Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2010 May 24;107(23):10719–10724. doi: 10.1073/pnas.1005866107

H2S mediates O2 sensing in the carotid body

Ying-Jie Peng a, Jayasri Nanduri a, Gayatri Raghuraman a, Dangjai Souvannakitti a, Moataz M Gadalla b, Ganesh K Kumar a, Solomon H Snyder b,c,d,1, Nanduri R Prabhakar a,1
PMCID: PMC2890835  PMID: 20556885

Abstract

Gaseous messengers, nitric oxide and carbon monoxide, have been implicated in O2 sensing by the carotid body, a sensory organ that monitors arterial blood O2 levels and stimulates breathing in response to hypoxia. We now show that hydrogen sulfide (H2S) is a physiologic gasotransmitter of the carotid body, enhancing its sensory response to hypoxia. Glomus cells, the site of O2 sensing in the carotid body, express cystathionine γ-lyase (CSE), an H2S-generating enzyme, with hypoxia increasing H2S generation in a stimulus-dependent manner. Mice with genetic deletion of CSE display severely impaired carotid body response and ventilatory stimulation to hypoxia, as well as a loss of hypoxia-evoked H2S generation. Pharmacologic inhibition of CSE elicits a similar phenotype in mice and rats. Hypoxia-evoked H2S generation in the carotid body seems to require interaction of CSE with hemeoxygenase-2, which generates carbon monoxide. CSE is also expressed in neonatal adrenal medullary chromaffin cells of rats and mice whose hypoxia-evoked catecholamine secretion is greatly attenuated by CSE inhibitors and in CSE knockout mice.

Keywords: cystathionine γ-lyase, hydrogen sulfide, hypoxia, hemeoxygenase-2


In adult mammals, carotid bodies are the sensory organs responsible for monitoring arterial blood O2 concentrations and relay sensory information to the brainstem neurons associated with regulation of breathing and the cardiovascular system (1). Carotid bodies are comprised mainly of two cell types: glomus (also called type I) and sustanticular (or type II) cells. Glomus cells, of neuronal nature, are considered the main hypoxia-sensing cells. The gaseous messengers, carbon monoxide (CO) and nitric oxide (NO), generated by hemeoxygenase-2 (HO-2) and neuronal nitric oxide synthase (nNOS), respectively, physiologically inhibit carotid body activity (24). Because HO-2 and nNOS require molecular O2 for their activity, stimulation of carotid body activity by hypoxia may reflect in part reduced formation of CO and NO (5).

Like NO and CO, hydrogen sulfide (H2S) is a gasotransmitter physiologically regulating neuronal transmission (6) and vascular tone (7). Cystathionine γ-lyase (CSE) (EC 4.4.1.1) and cystathionine β-synthase (CBS) (4.2.1.22) are the major enzymes associated with generation of endogenous H2S (8, 9). CBS is the predominant H2S-synthesizing enzyme in the brain, CSE preponderates in the peripheral tissues whose H2S levels are reduced 90% in CSE−/− mice (710). Given that carotid bodies are peripheral organs and that H2S is redox active, we hypothesized that CSE-derived H2S plays a role in hypoxic sensing by the carotid body. We examined carotid body response to hypoxia in wild-type (CSE+/+) and CSE−/− mice as well as in rats treated with CSE inhibitor. Genetic deletion or pharmacologic inhibition of CSE dramatically impairs hypoxic sensing by the carotid body as well as in neonatal adrenal medullary chromaffin cells (AMC).

Results

Loss of Carotid Body Response to Hypoxia in CSE−/− Mice.

CSE immunoreactivity was seen in glomus cells of carotid bodies from CSE+/+ mice as evidenced by colocalization with tyrosine hydroxylase (TH), an established marker of glomus cells (11, 12). CSE expression was absent in carotid bodies from CSE−/− mice (Fig. 1A). To assess the impact of CSE disruption on carotid body response to hypoxia, carotid bodies were isolated from CSE+/+ and mutant mice and “single” unit sensory discharges were recorded from the carotid sinus nerve. In CSE+/+ mice, hypoxia augmented the sensory activity in a stimulus-dependent manner (Fig. 1 B and C). In striking contrast, carotid bodies from CSE−/− mice exhibited severely impaired sensory response to hypoxia (Fig. 1 B and C). Similar loss of the hypoxic response was also seen in CSE+/+ mice treated systemically with DL-propargylglycine (PAG), an inhibitor of CSE (Fig. S1) (13, 14). On the other hand, acute application of PAG was ineffective in preventing the hypoxic response, a finding consistent with a recent report (15).

Fig. 1.

Fig. 1.

