2009.
 390. Song G , Wang H , Xu H , Poon CS . Kolliker‐Fuse neurons send collateral projections to multiple hypoxia‐activated and nonactivated structures in rat brainstem and spinal cord. Brain Struct Funct 217:835‐858, 2012.
 391. Song G , Xu H , Wang H , Macdonald SM , Poon CS . Hypoxia‐excited neurons in NTS send axonal projections to Kolliker‐Fuse/parabrachial complex in dorsolateral pons. Neurosci 175:145‐153, 2011.
 392. Loewy AD , Spyer KM. The Central Nervous Organization of Reflex Circulatory Control. In Central Regulation of Autonomic Functions. Loewy AD, Spyer KM. New York: Oxford University Press, 1990, pp. 168‐188.
 393. Spyer KM , Gourine AV . Chemosensory pathways in the brainstem controlling cardiorespiratory activity. Philos Trans R Soc Lond B Biol Sci 364:2603‐2610, 2009.
 394. St Croix CM , Satoh M , Morgan BJ , Skatrud JB , Dempsey JA . Role of respiratory motor output in within‐breath modulation of muscle sympathetic nerve activity in humans. Circ Res 85:457‐469, 1999.
 395. St‐John WM , Stornetta RL , Guyenet PG , Paton JF . Location and properties of respiratory neurones with putative intrinsic bursting properties in the rat in situ. J Physiol 587:3175‐3188, 2009.
 396. Sterling P. Principles of Allostasis: Optimal Design, Predictive Regulation, Pathophysiology and Rational Therapeutics. In Allostasis, Homeostasis, and the Costs of Physiological Adaptation. Schulkin Jay. New York, NY: Cambridge University Press, 2004, pp. 1‐36.
 397. Stocker SD , Simmons JR , Stornetta RL , Toney GM , Guyenet PG . Water deprivation activates a glutamatergic projection from the hypothalamic paraventricular nucleus to the rostral ventrolateral medulla. J Comp Neurol 494:673‐685, 2006.
 398. Stojicic S , Milutinovic‐Smiljanic S , Sarenac O , Milosavljevic S , Paton JF , Murphy D , Japundzic‐Zigon N . Blockade of central vasopressin receptors reduces the cardiovascular response to acute stress in freely moving rats. Neuropharmacology 54:824‐836, 2008.
 399. Stornetta RL . Neurochemistry of bulbospinal presympathetic neurons of the medulla oblongata. J Chem Neuroanat 38:222‐230, 2009.
 400. Stornetta RL , McQuiston TJ , Guyenet PG . GABAergic and glycinergic presympathetic neurons of rat medulla oblongata identified by retrograde transport of pseudorabies virus and in situ hybridization. J Comp Neurol 479:257‐270, 2004.
 401. Stornetta RL , Moreira TS , Takakura AC , Kang BJ , Chang DA , West GH , Brunet JF , Mulkey DK , Bayliss DA , Guyenet PG . Expression of Phox2b by brainstem neurons involved in chemosensory integration in the adult rat. J Neurosci 26:10305‐10314, 2006.
 402. Stornetta RL , Schreihofer AM , Pelaez NM , Sevigny CP , Guyenet PG . Preproenkephalin mRNA is expressed by C1 and non‐C1 barosensitive bulbospinal neurons in the rostral ventrolateral medulla of the rat. J Comp Neurol 435:111‐126, 2001.
 403. Stornetta RL , Sevigny CP , Schreihofer AM , Rosin DL , Guyenet PG . Vesicular glutamate transporter DNPI/GLUT2 is expressed by both C1 adrenergic and nonaminergic presympathetic vasomotor neurons of the rat medulla. J Comp Neurol 444:207‐220, 2002.
 404. Stornetta RL , Spirovski D , Moreira TS , Takakura AC , West GH , Gwilt JM , Pilowsky PM , Guyenet PG . Galanin is a selective marker of the retrotrapezoid nucleus in rats. J Comp Neurol 512:373‐383, 2009.
 405. Strack AM , Loewy AD . Pseudorabies virus: A highly specific transneuronal cell body marker in the sympathetic nervous system. J Neurosci 10:2139‐2147, 1990.
 406. Strack AM , Sawyer WB , Hughes JH , Platt KB , Loewy AD . A general pattern of CNS innervation of the sympathetic outflow demonstrated by transneuronal pseudorabies viral infections. Brain Res 491:156‐162, 1989.
 407. Strack AM , Sawyer WB , Platt KB , Loewy AD . CNS cell groups regulating the sympathetic outflow to adrenal gland as revealed by transneuronal cell body labeling with pseudorabies virus. Brain Res 491:274‐296, 1989.