CSE localization in the mouse carotid body and carotid body responses to hypoxia and hypercapnia in CSE+/+ and CSE−/− mice. (A) CSE expression in carotid bodies from CSE+/+ and CSE−/− mice. Carotid body sections were stained with antibodies specific for CSE or tyrosine hydroxylase (TH), a marker of glomus cells. (Scale bar: 20 μm.) (B) Sensory response of isolated carotid bodies to hypoxia (Hx) (Inline graphic ∼ 39 mmHg; at black bar) in CSE +/+ and CSE−/− mice. Integrated carotid body sensory activity (CB activity) is presented as impulses per second (imp/s). Superimposed action potentials from the single fiber are presented in Inset. (C) Carotid body responses to graded hypoxia from CSE+/+ and CSE−/− mice, measured as the difference in response between baseline and hypoxia (Δimp/s). Data are mean ± SEM of n = 24 (CSE+/+) and n = 23 (CSE−/−) fibers from eight mice each. (D) H2S levels (mean ± SEM) in carotid bodies from CSE+/+ and CSE−/− mice under normoxia (NOR) and hypoxia (Hx) (Inline graphic ∼ 40 mmHg) from four independent experiments. (E) Example illustrating carotid body responses to CO2 Inline graphic in CSE+/+ and CSE−/− mice. (F) Average data (mean ± SEM) of CO2 response from n = 24 (CSE +/+) and n = 19 (CSE−/−) fibers from eight mice in each group. *** and **, P < 0.001 and P < 0.01, respectively; n.s. (not significant), P > 0.05.

H2S generation was monitored in carotid bodies from CSE+/+ and CSE−/− mice under normoxia (Inline graphic ∼ 146 mmHg) and hypoxia (Inline graphic ∼ 40 mmHg). Basal H2S generation under normoxia was significantly less in CSE−/− compared to CSE+/+ mice (CSE+/+ = 55 ± 3 vs. CSE−/− = 29 ± 1 nmol/h/mg protein; 53% less in mutant mice). Hypoxia increased H2S generation in CSE+/+ but not in mutant mice (CSE+/+ mice 2.5-fold increase vs. CSE−/− mice 0.3-fold increase; P < 0.001; Fig. 1D).

To assess whether the attenuated hypoxic response in mutant mice was due to reduced excitability of the carotid body, sensory response to CO2, another physiological stimulus, was determined. Carotid bodies from CSE+/+ and CSE−/− mice responded to CO2 with a comparable increase in sensory discharge (Fig. 1 E and F).

Impaired Ventilatory Response to Acute Hypoxia in CSE−/− Mice.

Ventilatory responses to 21% O2 (normoxia) and 12% O2 (hypoxia) were determined by whole-body plethysmography in awake, nonsedated CSE+/+ and CSE−/− mice. Baseline ventilation (VE) was significantly lower in CSE−/− compared to CSE+/+ mice (Fig. 2 A and B), which was due to significantly lower respiratory rates (RR) in mutant mice (Table S1). More importantly, the magnitude of hypoxic ventilatory response was significantly less in CSE−/− compared to CSE+/+ mice (Fig. 2 A and B), which was due to decreased stimulation of RR and tidal volume (VT) (Table S1). However, changes in O2 consumption (Inline graphic) and CO2 production (Inline graphic) under hypoxia were comparable between mutant and CSE+/+ mice (Table S1). In contrast, CSE+/+ and CSE−/− mice responded to hypercapnia (5% CO2) with comparable increases in VE (Fig. 2 A and C), RR, and VT (Table S2).

Fig. 2.

Fig. 2.

Ventilatory responses to hypoxia and hypercapnia in CSE+/+ and CSE−/− mice. Ventilation was measured in unsedated mice by whole body plethysmography under normoxia (21% O2), hypoxia (12% O2), and hypercapnia (5% CO2). Hypoxia and hypercapnia lasted for 5 min. Representative tracings of breathing are shown in A and average data of minute ventilation (VE) in response to 12% O2, i.e., hypoxia (B) and 5% CO2 i.e., hypercapnia (C). The data presented are mean ± SEM from eight CSE+/+ and CSE−/− mice each. **, P < 0.01; n.s. (not significant), P > 0.05.

CSE and Hypoxic Sensitivity of the Rat Carotid Body.

To assess whether CSE contributes to the carotid body hypoxic response in species other than mouse, CSE expression and the effect of its pharmacologic inhibition by PAG were determined in rat carotid bodies. CSE immunoreactivity was found in glomus cells of the rat carotid body (Fig. 3A Left). Rat carotid bodies being ∼3 times bigger than those found in mouse (∼80 μg in rat vs. 25 μg in mouse), we were able to determine the effects of graded hypoxia on H2S generation. Basal H2S generation in rat carotid body was 266 ± 61 nmol/h/mg protein which was significantly higher than in mouse carotid body (55 ± 3 nmol/h/mg protein; P < 0.01). Hypoxia increased H2S levels in a stimulus-dependent manner (Fig. 3A Right). The magnitude of hypoxia-evoked H2S production was comparable in rat and mouse (∼5-fold increase in rats vs. ∼3.5-fold in mice at Inline graphic ∼ 40 mmHg). In PAG-treated rats, basal H2S levels were reduced by 55% (vehicle = 266 ± 61 vs. PAG = 147 ± 50 nmol/h/mg protein; P < 0.05) and hypoxia-evoked H2S generation was nearly absent in the carotid bodies (Fig. 3A Right).

Fig. 3.

Fig. 3.