 408. Su CK , Ho CM , Kuo HH , Wen YC , Chai CY . Sympathetic‐correlated c‐Fos expression in the neonatal rat spinal cord in vitro. J Biomed Sci 16:44, 2009.
 409. Su J , Yang L , Zhang X , Rojas A , Shi Y , Jiang C . High CO2 chemosensitivity versus wide sensing spectrum: A paradoxical problem and its solutions in cultured brainstem neurons. J Physiol 578:831‐841, 2007.
 410. Sun MK , Hackett JT , Guyenet PG . Sympathoexcitatory neurons of rostral ventrolateral medulla exhibit pacemaker properties in the presence of a glutamate‐ receptor antagonist. Brain Res 438:23‐40, 1988.
 411. Sun MK , Jeske IT , Reis DJ . Cyanide excites medullary sympathoexcitatory neurons in rats. Am J Physiol 262:R182‐R189, 1992.
 412. Sun MK , Reis DJ . Central neural mechanisms mediating excitation of sympathetic neurons by hypoxia. Prog Neurobiol 44:197‐219, 1994a.
 413. Sun MK , Reis DJ . Hypoxia selectively excites vasomotor neurons of rostral ventrolateral medulla in rats. Am J Physiol Regul Integr Comp Physiol 266:R245‐R256, 1994b.
 414. Sun MK , Reis DJ . Medullary vasomotor activity and hypoxic sympathoexcitation in pentobarbital‐anesthetized rats. Am J Physiol Regul Integr Comp Physiol 270:R348‐R355, 1996.
 415. Sun MK , Wahlestedt C , Reis DJ . Action of externally applied ATP on rat reticulospinal vasomotor neurons. Eur J Pharmacol 224:93‐96, 1992.
 416. Sunanaga J , Deng BS , Zhang W , Kanmura Y , Kuwaki T . CO(2) activates orexin‐containing neurons in mice. Respir Physiol Neurobiol 166:184‐186, 2009.
 417. Taddese A , Bean BP . Subthreshold sodium current from rapidly inactivating sodium channels drives spontaneous firing of tuberomammillary neurons. Neuron 33:587‐600, 2002.
 418. Tafil‐Klawe M , Trzebski A , Klawe J , Palko T . Augmented chemoreceptor reflex tonic drive in early human hypertension and in normotensive subjects with family background of hypertension. Acta Physiol Pol 36:51‐58, 1985.
 419. Tahawi Z , Orolinova N , Joshua IG , Bader M , Fletcher EC . Altered vascular reactivity in arterioles of chronic intermittent hypoxic rats. J Appl Physiol 90:2007‐2013, 2001.
 420. Takakura AC , Moreira TS . Arterial chemoreceptor activation reduces the activity of parapyramidal serotonergic neurons in rats. Neurosci 237:199‐207, 2013.
 421. Takakura AC , Moreira TS , Colombari E , West GH , Stornetta RL , Guyenet PG . Peripheral chemoreceptor inputs to retrotrapezoid nucleus (RTN) CO2‐sensitive neurons in rats. J Physiol 572:503‐523, 2006.
 422. Takakura AC , Moreira TS , De Paula PM , Menani JV , Colombari E . Control of breathing and blood pressure by parafacial neurons in conscious rats. Exp Physiol 98: 304‐315, 2012.
 423. Takakura AC , Moreira TS , Stornetta RL , West GH , Gwilt JM , Guyenet PG . Selective lesion of retrotrapezoid Phox2b‐expressing neurons raises the apnoeic threshold in rats. J Physiol 586:2975‐2991, 2008.
 424. Takakura AC , Moreira TS , West GH , Gwilt JM , Colombari E , Stornetta RL , Guyenet PG . GABAergic pump cells of solitary tract nucleus innervate retrotrapezoid nucleus chemoreceptors. J Neurophysiol 98:374‐381, 2007.
 425. Tan EM , Yamaguchi Y , Horwitz GD , Gosgnach S , Lein ES , Goulding M , Albright TD , Callaway EM . Selective and quickly reversible inactivation of mammalian neurons in vivo using the Drosophila allatostatin receptor. Neuron 51:157‐170, 2006.
 426. Tarkan O , Sari P , Demirhan O , Kiroglu M , Tuncer U , Surmelioglu O , Ozdemir S , Yilmaz MB , Kara K . Connexin 26 and 30 mutations in paediatric patients with congenital, non‐syndromic hearing loss treated with cochlear implantation in Mediterranean Turkey. J Laryngol Otol 127:33‐37, 2013.
 427. Teppema LJ , Berkenbosch A , Veening JG , Olievier CN . Hypercapnia induces c‐fos expression in neurons of retrotrapezoid nucleus in cats. Brain Res 635:353‐356, 1994.