CSE localization in the rat carotid body and rat carotid body responses to hypoxia and hypercapnia in the absence and presence of DL-propargylglycine. (A) Cystathionine γ-lyase (CSE) expression in rat carotid body. Carotid body sections were stained with antibodies specific for CSE and tyrosine hydroxylase (TH), a marker of glomus cells (Left). Effects of graded hypoxia on H2S levels in vehicle-(PAG−) and DL-propargylglycine (PAG+)-treated carotid bodies. Data are mean ± SEM from five individual experiments (Right). (B) Examples of carotid body response to hypoxia (Hx) (Inline graphic = 38 mmHg; at black bar) in vehicle- and PAG-treated rats (Left). Average (mean ± SEM) data of sensory response to graded hypoxia (Right), PAG− n = 12 fibers from six rats; PAG+ n = 10 fibers from six rats. (C) Example of carotid body response to CO2 in the same rats as in B (Inline graphic ∼ 68 mmHg; at black bar (Left)] and average (mean ± SEM) data of CO2 response (Right). Data derived from n = 9 fibers (PAG−) and n = 10 (PAG+) fibers from six rats each. In B and C, integrated carotid body sensory activity (CB activity) is presented as impulses per second (imp/s). Superimposed action potentials from the single fiber are presented in Inset. **, P < 0.01.

In vehicle-treated rats, hypoxia augmented sensory activity in a stimulus-dependent manner, and this response was significantly attenuated in PAG-treated rats (Fig. 3B). The attenuated hypoxic response in PAG-treated rats could be secondary to accumulation of cysteine resulting from CSE inhibition. However, 100 μM L-cysteine had no significant effect on the hypoxic response (Fig. S2). In sharp contrast, carotid body response to CO2 was significantly augmented in PAG-treated rats (same rats tested for hypoxia; Fig. 3C).

H2S Stimulates Carotid Body Sensory Activity.

The effects of exogenous administration of H2S on the sensory activity of the carotid body were examined. NaHS, an H2S donor, augmented carotid body activity in rats (Fig. 4A), and in CSE+/+ and CSE−/− mice (Fig. S3). Like hypoxia, carotid body response to NaHS was stimulus-dependent, occurred within seconds after its application, and sensory activity promptly returned to baseline after termination of the stimulus (Fig. 4A Center and Right).

Fig. 4.

Fig. 4.

Effect of NaHS on rat carotid body sensory activity. (A) Example of rat carotid body response to increasing concentrations of NaHS, an H2S donor (at black bar; Left). Average (mean ± SEM) data of dose–response to NaHS (Center) and time course of sensory response to NaHS (100 μM) and hypoxia [Inline graphic = 42 mmHg (Right)]. Data in middle and right panels were obtained from n = 13 fibers from six rats. (B) Effect of Ca2+ free medium on rat carotid body responses to 100 μM NaHS and hypoxia (Hx) (Inline graphic = 42 mmHg; at black bar). CaCl2 was replaced by 3 mM MgCl2 and 5 mM EGTA was added to the medium. (Left) Example and Right average (mean ± SEM) data from five rats (n = 8 fibers). In A and B, Integrated carotid body sensory activity (CB activity) is presented as impulses per second (imp/s). Superimposed action potentials from the single fiber are presented in the inset. **, P < 0.01; n.s. (not significant), P > 0.05.

Ca2+ influx into glomus cells is critical for carotid body response to hypoxia (1). To assess whether Ca2+ is also important for sensory excitation by H2S, carotid body responses to NaHS were examined in the presence of Ca2+-free and EGTA-containing medium. Carotid body responses to NaHS as well as to hypoxia were abolished in the presence of Ca2+-free and EGTA containing medium (Fig. 4B). On the other hand, glibenclamide, a potent inhibitor of ATP-sensitive potassium (KATP) channels which mediate H2S responses in vasculature (16), failed to prevent carotid body response either to NaHS or hypoxia (Fig. S4).

HO-2 Modulates H2S Generation by CSE.

Glomus cells express HO-2, an enzyme that generates CO (3). To assess whether the HO-2-CO system affects CSE activity, H2S generation was determined in CSE+/+ and CSE−/− carotid bodies in the presence and absence of Cr (III) mesoporphyrin IX chloride [Cr(III)MP], an inhibitor of HO-2 (17). As little as 1 μM Cr(III)MP increased H2S generation 6-fold in CSE+/+ mice carotid bodies under normoxia, and this response was abolished in mutant mice (Fig. 5A). Systemic administration of Cr(III)MP significantly increased baseline carotid body activity and potentiated the hypoxic response in CSE+/+ but not in mutant mice (Fig. 5 BD). In rat carotid bodies, Cr(III)MP also enhanced H2S generation under normoxia, and a CO donor (tricarbonyldichlororuthenium, [Ru(CO)3Cl2]2) inhibited H2S generation by hypoxia nearly to the levels seen under normoxia (Fig. S5).

Fig. 5.

Fig. 5.

Effect of HO-2 inhibition on carotid body H2S generation and sensory activity in CSE+/+ and CSE−/− mice. (A) Effect of Cr(III)MP (1 μM), an inhibitor of heme oxygenase 2 on H2S generation in carotid bodies under normoxia (Inline graphic ∼ 146 mmHg) from CSE+/+ and CSE−/− mice. Data presented are mean ± SEM from three experiments. Examples of baseline and hypoxic response (Hx) (Inline graphic ∼ 40 mmHg; at black bar) of carotid bodies from vehicle- and Cr(III)MP-treated CSE+/+ and CSE−/− mice (B) and average data (mean ± SEM) from six mice in each group (n = 8–12 fibers) in C and D. In B, Integrated carotid body sensory activity (CB activity) is presented as impulses per second (imp/s). Superimposed action potentials from the single fiber are presented in Inset. *** and *, P < 0.001 and P < 0.05, respectively; n.s. (not significant), P > 0.05.

CSE Disruption Impairs Hypoxic Sensing by Neonatal Adrenal Medullary Chromaffin Cells (AMC).

Hypoxia stimulates catecholamine secretion from neonatal AMC (18, 19). To assess the role of CSE in AMC, studies were performed on neonatal mice and rats at age P10. CSE immunoreactivity was seen in mice and rat AMC (Fig. 6 A and B, Top) but was absent in AMC from CSE−/− mice. Hypoxia-evoked catecholamine secretion was monitored by amperometry from single AMC harvested from CSE−/− mice and PAG-treated rat pups. Control studies were performed on age matched CSE+/+ mice and vehicle-treated rat pups, respectively. In CSE+/+ mice, hypoxia elicited robust catecholamine secretion, which was greatly reduced or absent in AMC from mutant mice (Fig. 6A Middle). A similar reduction in hypoxia-evoked catecholamine secretion was also seen in PAG-treated rat pups (Fig. 6B Middle). In contrast, K+-evoked (40 mM K+; nonselective depolarizing stimulus) catecholamine secretion was unaffected in CSE−/− mice and PAG-treated rat pups (Fig. 6 A and B, Middle and Bottom). H2S generation increased under hypoxia in adrenal glands from CSE+/+ mice and vehicle-treated rat pups, and this response was greatly reduced or absent in CSE−/− mice and PAG-treated rat pups, respectively.

Fig. 6.

Fig. 6.

CSE localization in the mouse and rat neonatal adrenal medullary chromaffin cells (AMC) and the effect of CSE disruption on mouse and rat AMC hypoxia sensing. (Top) Cystathionine γ-lyase (CSE) expression in AMC from mice (A) and rat pups (B). (Middle) Examples of catecholamine secretion from AMC from neonatal mice (A) and rats (B) in response to hypoxia (Hx) (Inline graphic = 36 mmHg) or high K+ (40 mM). Black bar represents the duration of hypoxia or K+ application. (Bottom) Average data (mean ± SEM) of total catecholamine (CA) secreted during Hx or K+ (CA molecules 107 i.e., number of secretory events multiplied by catecholamine molecules secreted per event). n = 9 cells each from CSE+/+ and CSE−/− and n = 10–12 cells from rat pups. **, P < 0.01; n.s. (not significant), P > 0.05 compared with CSE+/+ mice or vehicle-treated rat cells.

Discussion

The present study establishes a physiological role for H2S generated by CSE in mediating hypoxic sensing by the carotid body. CSE is expressed in rat and mouse glomus cells, the main site of O2 sensing in the carotid body. Moreover, chemoreceptor responses to acute hypoxia were markedly impaired in CSE knockout mice and following pharmacologic inhibition of CSE. The loss of hypoxic sensitivity does not likely reflect reduced carotid body excitability, because sensory response to CO2 was intact in mutant mice and CSE inhibitor-treated rats. Carotid bodies are the primary mediators of ventilatory stimulation by acute hypoxia. CSE−/− mice exhibited selective loss of ventilatory response to hypoxia but not to CO2, suggesting that CSE disruption profoundly impacts systemic responses to acute hypoxia by affecting the carotid body.

How might CSE contribute to hypoxic sensing? CSE catalyzes the formation of cysteine from cystathionine, and also generates H2S from cyst(e)ine (8, 9). The following observations indicate that H2S generated by CSE mediates carotid body hypoxic sensing. First, hypoxia increased H2S generation in the carotid body in a stimulus-dependent manner, an effect that was lost in CSE−/− mice as well as in rats treated with a CSE inhibitor. Second, loss of hypoxia-evoked H2S generation paralleled impaired hypoxic sensing by the carotid body. Third, an H2S donor, but not L-cysteine, stimulated the carotid body with a time-course and magnitude comparable to that evoked by low O2. An H2S donor stimulated carotid body activity in CSE knockout mice, indicating that the loss of hypoxic sensitivity is due to absence of H2S generation rather than impaired H2S signaling. These findings demonstrate that during hypoxia, CSE is the main source of H2S generation in the carotid body, and suggest that CSE contributes to hypoxic sensing by catalyzing H2S generation.

Interestingly, like the sensory activity, basal H2S generation in the carotid bodies was low under normoxia. Because HO-2 is an O2 requiring enzyme, it has been proposed that the low normoxemic sensory activity reflects tonic inhibition by HO-2 generated CO in the carotid body (3). We reasoned that a similar inhibitory influence by the HO-2-CO system might account for low normoxic levels of H2S. Indeed, an HO-2 inhibitor not only markedly elevated basal H2S levels but also augmented baseline sensory activity and potentiated the hypoxic response in CSE+/+ mice. These effects were absent in CSE−/− mice, implying that HO-2 tonically inhibits CSE. CO generated by HO-2 presumably mediates the inhibition of H2S generation from CSE, as a CO donor inhibited H2S generation during hypoxia. How might CO inhibit H2S generation by CSE? CO interacts with CBS, another enzyme that generates H2S, by binding to its heme moiety (20). Given that CSE is not a heme containing enzyme; it is likely that CO inhibits CSE activity by interacting with histidine residues, as it does with other proteins (21). Thus, CO appears to physiologically inhibit the CSE-H2S system with hypoxia reducing HO-2 activity to reverse the inhibition and augment H2S formation.