 428. Teppema LJ , Dahan A . The ventilatory response to hypoxia in mammals: Mechanisms, measurement, and analysis. Physiol Rev 90:675‐754, 2010.
 429. Teppema LJ , Smith CA . CrossTalk opposing view: Peripheral and central chemoreceptors have hyperadditive effects on respiratory motor control. J Physiol 591:4359‐4361, 2013.
 430. Teppema LJ , Veening JG , Kranenburg A , Dahan A , Berkenbosch A , Olievier C . Expression of c‐fos in the rat brainstem after exposure to hypoxia and to normoxic and hyperoxic hypercapnia. J Comp Neurol 388:169‐190, 1997.
 431. 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 12:1028‐1035, 2009.
 432. Thomas T , Ralevic V , Gadd CA , Spyer KM . Central CO2 chemoreception: A mechanism involving P2 purinoceptors localized in the ventrolateral medulla of the anaesthetized rat. J Physiol 517:899‐905, 1999.
 433. Tian JB , Bishop GA . Stimulus‐dependent activation of c‐Fos in neurons and glia in the rat cerebellum. J Chem Neuroanat 23:157‐170, 2002.
 434. Tin C , Song G , Poon CS . Hypercapnia attenuates inspiratory amplitude and expiratory time responsiveness to hypoxia in vagotomized and vagal‐intact rats. Respir Physiol Neurobiol 181:79‐87, 2012.
 435. Topor ZL , Johannson L , Kasprzyk J , Remmers JE . Dynamic ventilatory response to CO(2) in congestive heart failure patients with and without central sleep apnea. J Appl Physiol 91:408‐416, 2001.
 436. Toward MA , Abdala AP , Knopp SJ , Paton JF , Bissonnette JM . Increasing brain serotonin corrects CO2 chemosensitivity in methyl‐CpG‐binding protein 2 (Mecp2)‐deficient mice. Exp Physiol 98:842‐849, 2013.
 437. Tryba AK , Pena F , Ramirez JM . Gasping activity in vitro: A rhythm dependent on 5‐HT2A receptors. J Neurosci 26:2623‐2634, 2006.
 438. Ursino M , Magosso E , Avanzolini G . An integrated model of the human ventilatory control system: The response to hypercapnia. Clin Physiol 21:447‐464, 2001.
 439. Vardhan A , Kachroo A , Sapru HN . Excitatory amino acid receptors in commissural nucleus of the NTS mediate carotid chemoreceptor responses. Am J Physiol 264:R41‐R50, 1993.
 440. Veasey SC , Fornal CA , Metzler CW , Jacobs BL . Response of serotonergic caudal raphe neurons in relation to specific motor activities in freely moving cats. J Neurosci 15:5346‐5359, 1995.
 441. Veasey SC , Fornal CA , Metzler CW , Jacobs BL . Single‐unit responses of serotonergic dorsal raphe neurons to specific motor challenges in freely moving cats. Neurosci 79:161‐169, 1997.
 442. Viemari JC , Garcia AJ, III , Doi A , Elsen G , Ramirez JM . beta‐Noradrenergic receptor activation specifically modulates the generation of sighs in vivo and in vitro. Front Neural Circuits 7:1‐14, 2013.
 443. Wade JG , Larson CP, Jr. , Hickey RF , Ehrenfeld WK , Severinghaus JW . Effect of carotid endarterectomy on carotid chemoreceptor and baroreceptor function in man. N Engl J Med 282:823‐829, 1970.
 444. Wang HJ , Li YL , Zucker IH , Wang W . Exercise training prevents skeletal muscle afferent sensitization in rats with chronic heart failure. Am J Physiol Regul Integr Comp Physiol 302:R1260‐R1270, 2012.
 445. Wang S , Benamer N , Zanella S , Kumar NN , Shi Y , Bevengut M , Penton D , Guyenet PG , Lesage F , Gestreau C , Barhanin J , Bayliss DA . TASK‐2 channels contribute to pH sensitivity of retrotrapezoid nucleus chemoreceptor neurons. J Neurosci 33:16033‐16044, 2013.
 446. Wang S , Shi Y , Shu S , Guyenet PG , Bayliss DA . Phox2b‐expressing retrotrapezoid neurons are intrinsically responsive to acidification and CO2 . J Neurosci 33:7756‐7761, 2013.
 447. Wang W , Pizzonia JH , Richerson GB . Chemosensitivity of rat medullary raphe neurones in primary tissue culture. J Physiol 511:433‐450, 1998.
 448. Wang WA , Richerson GB . Chemosensitivity of non‐respiratory rat CNS neurons in tissue culture. Brain Res 860:119‐129, 2000.