Although KATP channels are targets of H2S (8, 9, 16), glibenclamide, a potent inhibitor of KATP channels, was ineffective in preventing carotid body stimulation by NaHS or hypoxia. On the other hand, Ca2+ influx appears critical for carotid body stimulation by H2S as well as hypoxia. Previous studies showed that Ca2+ influx via high voltage-gated Ca2+ channels, especially the L-type, is necessary for carotid body stimulation by hypoxia (1). L-type Ca2+ channels in glomus cells are redox sensitive, activated by hypoxia, and inhibited under normoxia, as well as by gaseous messengers such as NO (22, 23). A recent study demonstrated that H2S signaling involves covalent modification of redox sensitive cysteine residues in proteins through S-sulfhydration (10). It is conceivable that H2S generated by hypoxia activates L-type Ca2+ channels in glomus cells via S-sulfhydration. In addition, H2S might affect Ca2+-activated K+ currents in glomus cells as suggested recently (15, 24).

Carotid bodies are the main organs for sensing acute hypoxia in adults but in neonates they are relatively insensitive to low O2 (25, 26). On the other hand, AMC are extremely sensitive to hypoxia in neonates, and low O2 stimulates catecholamine secretion (18, 19), which is critical for maintaining homeostasis in neonates under hypoxic stress. Like glomus cells, neonatal AMC expressed CSE, and hypoxia-evoked catecholamine secretion was severely impaired in CSE−/− mice and in rats treated with a CSE inhibitor. Because hypoxia also increased H2S generation in adrenal glands, CSE-H2S system appears to mediate acute hypoxic sensing by neonatal AMC. However, hypoxic sensitivity of AMC decline with age (19). Whether age-related loss of hypoxic sensing by AMC is associated with developmental decline in CSE expression remains to be investigated. It is interesting to note that H2S has been demonstrated to mediate O2 sensing by the trout gill chemoreceptors, indicating that it is an ancient well conserved system across phyla (27).

Reflexes arising from the carotid body have been implicated in pathological situations including sleep-disordered breathing with recurrent apnea i.e., periodic cessations of breathing (28). Patients with recurrent apnea experience periodic hypoxemia or intermittent hypoxia and are prone to autonomic morbidities including hypertension (29). In rodent models chronic intermittent hypoxia enhances carotid body responses to hypoxia, and the ensuing chemo-reflex mediates increases in sympathetic nerve activity resulting in elevated blood pressure (28). By reducing carotid body activity, CSE inhibitors or other interventions that reduce H2S formation may afford therapeutic benefits for sleep apnea and related conditions.

Methods

Preparation of Animals.

Experiments were approved by the Institutional Animal Care and Use Committee of the University of Chicago and were performed on age-matched, male CSE+/+ and CSE−/− mice (2–3 mo old) and male Sprague–Dawley rats (2–3 mo old) except where otherwise noted. In the experiments requiring sedation, animals were anesthetized with i.p. injections of urethane (1.2 g/kg; Sigma). Supplemental doses, 10% of the initial dose, of urethane were given when corneal reflexes and responses to toe pinch persisted. Animals were allowed to breathe spontaneously. Core body temperature was monitored by a rectal thermistor probe and maintained at 37 ± 1 °C by a heating pad. At the end of the experiment, animals were euthanized by intracardiac injection (0.1 mL) of euthanasia solution (Beuthanasia-D Special; Schering-Plough).

Immunohistochemistry.

Carotid bodies and adrenal glands were harvested from anesthetized rats or mice (urethane 1.2 g/kg, i.p.) perfused with heparinized saline followed by 4% paraformaldehyde. The protocols for fixation of carotid bodies and adrenal glands were essentially the same as described in refs. 3 and 11. For assessing CSE immunoreactivity, sections (8 μm thick) were incubated at room temperature for 2 h with polyclonal rabbit anti-CSE antibody (1:400), this antibody was raised using bacterially purified full-length His-tagged CSE as antigen, and monoclonal mouse anti-tyrosine hydroxylase (1:2,000; Sigma), an established marker of glomus and chromaffin cells (11, 12), followed by Texas red-conjugated goat anti-rabbit IgG and FITC-conjugated goat anti-mouse IgG (1:250; Molecular Probes) in PBS with 1% normal goat serum and 0.2% Triton X-100. After washing with PBS, sections were mounted in DAPI-containing media and visualized using a fluorescent microscope (Eclipse E600; Nikon).

Carotid Body Sensory Activity.

Sensory activity from carotid bodies ex vivo was recorded as described in refs. 30 and 31. Briefly, carotid bodies along with the sinus nerves were harvested from anesthetized mice or rats, placed in a recording chamber (volume, 250 μL) and superfused with warm physiological saline (35 °C) at a rate of 2.5 mL/min having the composition 140 mM NaCl/5.4 mM KCl/2.5 mM CaCl2/0.5 mM MgCl2/5.5 mM Hepes/11 mM D-glucose/5 mM sucrose, and the solution was bubbled with 100% O2 and the pH was adjusted to 7.35. Carotid bodies were challenged with graded hypoxia by switching the perfusate equilibrated with gas mixtures containing varying levels of O2. For assessing the carotid body response to CO2, bicarbonate-buffered medium equilibrated with either 90% O2 + 5% CO2 (baseline) or 90% O2 + 10% CO2 balance N2 was used. Clearly identifiable action potentials (two to three active units) were recorded from one of the nerve bundles with a suction electrode and stored in a computer via a data acquisition system (PowerLab/8P; AD Instruments). ”Single“ units were selected based on the height and duration of the individual action potentials using a spike discrimination program (Spike Histogram Program, Power Laboratory; AD Instruments). In each carotid body, at least two chemoreceptor units were analyzed. The Inline graphic and Inline graphic of the superfusion medium were determined by a blood gas analyzer (ABL 5; Radiometer).

Measurements of Respiratory Variables.

Ventilation was monitored by whole-body plethysmograph, and O2 consumption and CO2 production were determined by the open-circuit method in unsedated mice as described in ref. 31. Ventilation was recorded while the mice breathed 21% or 12% O2-balance N2. Each gas challenge was given for 5 min. O2 consumption and CO2 production were measured at the end of each 5-min challenge. For determining ventilatory response to CO2, baseline ventilation was determined while mice breathed 21% O2 followed by hypercapnic challenge with 5% CO2–21% O2–balance N2. Sighs, sniffs, and movement-induced changes in breathing were monitored and excluded in the analysis. All recordings were made at an ambient temperature of 25 ± 1 °C.

Measurements of H2S Levels.

H2S levels in the carotid body were assayed as described in refs. 32 and 33 with few modifications. Briefly, carotid bodies were pooled (four carotid bodies per experiment in rats; six carotid bodies per experiment in mice) and tissue homogenates were prepared in 100 mM potassium phosphate buffer (pH 7.4). The enzyme reaction was carried out in sealed tubes flushed with either N2 or different levels of O2-N2 gas mixtures. The Inline graphic of the reaction medium was determined by blood gas analyzer (ABL5). The assay mixture in a total volume of 500 μL contained (in final concentration) 800 μM L-cysteine, 80 μM pyridoxal 5′-phosphate, 100 mM potassium phosphate buffer (pH 7.4), and tissue homogenate (2 μg of protein). The reaction mixture was incubated at 37 °C for 1 h and at the end of the reaction, alkaline zinc acetate (1% wt/vol; 250 μL) and trichloroacetic acid (10% vol/vol) was added sequentially to trap H2S generated and to stop the reaction, respectively. The zinc sulfide formed was reacted sequentially with acidic N,N-dimethyl-p-phenylenediamine sulfate (20 μM) and ferric chloride (30 μM) and the absorbance was measured at 670 nm using a microplate reader. A standard curve relating the concentration of Na2S and absorbance was used to calculate H2S concentration and expressed as nanomoles of H2S formed per hour per milligram protein.

Measurements of Catecholamine Secretion from Chromaffin Cells.

The protocols for preparation of chromaffin cells and measurements of catecholamine secretion by amperometry are essentially the same as described in ref. 18. Briefly, adrenal glands were harvested from anesthetized mice and rats aged P10. Chromaffin cells were enzymatically dissociated, plated on collagen (type VII; Sigma) coated coverslips, and maintained at 37°C in a 5% CO2 incubator for 12–24 h. The growth medium consisted of F-12 K medium (Invitrogen) supplemented with 10% horse serum, 5% FBS, and 1% penicillin/streptomycin/glutamine mixture (Invitrogen). Catecholamine secretion from chromaffin cells was monitored by amperometry using carbon fiber electrodes as described in ref. 18. Amperometric recordings were made from adherent cells that were superfused (flow rate ∼1.0 mL/min, chamber volume ∼80 μL) with a medium having the composition 1.26 mM CaCl2/0.49 mM MgCl2·6H2O/0.4 mM MgSO4·7H2O/5.33 mM KCl/0.441 mM KH2PO4/137.93 mM NaCl/0.34 mM Na2HPO4·7H2O/5.56 mM dextrose/20 mM Hepes (pH 7.35) and 300 mOsmol. Normoxic solutions were equilibrated with room air (Inline graphic ∼ 146 mmHg), and hypoxic solutions were equilibrated with appropriate gas mixtures that yielded medium Inline graphic of ∼30–40 mmHg as measured by blood gas analyzer.

Drugs.

Rats and mice were treated with DL-propargylglycine (Sigma; 200 mg/kg) or Cr (III) Mesoporphyrin IX chloride [1 mg/kg (CrM459; Frontier Scientific)] administered intraperitoneally 2–3 h before acute experiments on anesthetized animals. NaHS (Sigma) was given acutely to the ex vivo carotid body preparation. In the in vitro assays for H2S measurements, known concentrations of HO-2 inhibitor and CO donor (tricarbonyldichlororuthenium [II] dimer, [Ru(CO)3Cl2]2; Sigma) were added to cell lysates. All solutions with drugs were prepared fresh during the experiments.