 449. Wang WG , Tiwari JK , Bradley SR , Zaykin AV , Richerson GB . Acidosis‐stimulated neurons of the medullary raphe are serotonergic. J Neurophysiol 85:2224‐2235, 2001.
 450. Washburn CP , Bayliss DA , Guyenet PG . Cardiorespiratory neurons of the rat ventrolateral medulla contain TASK‐1 and TASK‐3 channel mRNA. Respir Physiol Neurobiol 138:19‐35, 2003.
 451. Weese‐Mayer DE , Berry‐Kravis EM , Ceccherini I , Keens TG , Loghmanee DA , Trang H . An official ATS clinical policy statement: Congenital central hypoventilation syndrome: Genetic basis, diagnosis, and management. Am J Respir Crit Care Med 181:626‐644, 2010.
 452. Weese‐Mayer DE , Rand CM , Berry‐Kravis EM , Jennings LJ , Loghmanee DA , Patwari PP , Ceccherini I . Congenital central hypoventilation syndrome from past to future: Model for translational and transitional autonomic medicine. Pediatr Pulmonol 44:521‐535, 2009.
 453. Wenker IC , Kreneisz O , Nishiyama A , Mulkey DK . Astrocytes in the retrotrapezoid nucleus sense H+ by inhibition of a Kir4.1‐Kir5.1‐like current and may contribute to chemoreception by a purinergic mechanism. J Neurophysiol 104:3042‐3052, 2010.
 454. Wenker IC , Sobrinho CR , Takakura AC , Moreira TS , Mulkey DK . Regulation of ventral surface CO2/H+‐sensitive neurons by purinergic signalling. J Physiol 590:2137‐2150, 2012.
 455. Wenker IC , Sobrinho CR , Takakura AC , Mulkey DK , Moreira TS . P2Y1 receptors expressed by C1 neurons determine peripheral chemoreceptor modulation of breathing, sympathetic activity, and blood pressure. Hypertension 62:263‐273, 2013.
 456. Weston MC , Stornetta RL , Guyenet PG . Glutamatergic neuronal projections from the marginal layer of the rostral ventral medulla to the respiratory centers in rats. J Comp Neurol 473:73‐85, 2004.
 457. Willette RN , Barcas PP , Krieger AJ , Sapru HN . Vasopressor and depressor areas in the rat medulla. Identification by microinjection of L‐glutamate. Neuropharmacology 22:1071‐1079, 1983.
 458. Williams RH , Jensen LT , Verkhratsky A , Fugger L , Burdakov D . Control of hypothalamic orexin neurons by acid and CO2 . Proc Natl Acad Sci USA 104:10685‐10690, 2007.
 459. Wilson RJ , Day TA . CrossTalk opposing view: Peripheral and central chemoreceptors have hypoadditive effects on respiratory motor output. J Physiol 591:4355‐4357, 2013.
 460. Wittmeier S , Song G , Duffin J , Poon CS . Pacemakers handshake synchronization mechanism of mammalian respiratory rhythmogenesis. Proc Natl Acad Sci USA 105:18000‐18005, 2008.
 461. Xie A , Skatrud JB , Morgan BJ , Chenuel B , Khayat R , Reichmuth K , Lin J , Dempsey JA . Influence of cerebrovascular function on the hypercapnic ventilatory response in healthy humans. J Physiol 577:319‐329, 2006.
 462. Yang Z , Coote JH . The role of supra‐spinal vasopressin and glutamate neurones in an increase in renal sympathetic activity in response to mild haemorrhage in the rat. Exp Physiol 91:791‐797, 2006.
 463. 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.
 464. Zhang W , Carreno FR , Cunningham JT , Mifflin SW . Chronic sustained and intermittent hypoxia reduce function of ATP‐sensitive potassium channels in nucleus of the solitary tract. Am J Physiol Regul Integr Comp Physiol 295:R1555‐R1562, 2008.
 465. Zhang W , Fukuda Y , Kuwaki T . Respiratory and cardiovascular actions of orexin‐A in mice. Neurosci Lett 385:131‐136, 2005.
 466. Zoccal DB , Machado BH . Coupling between respiratory and sympathetic activities as a novel mechanism underpinning neurogenic hypertension. Curr Hypertens Rep 13:229‐236, 2011.
 467. 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.
 468. Zwicker JD , Rajani V , Hahn LB , Funk GD . Purinergic modulation of preBotzinger complex inspiratory rhythm in rodents: The interaction between ATP and adenosine. J Physiol 589:4583‐4600, 2011.

Further Reading

Dutschmann M, Dick TE. Pontine mechanisms of respiratory control. Compr Physiol 2:2443-2469,2012

Forster HV, Haouzi P, Dempsey JA. Control of breathing during exercise. Compr Physiol 2:743-777,2012

Funk GD. Neuromodulation: purinergic signaling in respiratory control. Compr Physiol 3:331-363,2013

Gallego J. Genetic diseases: congenital central hypoventilation, Rett, and Prader-Willi syndromes. Compr Physiol 2:2255-2279,2012

Horner RL. Neural control of the upper airway: integrative physiological mechanisms and relevance for sleep disordered breathing. Compr Physiol 2:479-535,2012

Javaheri S, Dempsey JA. Central sleep apnea. Compr Physiol 3:141-163,2013

White DP, Younes MK. Obstructive sleep apnea. Compr Physiol 2:2541-2594,2012

Zhang SX, Wang Y, Gozal D. Pathological consequences of intermittent hypoxia in the central nervous system. Compr Physiol 2:1767-1777,2012


Related Articles:

Central Chemoreceptors: Locations and Functions
Peripheral Chemoreceptors: Function and Plasticity of the Carotid Body
The Cellular Building Blocks of Breathing
Interaction of Cardiovascular Reflexes in Circulatory Control
Computational Models and Emergent Properties of Respiratory Neural Networks
Pontine Mechanisms of Respiratory Control
Control of Breathing During Exercise
Neuromodulation: Purinergic Signaling in Respiratory Control
Genetic Diseases: Congenital Central Hypoventilation, Rett, and Prader‐Willi Syndromes
Neural Control of the Upper Airway: Integrative Physiological Mechanisms and Relevance for Sleep Disordered Breathing
Central Sleep Apnea
Obstructive Sleep Apnea
Pathological Consequences of Intermittent Hypoxia in the Central Nervous System
Pulmonary Vascular Disease

Contact Editor

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

* Required Field

How to Cite

Patrice G. Guyenet. Regulation of Breathing and Autonomic Outflows by Chemoreceptors. Compr Physiol 2014, 4: 1511-1562. doi: 10.1002/cphy.c140004