Data Analysis.

In unsedated mice RR (breaths/min), VT (μL), minute ventilation [VE (mL/min) = RR × VT], O2 consumption (Inline graphic, mL/min), and CO2 production (Inline graphic, mL/min) were analyzed. Respiratory variables (RR and VT) were averaged for at least 20 consecutive breaths over 5 min of inspired O2 and CO2 challenge. VT, VE, Inline graphic, and Inline graphic were normalized to body weight. Carotid body sensory activity (discharge from “single” units) was averaged during 3 min of baseline and during the 3 min of gas challenge and expressed as impulses per second unless otherwise stated. The number of secretory events and the amount of catecholamine secreted per secretory event were analyzed in each experiment and the data were expressed as total catecholamines secreted. Average data are presented as mean ± SEM. Statistical significance was assessed by either ANOVA or two-way ANOVA with repeated measures followed by Tukey's test. P < 0.05 was considered significant.

Supplementary Material

Supporting Information

Acknowledgments

We thank Dr. A. P. Fox for help with the experiments involving amperometry, Ms. Ning Wang for help with the experiments involving immunocytochemical assays, and Dr. Rui Wang for valued contributions. This work was supported by National Institutes of Health Grants HL-76537, HL-90554, and HL-86493 (to N.R.P.) and US Public Health Service Grant DA 000226 and Research Scientist Award DA 00074 (to S.H.S.). M.M.G. is supported by the National Institutes of Health Medical Scientist Training Program Award T32 GM007309.

Footnotes

The authors declare no conflict of interest.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1005866107/-/DCSupplemental.

References

  • 1.Prabhakar NR. Oxygen sensing by the carotid body chemoreceptors. J Appl Physiol. 2000;88:2287–2295. doi: 10.1152/jappl.2000.88.6.2287. [DOI] [PubMed] [Google Scholar]
  • 2.Prabhakar NR, Kumar GK, Chang CH, Agani FH, Haxhiu MA. Nitric oxide in the sensory function of the carotid body. Brain Res. 1993;625:16–22. doi: 10.1016/0006-8993(93)90132-7. [DOI] [PubMed] [Google Scholar]
  • 3.Prabhakar NR, Dinerman JL, Agani FH, Snyder SH. Carbon monoxide: A role in carotid body chemoreception. Proc Natl Acad Sci USA. 1995;92:1994–1997. doi: 10.1073/pnas.92.6.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Williams SE, et al. Hemoxygenase-2 is an oxygen sensor for a calcium-sensitive potassium channel. Science. 2004;306:2093–2097. doi: 10.1126/science.1105010. [DOI] [PubMed] [Google Scholar]
  • 5.Prabhakar NR. NO and CO as second messengers in oxygen sensing in the carotid body. Respir Physiol. 1999;115:161–168. doi: 10.1016/s0034-5687(99)00019-5. [DOI] [PubMed] [Google Scholar]
  • 6.Kimura H, Nagai Y, Umemura K, Kimura Y. Physiological roles of hydrogen sulfide: Synaptic modulation, neuroprotection, and smooth muscle relaxation. Antioxid Redox Signal. 2005;7:795–803. doi: 10.1089/ars.2005.7.795. [DOI] [PubMed] [Google Scholar]
  • 7.Yang G, et al. H2S as a physiologic vasorelaxant: Hypertension in mice with deletion of cystathionine γ-lyase. Science. 2008;322:587–590. doi: 10.1126/science.1162667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Mustafa AK, Gadalla MM, Snyder SH. Signaling by gasotransmitters. Sci Signal. 2009 doi: 10.1126/scisignal.268re2. 10.1126/scisignal.268re2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gadalla MM, Snyder SH. Hydrogen sulfide as a gasotransmitter. J Neurochem. 2010;113:14–26. doi: 10.1111/j.1471-4159.2010.06580.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Mustafa AK, et al. H2S signals through protein S-sulfhydration. Sci Signal. 2009 doi: 10.1126/scisignal.2000464. 10.1126/scisignal.2000464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kline DD, et al. Defective carotid body function and impaired ventilatory responses to chronic hypoxia in mice partially deficient for hypoxia-inducible factor 1α. Proc Natl Acad Sci USA. 2002;99:821–826. doi: 10.1073/pnas.022634199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nurse CA, Fearon IM. Carotid body chemoreceptors in dissociated cell culture. Microsc Res Tech. 2002;59:249–255. doi: 10.1002/jemt.10199. [DOI] [PubMed] [Google Scholar]
  • 13.Abeles RH, Walsh CT. Acetylenic enzyme inactivators. Inactivation of γ-cystathionase, in vitro and in vivo, by propargylglycine. J Am Chem Soc. 1973;95:6124–6125. doi: 10.1021/ja00799a053. [DOI] [PubMed] [Google Scholar]
  • 14.Washtien W, Abeles RH. Mechanism of inactivation of γ-cystathionase by the acetylenic substrate analogue propargylglycine. Biochemistry. 1977;16:2485–2491. doi: 10.1021/bi00630a026. [DOI] [PubMed] [Google Scholar]
  • 15.Li Q, et al. A crucial role for hydrogen sulfide in oxygen sensing via modulating large conductance calcium-activated potassium channels. Antioxid Redox Signal. 2010;12:1179–1189. doi: 10.1089/ars.2009.2926. [DOI] [PubMed] [Google Scholar]
  • 16.Zhao W, Zhang J, Lu Y, Wang R. The vasorelaxant effect of H2S as a novel endogenous gaseous KATP channel opener. EMBO J. 2001;20:6008–6016. doi: 10.1093/emboj/20.21.6008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Vreman HJ, Ekstrand BC, Stevenson DK. Selection of metalloporphyrin heme oxygenase inhibitors based on potency and photoreactivity. Pediatr Res. 1993;33:195–200. doi: 10.1203/00006450-199302000-00021. [DOI] [PubMed] [Google Scholar]
  • 18.Souvannakitti D, Kumar GK, Fox A, Prabhakar NR. Neonatal intermittent hypoxia leads to long-lasting facilitation of acute hypoxia-evoked catecholamine secretion from rat chromaffin cells. J Neurophysiol. 2009;101:2837–2846. doi: 10.1152/jn.00036.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Thompson RJ, Jackson A, Nurse CA. Developmental loss of hypoxic chemosensitivity in rat adrenomedullary chromaffin cells. J Physiol. 1997;498:503–510. doi: 10.1113/jphysiol.1997.sp021876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Taoka S, Banerjee R. Characterization of NO binding to human cystathionine β-synthase: Possible implications of the effects of CO and NO binding to the human enzyme. J Inorg Biochem. 2001;87:245–251. doi: 10.1016/s0162-0134(01)00335-x. [DOI] [PubMed] [Google Scholar]
  • 21.Wang R, Wu L. The chemical modification of KCa channels by carbon monoxide in vascular smooth muscle cells. J Biol Chem. 1997;272:8222–8226. doi: 10.1074/jbc.272.13.8222. [DOI] [PubMed] [Google Scholar]
  • 22.Summers BA, Overholt JL, Prabhakar NR. Augmentation of L-type calcium current by hypoxia in rabbit carotid body glomus cells: Evidence for a PKC-sensitive pathway. J Neurophysiol. 2000;84:1636–1644. doi: 10.1152/jn.2000.84.3.1636. [DOI] [PubMed] [Google Scholar]
  • 23.Summers BA, Overholt JL, Prabhakar NR. Nitric oxide inhibits L-type Ca2+ current in glomus cells of the rabbit carotid body via a cGMP-independent mechanism. J Neurophysiol. 1999;81:1449–1457. doi: 10.1152/jn.1999.81.4.1449. [DOI] [PubMed] [Google Scholar]
  • 24.Telezhkin V, et al. Hydrogen sulfide inhibits human BK(Ca) channels. Adv Exp Med Biol. 2009;648:65–72. doi: 10.1007/978-90-481-2259-2_7. [DOI] [PubMed] [Google Scholar]
  • 25.Donnelly DF. Developmental aspects of oxygen sensing by the carotid body. J Appl Physiol. 2000;88:2296–2301. doi: 10.1152/jappl.2000.88.6.2296. [DOI] [PubMed] [Google Scholar]
  • 26.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. 2008;104:1287–1294. doi: 10.1152/japplphysiol.00644.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Olson KR, et al. Hydrogen sulfide as an oxygen sensor in trout gill chemoreceptors. Am J Physiol. 2008;295:R669–R680. doi: 10.1152/ajpregu.00807.2007. [DOI] [PubMed] [Google Scholar]
  • 28.Prabhakar NR, Dick TE, Nanduri J, Kumar GK. Systemic, cellular and molecular analysis of chemoreflex-mediated sympathoexcitation by chronic intermittent hypoxia. Exp Physiol. 2007;92:39–44. doi: 10.1113/expphysiol.2006.036434. [DOI] [PubMed] [Google Scholar]
  • 29.Nieto FJ, et al. Association of sleep-disordered breathing, sleep apnea, and hypertension in a large community-based study. Sleep Heart Health Study. JAMA. 2000;283:1829–1836. doi: 10.1001/jama.283.14.1829. [DOI] [PubMed] [Google Scholar]
  • 30.Peng YJ, Overholt JL, Kline D, Kumar GK, Prabhakar NR. Induction of sensory long-term facilitation in the carotid body by intermittent hypoxia: Implications for recurrent apneas. Proc Natl Acad Sci USA. 2003;100:10073–10078. doi: 10.1073/pnas.1734109100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Peng YJ, et al. Heterozygous HIF-1α deficiency impairs carotid body-mediated systemic responses and reactive oxygen species generation in mice exposed to intermittent hypoxia. J Physiol. 2006;577:705–716. doi: 10.1113/jphysiol.2006.114033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Li L, et al. Hydrogen sulfide is a novel mediator of lipopolysaccharide-induced inflammation in the mouse. FASEB J. 2005;19:1196–1198. doi: 10.1096/fj.04-3583fje. [DOI] [PubMed] [Google Scholar]
  • 33.Qu K, Chen CP, Halliwell B, Moore PK, Wong PT. Hydrogen sulfide is a mediator of cerebral ischemic damage. Stroke. 2006;37:889–893. doi: 10.1161/01.STR.0000204184.34946.41. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES