1
|
Leonard EM, Porteus CS, Brink D, Milsom WK. Fish gill chemosensing: knowledge gaps and inconsistencies. J Comp Physiol B 2024; 194:1-33. [PMID: 38758303 DOI: 10.1007/s00360-024-01553-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/04/2024] [Indexed: 05/18/2024]
Abstract
In this review, we explore the inconsistencies in the data and gaps in our knowledge that exist in what is currently known regarding gill chemosensors which drive the cardiorespiratory reflexes in fish. Although putative serotonergic neuroepithelial cells (NEC) dominate the literature, it is clear that other neurotransmitters are involved (adrenaline, noradrenaline, acetylcholine, purines, and dopamine). And although we assume that these agents act on neurons synapsing with the NECs or in the afferent or efferent limbs of the paths between chemosensors and central integration sites, this process remains elusive and may explain current discrepancies or species differences in the literature. To date it has been impossible to link the distribution of NECs to species sensitivity to different stimuli or fish lifestyles and while the gills have been shown to be the primary sensing site for respiratory gases, the location (gills, oro-branchial cavity or elsewhere) and orientation (external/water or internal/blood sensing) of the NECs are highly variable between species of water and air breathing fish. Much of what has been described so far comes from studies of hypoxic responses in fish, however, changes in CO2, ammonia and lactate have all been shown to elicit cardio-respiratory responses and all have been suggested to arise from stimulation of gill NECs. Our view of the role of NECs is broadening as we begin to understand the polymodal nature of these cells. We begin by presenting the fundamental picture of gill chemosensing that has developed, followed by some key unanswered questions about gill chemosensing in general.
Collapse
Affiliation(s)
- Erin M Leonard
- Department of Biology, Wilfrid Laurier University, Waterloo, ON, Canada
| | - Cosima S Porteus
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, ON, Canada.
| | - Deidre Brink
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - William K Milsom
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Khalilpour J, Soltani Zangbar H, Alipour MR, Shahabi P. The hypoxic respiratory response of the pre-Bötzinger complex. Heliyon 2024; 10:e34491. [PMID: 39114066 PMCID: PMC11305331 DOI: 10.1016/j.heliyon.2024.e34491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
Since the discovery of the pre-Bötzinger Complex (preBötC) as a crucial region for generating the main respiratory rhythm, our understanding of its cellular and molecular aspects has rapidly increased within the last few decades. It is now apparent that preBötC is a highly flexible neuronal network that reconfigures state-dependently to produce the most appropriate respiratory output in response to various metabolic challenges, such as hypoxia. However, the responses of the preBötC to hypoxic conditions can be varied based on the intensity, pattern, and duration of the hypoxic challenge. This review discusses the preBötC response to hypoxic challenges at the cellular and network level. Particularly, the involvement of preBötC in the classical biphasic response of the respiratory network to acute hypoxia is illuminated. Furthermore, the article discusses the functional and structural changes of preBötC neurons following intermittent and sustained hypoxic challenges. Accumulating evidence shows that the preBötC neural circuits undergo substantial changes following hypoxia and contribute to several types of the respiratory system's hypoxic ventilatory responses.
Collapse
Affiliation(s)
- Jamal Khalilpour
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Soltani Zangbar
- Department of Neuroscience, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Getsy PM, Coffee GA, May WJ, Baby SM, Bates JN, Lewis SJ. The Reducing Agent Dithiothreitol Modulates the Ventilatory Responses That Occur in Freely Moving Rats during and following a Hypoxic-Hypercapnic Challenge. Antioxidants (Basel) 2024; 13:498. [PMID: 38671945 PMCID: PMC11047747 DOI: 10.3390/antiox13040498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/01/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
The present study examined the hypothesis that changes in the oxidation-reduction state of thiol residues in functional proteins play a major role in the expression of the ventilatory responses in conscious rats that occur during a hypoxic-hypercapnic (HH) gas challenge and upon return to room air. A HH gas challenge in vehicle-treated rats elicited robust and sustained increases in minute volume (via increases in frequency of breathing and tidal volume), peak inspiratory and expiratory flows, and inspiratory and expiratory drives while minimally affecting the non-eupneic breathing index (NEBI). The HH-induced increases in these parameters, except for frequency of breathing, were substantially diminished in rats pre-treated with the potent and lipophilic disulfide-reducing agent, L,D-dithiothreitol (100 µmol/kg, IV). The ventilatory responses that occurred upon return to room air were also substantially different in dithiothreitol-treated rats. In contrast, pre-treatment with a substantially higher dose (500 µmol/kg, IV) of the lipophilic congener of the monosulfide, N-acetyl-L-cysteine methyl ester (L-NACme), only minimally affected the expression of the above-mentioned ventilatory responses that occurred during the HH gas challenge or upon return to room air. The effectiveness of dithiothreitol suggests that the oxidation of thiol residues occurs during exposure to a HH gas challenge and that this process plays an essential role in allowing for the expression of the post-HH excitatory phase in breathing. However, this interpretation is contradicted by the lack of effects of L-NACme. This apparent conundrum may be explained by the disulfide structure affording unique functional properties to dithiothreitol in comparison to monosulfides. More specifically, the disulfide structure may give dithiothreitol the ability to alter the conformational state of functional proteins while transferring electrons. It is also possible that dithiothreitol is simply a more efficient reducing agent following systemic injection, although one interpretation of the data is that the effects of dithiothreitol are not due to its reducing ability.
Collapse
Affiliation(s)
- Paulina M. Getsy
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.G.); (G.A.C.)
| | - Gregory A. Coffee
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.G.); (G.A.C.)
| | - Walter J. May
- Department of Pediatrics, University of Virginia, Charlottesville, VA 22903, USA;
| | - Santhosh M. Baby
- Galleon Pharmaceuticals, Inc., 213 Witmer Road, Horsham, PA 19044, USA;
| | - James N. Bates
- Department of Anesthesiology, University of Iowa Hospitals and Clinics, Iowa, IA 52242, USA;
| | - Stephen J. Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, USA; (P.M.G.); (G.A.C.)
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
- Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
4
|
Steppan J, Nandakumar K, Wang H, Jang R, Smith L, Kang S, Savage W, Bauer M, Choi R, Brady T, Wodu BP, Scafidi S, Scafidi J, Santhanam L. Neonatal exposure to hypoxia induces early arterial stiffening via activation of lysyl oxidases. Physiol Rep 2023; 11:e15656. [PMID: 37038896 PMCID: PMC10086679 DOI: 10.14814/phy2.15656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 04/12/2023] Open
Abstract
Hypoxia in the neonatal period is associated with early manifestations of adverse cardiovascular health in adulthood including higher risk of hypertension and atherosclerosis. We hypothesize that this occurs due to activation of lysyl oxidases (LOXs) and the remodeling of the large conduit vessels, leading to early arterial stiffening. Newborn C57Bl/6 mice were exposed to hypoxia (FiO2 = 11.5%) from postnatal day 1 (P1) to postnatal day 11 (P11), followed by resumption of normoxia. Controls were maintained in normoxia. Using in vivo (pulse wave velocity; PWV) and ex vivo (tensile testing) arterial stiffness indexes, we determined that mice exposed to neonatal hypoxia had significantly higher arterial stiffness compared with normoxia controls by young adulthood (P60), and it increased further by P120. Echocardiography performed at P60 showed that mice exposed to hypoxia displayed a compensated dilated cardiomyopathy. Western blotting revelated that neonatal hypoxia accelerated age-related increase in LOXL2 protein expression in the aorta and elevated LOXL2 expression in the PA at P11 with a delayed decay toward normoxic controls. In the heart and lung, gene and protein expression of LOX/LOXL2 were upregulated at P11, with a delayed decay when compared to normoxic controls. Neonatal hypoxia results in a significant increase in arterial stiffness in early adulthood due to aberrant LOX/LOXL2 expression. This suggests an acceleration in the mechanical decline of the cardiovascular system, that contributes to increased risk of hypertension in young adults exposed to neonatal hypoxia that may increase susceptibility to further insults.
Collapse
Affiliation(s)
- Jochen Steppan
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Kavitha Nandakumar
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Huilei Wang
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Rosie Jang
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Logan Smith
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Sara Kang
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - William Savage
- Department of Chemical and Biomolecular EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Maria Bauer
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Rira Choi
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Travis Brady
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Bulouere Princess Wodu
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Susanna Scafidi
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| | - Joseph Scafidi
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of NeurologyJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of PediatricsJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Michael V. Johnston Center for Developmental NeuroscienceKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care MedicineJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins University School of Medicine, Kennedy Krieger InstituteBaltimoreMarylandUSA
| |
Collapse
|
5
|
Gray OA, Yoo J, Sobreira DR, Jousma J, Witonsky D, Sakabe NJ, Peng YJ, Prabhakar NR, Fang Y, Nobréga MA, Di Rienzo A. A pleiotropic hypoxia-sensitive EPAS1 enhancer is disrupted by adaptive alleles in Tibetans. SCIENCE ADVANCES 2022; 8:eade1942. [PMID: 36417539 PMCID: PMC9683707 DOI: 10.1126/sciadv.ade1942] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 10/25/2022] [Indexed: 06/16/2023]
Abstract
In Tibetans, noncoding alleles in EPAS1-whose protein product hypoxia-inducible factor 2α (HIF-2α) drives the response to hypoxia-carry strong signatures of positive selection; however, their functional mechanism has not been systematically examined. Here, we report that high-altitude alleles disrupt the activity of four EPAS1 enhancers in one or more cell types. We further characterize one enhancer (ENH5) whose activity is both allele specific and hypoxia dependent. Deletion of ENH5 results in down-regulation of EPAS1 and HIF-2α targets in acute hypoxia and in a blunting of the transcriptional response to sustained hypoxia. Deletion of ENH5 in mice results in dysregulation of gene expression across multiple tissues. We propose that pleiotropic adaptive effects of the Tibetan alleles in EPAS1 underlie the strong selective signal at this gene.
Collapse
Affiliation(s)
- Olivia A. Gray
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Jennifer Yoo
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Débora R. Sobreira
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Jordan Jousma
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - David Witonsky
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Noboru J. Sakabe
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Ying-Jie Peng
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL 60637, USA
| | - Nanduri R. Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL 60637, USA
| | - Yun Fang
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Marcelo A. Nobréga
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Anna Di Rienzo
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
6
|
Peng Y, Nanduri J, Wang N, Khan SA, Pamenter M, Prabhakar NR. Carotid body responses to O 2 and CO 2 in hypoxia-tolerant naked mole rats. Acta Physiol (Oxf) 2022; 236:e13851. [PMID: 35757963 PMCID: PMC9787741 DOI: 10.1111/apha.13851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 02/02/2023]
Abstract
AIM Naked mole rats (NMRs) exhibit blunted hypoxic (HVR) and hypercapnic ventilatory responses (HCVR). The mechanism(s) underlying these responses are largely unknown. We hypothesized that attenuated carotid body (CB) sensitivity to hypoxia and hypercapnia contributes to the near absence of ventilatory responses to hypoxia and CO2 in NMRs. METHODS We measured ex vivo CB sensory nerve activity, phrenic nerve activity (an estimation of ventilation), and blood gases in urethane-anesthetized NMRs and C57BL/6 mice breathing normoxic, hypoxic, or hypercapnic gases. CB morphology, carbon monoxide, and H2 S levels were also determined. RESULTS Relative to mice, NMRs had blunted CB and HVR. Morphologically, NMRs have larger CBs, which contained more glomus cells than in mice. Furthermore, NMR glomus cells form a dispersed pattern compared to a clustered pattern in mice. Hemeoxygenase (HO)-1 mRNA was elevated in NMR CBs, and an HO inhibitor increased CB sensitivity to hypoxia in NMRs. This increase was blocked by an H2 S synthesis inhibitor, suggesting that interrupted gas messenger signaling contributes to the blunted CB responses and HVR in NMRs. Regarding hypercapnia, CB and ventilatory responses to CO2 in NMRs were larger than in mice. Carbonic anhydrase (CA)-2 mRNA is elevated in NMR CBs, and a CA inhibitor blocked the augmented CB response to CO2 in NMRs, indicating CA activity regulates augmented CB response to CO2 . CONCLUSIONS Consistent with our hypothesis, impaired CB responses to hypoxia contribute in part to the blunted HVR in NMRs. Conversely, the HCVR and CB are more sensitive to CO2 in NMRs.
Collapse
Affiliation(s)
- Ying‐Jie Peng
- Institute for Integrative Physiology and Center for Systems Biology of O2 SensingUniversity of ChicagoChicagoIllinoisUSA
| | - Jayasri Nanduri
- Institute for Integrative Physiology and Center for Systems Biology of O2 SensingUniversity of ChicagoChicagoIllinoisUSA
| | - Ning Wang
- Institute for Integrative Physiology and Center for Systems Biology of O2 SensingUniversity of ChicagoChicagoIllinoisUSA
| | - Shakil A. Khan
- Institute for Integrative Physiology and Center for Systems Biology of O2 SensingUniversity of ChicagoChicagoIllinoisUSA
| | - Matthew E. Pamenter
- Department of BiologyUniversity of OttawaOttawaOntarioCanada,University of Ottawa Brain and Mind Research InstituteOttawaOntarioCanada
| | - Nanduri R. Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 SensingUniversity of ChicagoChicagoIllinoisUSA
| |
Collapse
|
7
|
Verheyen M, Puschkarow M, Gnipp S, Koesling D, Peters M, Mergia E. The differential roles of the two NO-GC isoforms in adjusting airway reactivity. Am J Physiol Lung Cell Mol Physiol 2022; 323:L450-L463. [PMID: 35972838 PMCID: PMC9529264 DOI: 10.1152/ajplung.00404.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The enzyme, nitric oxide-sensitive guanylyl cyclase (NO-GC), is activated by binding NO to its prosthetic heme group and catalyzes the formation of cGMP. The NO-GC is primarily known to mediate vascular smooth muscle relaxation in the lung, and inhaled NO has been successfully used as a selective pulmonary vasodilator. In comparison, NO-GC’s impact on the regulation of airway tone is less acknowledged and, most importantly, little is known about the issue that NO-GC signaling is accomplished by two isoforms: NO-GC1 and NO-GC2, implying the existence of distinct “cGMP pools.” Herein, we investigated the functional role of the NO-GC isoforms in respiration by measuring lung function parameters of isoform-specific knockout (KO) mice using noninvasive and invasive techniques. Our data revealed the participation and ongoing influence of NO-GC1-derived cGMP in the regulation of airway tone by showing that respiratory resistance was enhanced in NO-GC1-KOs and increased more pronouncedly after the challenge with the bronchoconstrictor methacholine. The tissue resistance and stiffness of NO-GC1-KOs were also higher because of narrowed airways that cause tissue distortion. Contrariwise, NO-GC2-KOs displayed reduced tissue elasticity, elastic recoil, and airway reactivity to methacholine, which did not even increase in an ovalbumin model of asthma that induced hyperresponsiveness in NO-GC1-KOs. In addition, conscious NO-GC2-KOs showed a higher breathing rate with a shorter duration of inspiration and expiration time, which remained faster even in the presence of bronchoconstrictors that slow down breathing. Thus, we provide evidence of two distinct NO/cGMP pathways in airways, accomplished by either NO-GC1 or NO-GC2, adjusting differentially the airway reactivity.
Collapse
Affiliation(s)
- Malte Verheyen
- Institute of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany
| | - Michelle Puschkarow
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, NRW, Germany
| | - Stefanie Gnipp
- Department of Experimental Pneumology, Ruhr University Bochum, Bochum, NRW, Germany
| | - Doris Koesling
- Institute of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany
| | - Marcus Peters
- Department of Molecular Immunology, Ruhr University Bochum, Bochum, NRW, Germany
| | - Evanthia Mergia
- Institute of Pharmacology and Toxicology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
8
|
Bruggink S, Kentch K, Kronenfeld J, Renquist BJ. A Leak-Free Head-Out Plethysmography System to Accurately Assess Lung Function in Mice. J Appl Physiol (1985) 2022; 133:104-118. [DOI: 10.1152/japplphysiol.00835.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mice are a valuable model for elegant studies of complex, systems-dependent diseases, including pulmonary diseases. Current tools to assess lung function in mice are either terminal or lack accuracy. We set out to develop a low-cost, accurate, head-out variable-pressure plethysmography system to allow for repeated, non-terminal measurements of lung function in mice. Current head-out plethysmography systems are limited by air leaks that prevent accurate measures of volume and flow. We designed an inflatable cuff that encompasses the mouse's neck preventing air leak. We wrote corresponding software to collect and analyze the data, remove movement artifacts, and automatically calibrate each dataset. This software calculates inspiratory/expiratory volume, inspiratory/expiratory time, breaths per minute, mid-expiratory flow, and end-inspiratory pause. To validate the use, we established that our plethysmography system accurately measured tidal breathing, the bronchoconstrictive response to methacholine, sex and age associated changes in breathing, and breathing changes associated with house dust mite sensitization. Our estimates of volume, flow, and timing of breaths are in line with published estimates, we observed dose-dependent decreases in volume and flow in response to methacholine (P < 0.05), increased lung volume and decreased breathing rate with aging (P < 0.05), and that house dust mite sensitization decreased volume and flow (P <0.05) while exacerbating the methacholine induced increases in inspiratory and expiratory time (P < 0.05). We describe an accurate, sensitive, low-cost, head-out plethysmography system that allows for longitudinal studies of pulmonary disease in mice.
Collapse
Affiliation(s)
- Stephanie Bruggink
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
- Physiological Sciences GIDP, University of Arizona, Tucson, AZ, United States
| | - Kyle Kentch
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Jason Kronenfeld
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
| | - Benjamin Jennings Renquist
- Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, United States
- Physiological Sciences GIDP, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
9
|
Biophysicochemical studies of a ruthenium (II) nitrosyl thioether‐thiolate complex binding to BSA: Mechanistic information, molecular docking, and relationship to antibacterial and cytotoxic activities. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
10
|
Ventilatory responses during and following hypercapnic gas challenge are impaired in male but not female endothelial NOS knock-out mice. Sci Rep 2021; 11:20557. [PMID: 34663876 PMCID: PMC8523677 DOI: 10.1038/s41598-021-99922-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 09/24/2021] [Indexed: 11/28/2022] Open
Abstract
The roles of endothelial nitric oxide synthase (eNOS) in the ventilatory responses during and after a hypercapnic gas challenge (HCC, 5% CO2, 21% O2, 74% N2) were assessed in freely-moving female and male wild-type (WT) C57BL6 mice and eNOS knock-out (eNOS-/-) mice of C57BL6 background using whole body plethysmography. HCC elicited an array of ventilatory responses that were similar in male and female WT mice, such as increases in breathing frequency (with falls in inspiratory and expiratory times), and increases in tidal volume, minute ventilation, peak inspiratory and expiratory flows, and inspiratory and expiratory drives. eNOS-/- male mice had smaller increases in minute ventilation, peak inspiratory flow and inspiratory drive, and smaller decreases in inspiratory time than WT males. Ventilatory responses in female eNOS-/- mice were similar to those in female WT mice. The ventilatory excitatory phase upon return to room-air was similar in both male and female WT mice. However, the post-HCC increases in frequency of breathing (with decreases in inspiratory times), and increases in tidal volume, minute ventilation, inspiratory drive (i.e., tidal volume/inspiratory time) and expiratory drive (i.e., tidal volume/expiratory time), and peak inspiratory and expiratory flows in male eNOS-/- mice were smaller than in male WT mice. In contrast, the post-HCC responses in female eNOS-/- mice were equal to those of the female WT mice. These findings provide the first evidence that the loss of eNOS affects the ventilatory responses during and after HCC in male C57BL6 mice, whereas female C57BL6 mice can compensate for the loss of eNOS, at least in respect to triggering ventilatory responses to HCC.
Collapse
|
11
|
Getsy PM, Sundararajan S, May WJ, von Schill GC, McLaughlin DK, Palmer LA, Lewis SJ. Short-term facilitation of breathing upon cessation of hypoxic challenge is impaired in male but not female endothelial NOS knock-out mice. Sci Rep 2021; 11:18346. [PMID: 34526532 PMCID: PMC8443732 DOI: 10.1038/s41598-021-97322-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 08/09/2021] [Indexed: 02/08/2023] Open
Abstract
Decreases in arterial blood oxygen stimulate increases in minute ventilation via activation of peripheral and central respiratory structures. This study evaluates the role of endothelial nitric oxide synthase (eNOS) in the expression of the ventilatory responses during and following a hypoxic gas challenge (HXC, 10% O2, 90% N2) in freely moving male and female wild-type (WT) C57BL6 and eNOS knock-out (eNOS-/-) mice. Exposure to HXC caused an array of responses (of similar magnitude and duration) in both male and female WT mice such as, rapid increases in frequency of breathing, tidal volume, minute ventilation and peak inspiratory and expiratory flows, that were subject to pronounced roll-off. The responses to HXC in male eNOS-/- mice were similar to male WT mice. In contrast, several of the ventilatory responses in female eNOS-/- mice (e.g., frequency of breathing, and expiratory drive) were greater compared to female WT mice. Upon return to room-air, male and female WT mice showed similar excitatory ventilatory responses (i.e., short-term potentiation phase). These responses were markedly reduced in male eNOS-/- mice, whereas female eNOS-/- mice displayed robust post-HXC responses that were similar to those in female WT mice. Our data demonstrates that eNOS plays important roles in (1) ventilatory responses to HXC in female compared to male C57BL6 mice; and (2) expression of post-HXC responses in male, but not female C57BL6 mice. These data support existing evidence that sex, and the functional roles of specific proteins (e.g., eNOS) have profound influences on ventilatory processes, including the responses to HXC.
Collapse
Affiliation(s)
- Paulina M. Getsy
- grid.67105.350000 0001 2164 3847Department of Pediatrics, Biomedical Research Building BRB 319, Case Western Reserve University, 10900 Euclid Avenue Mail Stop 1714, Cleveland, OH 44106-1714 USA ,grid.67105.350000 0001 2164 3847Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH USA
| | - Sripriya Sundararajan
- grid.27755.320000 0000 9136 933XPediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA USA ,grid.411024.20000 0001 2175 4264Present Address: Division of Neonatology, Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Walter J. May
- grid.27755.320000 0000 9136 933XPediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Graham C. von Schill
- grid.27755.320000 0000 9136 933XPediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Dylan K. McLaughlin
- grid.27755.320000 0000 9136 933XPediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Lisa A. Palmer
- grid.27755.320000 0000 9136 933XPediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA USA
| | - Stephen J. Lewis
- grid.67105.350000 0001 2164 3847Department of Pediatrics, Biomedical Research Building BRB 319, Case Western Reserve University, 10900 Euclid Avenue Mail Stop 1714, Cleveland, OH 44106-1714 USA ,grid.67105.350000 0001 2164 3847Department of Pharmacology, Case Western Reserve University, Cleveland, OH USA ,grid.67105.350000 0001 2164 3847Functional Electrical Stimulation Center, Case Western Reserve University, Cleveland, OH USA
| |
Collapse
|
12
|
Peng YJ, Gridina A, Wang B, Nanduri J, Fox AP, Prabhakar NR. Olfactory receptor 78 participates in carotid body response to a wide range of low O 2 levels but not severe hypoxia. J Neurophysiol 2020; 123:1886-1895. [PMID: 32208891 DOI: 10.1152/jn.00075.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The role of olfactory receptor 78 (Olfr78) in carotid body (CB) response to hypoxia was examined. BL6 mice with global deletion of Olfr78 manifested an impaired hypoxic ventilatory response (HVR), a hallmark of the CB chemosensory reflex, CB sensory nerve activity, and reduced intracellular Ca2+ concentration ([Ca2+]i) response of glomus cells to hypoxia (Po2 ~ 40 mmHg). In contrast, severe hypoxia (Po2 ~ 10 mmHg) depressed breathing and produced a very weak CB sensory nerve excitation but robust elevation of [Ca2+]i in Olfr78 null glomus cells. CB sensory nerve excitation evoked by Olfr78 ligands, lactate, propionate, acetate, and butyrate were unaffected in mutant mice and were smaller than that evoked by hypoxia (Po2 ~ 40mmHg). Similar results were obtained in Olfr78 null mice on a JAX genetic background. These results demonstrate a role for Olfr78 in CB responses to a wide range of hypoxia, but not severe hypoxia, and do not require either lactate or any other short-chain fatty acids.NEW & NOTEWORTHY The current study demonstrates that olfactory receptor 78 (Olfr78), a G protein-coupled receptor, is an integral component of the hypoxic sensing mechanism of the carotid body to a wide range of low oxygen levels, but not severe hypoxia, and that Olfr78 participation does not require either lactate or any other short-chain fatty acids, proposed ligands of Olfr78.
Collapse
Affiliation(s)
- Ying-Jie Peng
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| | - Anna Gridina
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| | - Benjamin Wang
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| | - Jayasri Nanduri
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| | - Aaron P Fox
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| | - Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| |
Collapse
|
13
|
Fields DP, Roberts BM, Simon AK, Judge AR, Fuller DD, Mitchell GS. Cancer cachexia impairs neural respiratory drive in hypoxia but not hypercapnia. J Cachexia Sarcopenia Muscle 2019; 10:63-72. [PMID: 30362273 PMCID: PMC6438337 DOI: 10.1002/jcsm.12348] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/26/2018] [Accepted: 08/19/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Cancer cachexia is an insidious process characterized by muscle atrophy with associated motor deficits, including diaphragm weakness and respiratory insufficiency. Although neuropathology contributes to muscle wasting and motor deficits in many clinical disorders, neural involvement in cachexia-linked respiratory insufficiency has not been explored. METHODS We first used whole-body plethysmography to assess ventilatory responses to hypoxic and hypercapnic chemoreflex activation in mice inoculated with the C26 colon adenocarcinoma cell line. Mice were exposed to a sequence of inspired gas mixtures consisting of (i) air, (ii) hypoxia (11% O2 ) with normocapnia, (iii) hypercapnia (7% CO2 ) with normoxia, and (iv) combined hypercapnia with hypoxia (i.e. maximal chemoreflex response). We also tested the respiratory neural network directly by recording inspiratory burst output from ligated phrenic nerves, thereby bypassing influences from changes in diaphragm muscle strength, respiratory mechanics, or compensation through recruitment of accessory motor pools. RESULTS Cachectic mice demonstrated a significant attenuation of the hypoxic tidal volume (0.26mL±0.01mL vs 0.30mL±0.01mL; p<0.05), breathing frequency (317±10bpm vs 344±6bpm; p<0.05) and phrenic nerve (29.5±2.6% vs 78.8±11.8%; p<0.05) responses. On the other hand, the much larger hypercapnic tidal volume (0.46±0.01mL vs 0.46±0.01mL; p>0.05), breathing frequency (392±5bpm vs 408±5bpm; p>0.05) and phrenic nerve (93.1±8.8% vs 111.1±13.2%; p>0.05) responses were not affected. Further, the concurrent hypercapnia/hypoxia tidal volume (0.45±0.01mL vs 0.45±0.01mL; p>0.05), breathing frequency (395±7bpm vs 400±3bpm; p>0.05), and phrenic nerve (106.8±7.1% vs 147.5±38.8%; p>0.05) responses were not different between C26 cachectic and control mice. CONCLUSIONS Breathing deficits associated with cancer cachexia are specific to the hypoxic ventilatory response and, thus, reflect disruptions in the hypoxic chemoafferent neural network. Diagnostic techniques that detect decompensation and therapeutic approaches that support the failing hypoxic respiratory response may benefit patients at risk for cancer cachectic-associated respiratory failure.
Collapse
Affiliation(s)
- Daryl P Fields
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Brandon M Roberts
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Alec K Simon
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Andrew R Judge
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - David D Fuller
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Gordon S Mitchell
- Center for Respiratory Research and Rehabilitation, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
14
|
Peng YJ, Makarenko VV, Gridina A, Chupikova I, Zhang X, Kumar GK, Fox AP, Prabhakar NR. H 2S mediates carotid body response to hypoxia but not anoxia. Respir Physiol Neurobiol 2019; 259:75-85. [PMID: 30086385 PMCID: PMC6252114 DOI: 10.1016/j.resp.2018.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/16/2018] [Accepted: 08/03/2018] [Indexed: 02/05/2023]
Abstract
The role of cystathionine-γ-lyase (CSE) derived H2S in the hypoxic and anoxic responses of the carotid body (CB) were examined. Experiments were performed on Sprague-Dawley rats, wild type and CSE knockout mice on C57BL/6 J background. Hypoxia (pO2 = 37 ± 3 mmHg) increased the CB sensory nerve activity and elevated H2S levels in rats. In contrast, anoxia (pO2 = 5 ± 4 mmHg) produced only a modest CB sensory excitation with no change in H2S levels. DL-propargylglycine (DL-PAG), a blocker of CSE, inhibited hypoxia but not anoxia-evoked CB sensory excitation and [Ca2+]i elevation of glomus cells. The inhibitory effects of DL-PAG on hypoxia were seen: a) when it is dissolved in saline but not in dimethyl sulfoxide (DMSO), and b) in glomus cells cultured for18 h but not in cells either soon after isolation or after prolonged culturing (72 h) requiring 1-3 h of incubation. On the other hand, anoxia-induced [Ca2+]i responses of glomus cell were blocked by high concentration of DL-PAG (300μM) either alone or in combination with aminooxyacetic acid (AOAA; 300μM) with a decreased cell viability. Anoxia produced a weak CB sensory excitation and robust [Ca2+]i elevation in glomus cells of both wild-type and CSE null mice. As compared to wild-type, CSE null mice exhibited impaired CB chemo reflex as evidenced by attenuated efferent phrenic nerve responses to brief hyperoxia (Dejours test), and hypoxia. Inhalation of 100% N2 (anoxia) depressed breathing in both CSE null and wild-type mice. These observations demonstrate that a) hypoxia and anoxia are not analogous stimuli for studying CB physiology and b) CSE-derived H2S contributes to CB response to hypoxia but not to that of anoxia.
Collapse
Affiliation(s)
- Ying-Jie Peng
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA.
| | - Vladislav V Makarenko
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA
| | - Anna Gridina
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA
| | - Irina Chupikova
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA
| | - Xiuli Zhang
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA
| | - Ganesh K Kumar
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA
| | - Aaron P Fox
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA
| | - Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2Sensing, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
15
|
Gonzalez NC, Kuwahira I. Systemic Oxygen Transport with Rest, Exercise, and Hypoxia: A Comparison of Humans, Rats, and Mice. Compr Physiol 2018; 8:1537-1573. [PMID: 30215861 DOI: 10.1002/cphy.c170051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The objective of this article is to compare and contrast the known characteristics of the systemic O2 transport of humans, rats, and mice at rest and during exercise in normoxia and hypoxia. This analysis should help understand when rodent O2 transport findings can-and cannot-be applied to human responses to similar conditions. The O2 -transport system was analyzed as composed of four linked conductances: ventilation, alveolo-capillary diffusion, circulatory convection, and tissue capillary-cell diffusion. While the mechanisms of O2 transport are similar in the three species, the quantitative differences are naturally large. There are abundant data on total O2 consumption and on ventilatory and pulmonary diffusive conductances under resting conditions in the three species; however, there is much less available information on pulmonary gas exchange, circulatory O2 convection, and tissue O2 diffusion in mice. The scarcity of data largely derives from the difficulty of obtaining blood samples in these small animals and highlights the need for additional research in this area. In spite of the large quantitative differences in absolute and mass-specific O2 flux, available evidence indicates that resting alveolar and arterial and venous blood PO2 values under normoxia are similar in the three species. Additionally, at least in rats, alveolar and arterial blood PO2 under hypoxia and exercise remain closer to the resting values than those observed in humans. This is achieved by a greater ventilatory response, coupled with a closer value of arterial to alveolar PO2 , suggesting a greater efficacy of gas exchange in the rats. © 2018 American Physiological Society. Compr Physiol 8:1537-1573, 2018.
Collapse
Affiliation(s)
- Norberto C Gonzalez
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ichiro Kuwahira
- Department of Pulmonary Medicine, Tokai University School of Medicine, Tokai University Tokyo Hospital, Tokyo, Japan
| |
Collapse
|
16
|
Pamenter ME, Powell FL. Time Domains of the Hypoxic Ventilatory Response and Their Molecular Basis. Compr Physiol 2016; 6:1345-85. [PMID: 27347896 DOI: 10.1002/cphy.c150026] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ventilatory responses to hypoxia vary widely depending on the pattern and length of hypoxic exposure. Acute, prolonged, or intermittent hypoxic episodes can increase or decrease breathing for seconds to years, both during the hypoxic stimulus, and also after its removal. These myriad effects are the result of a complicated web of molecular interactions that underlie plasticity in the respiratory control reflex circuits and ultimately control the physiology of breathing in hypoxia. Since the time domains of the physiological hypoxic ventilatory response (HVR) were identified, considerable research effort has gone toward elucidating the underlying molecular mechanisms that mediate these varied responses. This research has begun to describe complicated and plastic interactions in the relay circuits between the peripheral chemoreceptors and the ventilatory control circuits within the central nervous system. Intriguingly, many of these molecular pathways seem to share key components between the different time domains, suggesting that varied physiological HVRs are the result of specific modifications to overlapping pathways. This review highlights what has been discovered regarding the cell and molecular level control of the time domains of the HVR, and highlights key areas where further research is required. Understanding the molecular control of ventilation in hypoxia has important implications for basic physiology and is emerging as an important component of several clinical fields. © 2016 American Physiological Society. Compr Physiol 6:1345-1385, 2016.
Collapse
Affiliation(s)
| | - Frank L Powell
- Physiology Division, Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
17
|
The sensing of respiratory gases in fish: Mechanisms and signalling pathways. Respir Physiol Neurobiol 2016; 224:71-9. [DOI: 10.1016/j.resp.2015.06.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/16/2015] [Accepted: 06/17/2015] [Indexed: 12/29/2022]
|
18
|
King TL, Ruyle BC, Kline DD, Heesch CM, Hasser EM. Catecholaminergic neurons projecting to the paraventricular nucleus of the hypothalamus are essential for cardiorespiratory adjustments to hypoxia. Am J Physiol Regul Integr Comp Physiol 2015; 309:R721-31. [PMID: 26157062 PMCID: PMC4666929 DOI: 10.1152/ajpregu.00540.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 07/01/2015] [Indexed: 12/31/2022]
Abstract
Brainstem catecholamine neurons modulate sensory information and participate in control of cardiorespiratory function. These neurons have multiple projections, including to the paraventricular nucleus (PVN), which contributes to cardiorespiratory and neuroendocrine responses to hypoxia. We have shown that PVN-projecting catecholaminergic neurons are activated by hypoxia, but the function of these neurons is not known. To test the hypothesis that PVN-projecting catecholamine neurons participate in responses to respiratory challenges, we injected IgG saporin (control; n = 6) or anti-dopamine β-hydroxylase saporin (DSAP; n = 6) into the PVN to retrogradely lesion catecholamine neurons projecting to the PVN. After 2 wk, respiratory measurements (plethysmography) were made in awake rats during normoxia, increasing intensities of hypoxia (12, 10, and 8% O2) and hypercapnia (5% CO2-95% O2). DSAP decreased the number of tyrosine hydroxylase-immunoreactive terminals in PVN and cells counted in ventrolateral medulla (VLM; -37%) and nucleus tractus solitarii (nTS; -36%). DSAP produced a small but significant decrease in respiratory rate at baseline (during normoxia) and at all intensities of hypoxia. Tidal volume and minute ventilation (VE) index also were impaired at higher hypoxic intensities (10-8% O2; e.g., VE at 8% O2: IgG = 181 ± 22, DSAP = 91 ± 4 arbitrary units). Depressed ventilation in DSAP rats was associated with significantly lower arterial O2 saturation at all hypoxic intensities. PVN DSAP also reduced ventilatory responses to 5% CO2 (VE: IgG = 176 ± 21 and DSAP = 84 ± 5 arbitrary units). Data indicate that catecholamine neurons projecting to the PVN are important for peripheral and central chemoreflex respiratory responses and for maintenance of arterial oxygen levels during hypoxic stimuli.
Collapse
Affiliation(s)
| | | | - David D Kline
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, and
| | - Cheryl M Heesch
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, and
| | - Eileen M Hasser
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, and Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
19
|
Hartmann SE, Kissel CK, Szabo L, Walker BL, Leigh R, Anderson TJ, Poulin MJ. Increased ventilatory response to carbon dioxide in COPD patients following vitamin C administration. ERJ Open Res 2015; 1:00017-2015. [PMID: 27730137 PMCID: PMC5005137 DOI: 10.1183/23120541.00017-2015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 07/12/2015] [Indexed: 11/17/2022] Open
Abstract
Patients with chronic obstructive pulmonary disease (COPD) have decreased ventilatory and cerebrovascular responses to hypercapnia. Antioxidants increase the ventilatory response to hypercapnia in healthy humans. Cerebral blood flow is an important determinant of carbon dioxide/hydrogen ion concentration at the central chemoreceptors and may be affected by antioxidants. It is unknown whether antioxidants can improve the ventilatory and cerebral blood flow response in individuals in whom these are diminished. Thus, we aimed to determine the effect of vitamin C administration on the ventilatory and cerebrovascular responses to hypercapnia during healthy ageing and in COPD. Using transcranial Doppler ultrasound, we measured the ventilatory and cerebral blood flow responses to hyperoxic hypercapnia before and after an intravenous vitamin C infusion in healthy young (Younger) and older (Older) subjects and in moderate COPD. Vitamin C increased the ventilatory response in COPD patients (mean (95% CI) 1.1 (0.9-1.1) versus 1.5 (1.1-2.0) L·min-1·mmHg-1, p<0.05) but not in Younger (2.5 (1.9-3.1) versus 2.4 (1.9-2.9) L·min-1·mmHg-1, p>0.05) or Older (1.3 (1.0-1.7) versus 1.3 (1.0-1.7) L·min-1·mmHg-1, p>0.05) healthy subjects. Vitamin C did not affect the cerebral blood flow response in the young or older healthy subjects or COPD subjects (p>0.05). Vitamin C increases the ventilatory but not cerebrovascular response to hyperoxic hypercapnia in patients with moderate COPD.
Collapse
Affiliation(s)
- Sara E. Hartmann
- Dept of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Christine K. Kissel
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Lian Szabo
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Medicine, University of Calgary, Calgary, AB, Canada
| | - Brandie L. Walker
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Richard Leigh
- Dept of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Todd J. Anderson
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Dept of Cardiac Sciences, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
- Dept of Medicine, University of Calgary, Calgary, AB, Canada
| | - Marc J. Poulin
- Dept of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
- Dept of Clinical Neuroscience, University of Calgary, Calgary, AB, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
20
|
Pamenter ME, Go A, Fu Z, Powell FL. No evidence of a role for neuronal nitric oxide synthase in the nucleus tractus solitarius in ventilatory responses to acute or chronic hypoxia in awake rats. J Appl Physiol (1985) 2015; 118:750-9. [PMID: 25571988 PMCID: PMC4360023 DOI: 10.1152/japplphysiol.00333.2014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 01/02/2015] [Indexed: 11/22/2022] Open
Abstract
When exposed to a hypoxic environment, the body's first response is a reflex increase in ventilation, termed the hypoxic ventilatory response (HVR). With chronic sustained hypoxia (CSH), such as during acclimatization to high altitude, an additional time-dependent increase in ventilation occurs, which increases the HVR and is termed ventilatory acclimatization to hypoxia (VAH). This secondary increase persists after exposure to CSH and involves plasticity within the circuits in the central nervous system that control breathing. The mechanisms of HVR plasticity are currently poorly understood. We hypothesized that changes in neuronal nitric oxide synthase (nNOS) activity or expression in the nucleus tractus solitarius contribute to this plasticity and underlie VAH in rats. To test this, we treated rats held in normoxia or 10% O2 (CSH, PIO2 = 70 Torr) for 7-9 days and measured ventilation in conscious, unrestrained animals before and after microinjecting the general NOS antagonist L-NG-Nitroarginine methyl ester into the nucleus tractus solitarius (NTS) or systemically injecting the nNOS-specific antagonist S-methyl-l-thiocitrulline. Localization of injection sites in the NTS was confirmed by histology following the experiment. We found that 1) neither NTS-specific nor systemic nNOS antagonism had any effect on hypoxia-mediated changes in breathing or metabolism (P > 0.05), but 2) nNOS protein expression was increased in the middle and caudal NTS by CSH. A persistent HVR after nNOS blockade in the NTS contrasts with results in awake mice, and our findings do not support the hypotheses that nNOS in the NTS contribute to the HVR or VAH in awake rats.
Collapse
Affiliation(s)
- Matthew E Pamenter
- Division of Physiology, Department of Medicine, University of California, San Diego, La Jolla, California; and Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ariel Go
- Division of Physiology, Department of Medicine, University of California, San Diego, La Jolla, California; and
| | - Zhenxing Fu
- Division of Physiology, Department of Medicine, University of California, San Diego, La Jolla, California; and
| | - Frank L Powell
- Division of Physiology, Department of Medicine, University of California, San Diego, La Jolla, California; and
| |
Collapse
|
21
|
Palmer LA, Kimberly deRonde, Brown-Steinke K, Gunter S, Jyothikumar V, Forbes MS, Lewis SJ. Hypoxia-induced changes in protein s-nitrosylation in female mouse brainstem. Am J Respir Cell Mol Biol 2015; 52:37-45. [PMID: 24922346 DOI: 10.1165/rcmb.2013-0359oc] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Exposure to hypoxia elicits an increase in minute ventilation that diminishes during continued exposure (roll-off). Brainstem N-methyl-D-aspartate receptors (NMDARs) and neuronal nitric oxide synthase (nNOS) contribute to the initial hypoxia-induced increases in minute ventilation. Roll-off is regulated by platelet-derived growth factor receptor-β (PDGFR-β) and S-nitrosoglutathione (GSNO) reductase (GSNOR). S-nitrosylation inhibits activities of NMDAR and nNOS, but enhances GSNOR activity. The importance of S-nitrosylation in the hypoxic ventilatory response is unknown. This study confirms that ventilatory roll-off is virtually absent in female GSNOR(+/-) and GSNO(-/-) mice, and evaluated the location of GSNOR in female mouse brainstem, and temporal changes in GSNOR activity, protein expression, and S-nitrosylation status of GSNOR, NMDAR (1, 2A, 2B), nNOS, and PDGFR-β during hypoxic challenge. GSNOR-positive neurons were present throughout the brainstem, including the nucleus tractus solitarius. Protein abundances for GSNOR, nNOS, all NMDAR subunits and PDGFR-β were not altered by hypoxia. GSNOR activity and S-nitrosylation status temporally increased with hypoxia. In addition, nNOS S-nitrosylation increased with 3 and 15 minutes of hypoxia. Changes in NMDAR S-nitrosylation were detected in NMDAR 2B at 15 minutes of hypoxia. No hypoxia-induced changes in PDGFR-β S-nitrosylation were detected. However, PDGFR-β phosphorylation increased in the brainstems of wild-type mice during hypoxic exposure (consistent with roll-off), whereas it did not rise in GSNOR(+/-) mice (consistent with lack of roll-off). These data suggest that: (1) S-nitrosylation events regulate hypoxic ventilatory response; (2) increases in S-nitrosylation of NMDAR 2B, nNOS, and GSNOR may contribute to ventilatory roll-off; and (3) GSNOR regulates PDGFR-β phosphorylation.
Collapse
Affiliation(s)
- Lisa A Palmer
- 1 Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia
| | | | | | | | | | | | | |
Collapse
|
22
|
Porteus CS, Pollack J, Tzaneva V, Kwong RW, Kumai Y, Abdallah SJ, Zaccone G, Lauriano ER, Milsom WK, Perry SF. A role for nitric oxide in the control of breathing in zebrafish (Danio rerio). J Exp Biol 2015; 218:3746-53. [DOI: 10.1242/jeb.127795] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/28/2015] [Indexed: 11/20/2022]
Abstract
Nitric oxide (NO) is a gaseous neurotransmitter, which in adult mammals, modulates the acute hypoxic ventilatory response; its role in the control of breathing in fish during development is unknown. We addressed the interactive effects of developmental age and NO in the control of piscine breathing by measuring the ventilatory response of zebrafish (Danio rerio) adults and larvae to NO donors and by inhibiting endogenous production of NO. In adults, sodium nitroprusside (SNP), a NO donor, inhibited ventilation; the extent of the ventilatory inhibition was related to the pre-existing ventilatory drive, with the greatest inhibition exhibited during exposure to hypoxia (PO2=5.6 kPa). Inhibition of endogenous NO production using L-NAME supressed the hypoventilatory response to hyperoxia, supporting an inhibitory role of NO in adult zebrafish. Neuroepithelial cells, the putative oxygen chemoreceptors of fish, contain neuronal nitric oxide synthase (nNOS). In zebrafish larvae at 4 days post fertilization, SNP increased ventilation in a concentration-dependent manner. Inhibition of NOS activity with L-NAME or knockdown of nNOS inhibited the hypoxic (PO2=3.5 kPa) ventilatory response. Immunohistochemistry revealed the presence of nNOS in the NECs of larvae. Taken together, these data suggest that NO plays an inhibitory role in the control ventilation in adult zebrafish, but an excitatory role in larvae.
Collapse
Affiliation(s)
- Cosima S. Porteus
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Jacob Pollack
- Department of Biology, University of Ottawa, ON, K1N 6N5, Canada
| | | | | | - Yusuke Kumai
- Department of Biology, University of Ottawa, ON, K1N 6N5, Canada
| | - Sara J. Abdallah
- Department of Biology, University of Ottawa, ON, K1N 6N5, Canada
| | - Giacomo Zaccone
- Department of Food and Environmental Science, Messina University, I-98166, Italy
| | | | - William K. Milsom
- Department of Zoology, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Steve F. Perry
- Department of Biology, University of Ottawa, ON, K1N 6N5, Canada
| |
Collapse
|
23
|
Prabhakar NR, Peers C. Gasotransmitter regulation of ion channels: a key step in O2 sensing by the carotid body. Physiology (Bethesda) 2014; 29:49-57. [PMID: 24382871 PMCID: PMC3929115 DOI: 10.1152/physiol.00034.2013] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Carotid bodies detect hypoxia in arterial blood, translating this stimulus into physiological responses via the CNS. It is long established that ion channels are critical to this process. More recent evidence indicates that gasotransmitters exert powerful influences on O2 sensing by the carotid body. Here, we review current understanding of hypoxia-dependent production of gasotransmitters, how they regulate ion channels in the carotid body, and how this impacts carotid body function.
Collapse
Affiliation(s)
- Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, Biological Sciences Division, University of Chicago, Chicago, Illinois; and
| | | |
Collapse
|
24
|
Ruthenium complexes as NO donors for vascular relaxation induction. Molecules 2014; 19:9628-54. [PMID: 25004072 PMCID: PMC6271244 DOI: 10.3390/molecules19079628] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/09/2014] [Accepted: 06/26/2014] [Indexed: 11/17/2022] Open
Abstract
Nitric oxide (NO) donors are substances that can release NO. Vascular relaxation induction is among the several functions of NO, and the administration of NO donors is a pharmacological alternative to treat hypertension. This review will focus on the physicochemical description of ruthenium-derived NO donor complexes that release NO via reduction and light stimulation. In particular, we will discuss the complexes synthesized by our research group over the last ten years, and we will focus on the vasodilation and arterial pressure control elicited by these complexes. Soluble guanylyl cyclase (sGC) and potassium channels are the main targets of the NO species released from the inorganic compounds. We will consider the importance of the chemical structure of the ruthenium complexes and their vascular effects.
Collapse
|
25
|
Mendoza JP, Passafaro RJ, Baby SM, Young AP, Bates JN, Gaston B, Lewis SJ. Role of nitric oxide-containing factors in the ventilatory and cardiovascular responses elicited by hypoxic challenge in isoflurane-anesthetized rats. J Appl Physiol (1985) 2014; 116:1371-81. [PMID: 24744389 DOI: 10.1152/japplphysiol.00842.2013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Exposure to hypoxia elicits changes in mean arterial blood pressure (MAP), heart rate, and frequency of breathing (fR). The objective of this study was to determine the role of nitric oxide (NO) in the cardiovascular and ventilatory responses elicited by brief exposures to hypoxia in isoflurane-anesthetized rats. The rats were instrumented to record MAP, heart rate, and fR and then exposed to 90 s episodes of hypoxia (10% O2, 90% N2) before and after injection of vehicle, the NO synthase inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME), or the inactive enantiomer D-NAME (both at 50 μmol/kg iv). Each episode of hypoxia elicited a decrease in MAP, bidirectional changes in heart rate (initial increase and then a decrease), and an increase in fR. These responses were similar before and after injection of vehicle or D-NAME. In contrast, the hypoxia-induced decreases in MAP were attenuated after administration of L-NAME. The initial increases in heart rate during hypoxia were amplified whereas the subsequent decreases in heart rate were attenuated in L-NAME-treated rats. Finally, the hypoxia-induced increases in fR were virtually identical before and after administration of L-NAME. These findings suggest that NO factors play a vital role in the expression of the cardiovascular but not the ventilatory responses elicited by brief episodes of hypoxia in isoflurane-anesthetized rats. Based on existing evidence that NO factors play a vital role in carotid body and central responses to hypoxia in conscious rats, our findings raise the novel possibility that isoflurane blunts this NO-dependent signaling.
Collapse
Affiliation(s)
- James P Mendoza
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Rachael J Passafaro
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Santhosh M Baby
- Division of Biology, Galleon Pharmaceuticals, Horsham, Pennsylvania
| | - Alex P Young
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - James N Bates
- Department of Anesthesia, University of Iowa Hospitals and Clinics, Iowa City, Iowa; and
| | - Benjamin Gaston
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Stephen J Lewis
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
26
|
Lowe M, Park SJ, Nurse CA, Campanucci VA. Purinergic stimulation of carotid body efferent glossopharyngeal neurones increases intracellular Ca2+ and nitric oxide production. Exp Physiol 2013; 98:1199-212. [PMID: 23525247 DOI: 10.1113/expphysiol.2013.072058] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mammalian carotid body (CB) is a peripheral chemosensory organ that controls ventilation and is innervated by both afferent and efferent nerve fibres. The afferent pathway is stimulated by chemoexcitants, such as hypoxia, hypercapnia and acidosis. The efferent pathway causes inhibition of the sensory discharge via release of NO that originates mainly from neuronal nitric oxide synthase (nNOS)-positive autonomic neurones within the glossopharyngeal nerve (GPN). Recent studies in the rat indicate that these inhibitory GPN neurones and their processes express purinergic P2X receptors and can be activated by ATP, a key excitatory CB neurotransmitter. Here we tested the hypothesis that purinergic agonists stimulate a rise in [Ca(2+)]i, leading to nNOS activation and NO production in isolated GPN neurones, using the fluorescent probes fura-2 and 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM DA), respectively. ATP caused a dose-dependent increase in [Ca(2+)]i in GPN neurones (EC50 ≈ 1.92 μm) that was markedly inhibited by a combination of 100 μm suramin (a non-specific P2X blocker) and 100 nm Brilliant Blue G (a selective P2X7 blocker). ATP also stimulated NO production in GPN neurones, as revealed by an increase in DAF fluorescence; this NO signal was inhibited by purinergic blockers, chelators of extracellular Ca(2+), the nNOS inhibitor l-NAME and the NO scavenger carboxy-PTIO. The P2X2/3 and P2X7 agonists α,β,-methylene ATP and benzoyl ATP mimicked the effects of ATP. Taken together, these data indicate that ATP may contribute to negative feedback inhibition of CB sensory discharge via purinergic stimulation of NO production in efferent fibres.
Collapse
Affiliation(s)
- Michael Lowe
- Department of Biology, McMaster University, Hamilton, Ontario, Canada L8S 4K1
| | | | | | | |
Collapse
|
27
|
Ai H, Huang L, Ren J. Genetic diversity, linkage disequilibrium and selection signatures in chinese and Western pigs revealed by genome-wide SNP markers. PLoS One 2013; 8:e56001. [PMID: 23409110 PMCID: PMC3567019 DOI: 10.1371/journal.pone.0056001] [Citation(s) in RCA: 172] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 01/03/2013] [Indexed: 11/18/2022] Open
Abstract
To investigate population structure, linkage disequilibrium (LD) pattern and selection signature at the genome level in Chinese and Western pigs, we genotyped 304 unrelated animals from 18 diverse populations using porcine 60 K SNP chips. We confirmed the divergent evolution between Chinese and Western pigs and showed distinct topological structures of the tested populations. We acquired the evidence for the introgression of Western pigs into two Chinese pig breeds. Analysis of runs of homozygosity revealed that historical inbreeding reduced genetic variability in several Chinese breeds. We found that intrapopulation LD extents are roughly comparable between Chinese and Western pigs. However, interpopulation LD is much longer in Western pigs compared with Chinese pigs with average r(2) (0.3) values of 125 kb for Western pigs and only 10.5 kb for Chinese pigs. The finding indicates that higher-density markers are required to capture LD with causal variants in genome-wide association studies and genomic selection on Chinese pigs. Further, we looked across the genome to identify candidate loci under selection using F(ST) outlier tests on two contrast samples: Tibetan pigs versus lowland pigs and belted pigs against non-belted pigs. Interestingly, we highlighted several genes including ADAMTS12, SIM1 and NOS1 that show signatures of natural selection in Tibetan pigs and are likely important for genetic adaptation to high altitude. Comparison of our findings with previous reports indicates that the underlying genetic basis for high-altitude adaptation in Tibetan pigs, Tibetan peoples and yaks is likely distinct from one another. Moreover, we identified the strongest signal of directional selection at the EDNRB loci in Chinese belted pigs, supporting EDNRB as a promising candidate gene for the white belt coat color in Chinese pigs. Altogether, our findings advance the understanding of the genome biology of Chinese and Western pigs.
Collapse
Affiliation(s)
- Huashui Ai
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, China
| | - Lusheng Huang
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, China
- * E-mail: (JR); (LH)
| | - Jun Ren
- Key Laboratory for Animal Biotechnology of Jiangxi Province and the Ministry of Agriculture of China, Jiangxi Agricultural University, Nanchang, China
- * E-mail: (JR); (LH)
| |
Collapse
|
28
|
Modulation of the carotid body sensory discharge by NO: An up-dated hypothesis. Respir Physiol Neurobiol 2012; 184:149-57. [DOI: 10.1016/j.resp.2012.04.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/08/2012] [Accepted: 04/15/2012] [Indexed: 11/23/2022]
|
29
|
Nurse CA, Piskuric NA. Signal processing at mammalian carotid body chemoreceptors. Semin Cell Dev Biol 2012; 24:22-30. [PMID: 23022231 DOI: 10.1016/j.semcdb.2012.09.006] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 09/19/2012] [Indexed: 10/27/2022]
Abstract
Mammalian carotid bodies are richly vascularized chemosensory organs that sense blood levels of O(2), CO(2)/H(+), and glucose and maintain homeostatic regulation of these levels via the reflex control of ventilation. Carotid bodies consist of innervated clusters of type I (or glomus) cells in intimate association with glial-like type II cells. Carotid bodies make afferent connections with fibers from sensory neurons in the petrosal ganglia and receive efferent inhibitory innervation from parasympathetic neurons located in the carotid sinus and glossopharyngeal nerves. There are synapses between type I (chemosensory) cells and petrosal afferent terminals, as well as between neighboring type I cells. There is a broad array of neurotransmitters and neuromodulators and their ionotropic and metabotropic receptors in the carotid body. This allows for complex processing of sensory stimuli (e.g., hypoxia and acid hypercapnia) involving both autocrine and paracrine signaling pathways. This review summarizes and evaluates current knowledge of these pathways and presents an integrated working model on information processing in carotid bodies. Included in this model is a novel hypothesis for a potential role of type II cells as an amplifier for the release of a key excitatory carotid body neurotransmitter, ATP, via P2Y purinoceptors and pannexin-1 channels.
Collapse
Affiliation(s)
- Colin A Nurse
- Department of Biology, McMaster University, 1280 Main St. W., Hamilton, Ontario, Canada L8S 4K1.
| | | |
Collapse
|
30
|
Affiliation(s)
- Gregg L Semenza
- Vascular Program, Institute for Cell Engineering, Department of Pediatrics, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | |
Collapse
|
31
|
Prabhakar NR, Semenza GL. Gaseous messengers in oxygen sensing. J Mol Med (Berl) 2012; 90:265-72. [PMID: 22349394 DOI: 10.1007/s00109-012-0876-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 01/30/2012] [Accepted: 02/01/2012] [Indexed: 12/23/2022]
Abstract
The carotid body is a sensory organ that detects acute changes in arterial blood oxygen (O(2)) levels and reflexly mediates systemic cardiac, vascular, and respiratory responses to hypoxia. This article provides a brief update of the roles of gas messengers as well as redox homeostasis by hypoxia-inducible factors (HIFs) in hypoxic sensing by the carotid body. Carbon monoxide (CO) and nitric oxide (NO), generated by heme oxygenase-2 (HO-2) and neuronal nitric oxide synthase (nNOS), respectively, inhibit carotid body activity. Molecular O(2) is a required substrate for the enzymatic activities of HO-2 and nNOS. Stimulation of carotid body activity by hypoxia may reflect reduced formation of CO and NO. Glomus cells, the site of O(2) sensing in the carotid body, express cystathionine γ-lyase (CSE), an H(2)S generating enzyme. Cth ( -/- ) mice, which lack CSE, exhibit severely impaired hypoxia-induced H(2)S generation, sensory excitation, and stimulation of breathing in response to low O(2). Hypoxia-evoked H(2)S generation in the carotid body requires the interaction of CSE with HO-2, which generates CO. Carotid bodies from Hif1a ( +/- ) mice with partial HIF-1α deficiency do not respond to hypoxia, whereas carotid bodies from mice with partial HIF-2α deficiency are hyper-responsive to hypoxia. The opposing roles of HIF-1α and HIF-2α in the carotid body have provided novel insight into molecular mechanisms of redox homeostasis and its role in hypoxia sensing. Heightened carotid body activity has been implicated in the pathogenesis of autonomic morbidities associated with sleep-disordered breathing, congestive heart failure, and essential hypertension. The enzymes that generate gas messengers and redox regulation by HIFs represent potential therapeutic targets for normalizing carotid body function and downstream autonomic output in these disease states.
Collapse
Affiliation(s)
- Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
32
|
A robust experimental protocol for pharmacological fMRI in rats and mice. J Neurosci Methods 2012; 204:9-18. [DOI: 10.1016/j.jneumeth.2011.10.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/20/2011] [Accepted: 10/22/2011] [Indexed: 01/03/2023]
|
33
|
Abstract
The discovery of the sensory nature of the carotid body dates back to the beginning of the 20th century. Following these seminal discoveries, research into carotid body mechanisms moved forward progressively through the 20th century, with many descriptions of the ultrastructure of the organ and stimulus-response measurements at the level of the whole organ. The later part of 20th century witnessed the first descriptions of the cellular responses and electrophysiology of isolated and cultured type I and type II cells, and there now exist a number of testable hypotheses of chemotransduction. The goal of this article is to provide a comprehensive review of current concepts on sensory transduction and transmission of the hypoxic stimulus at the carotid body with an emphasis on integrating cellular mechanisms with the whole organ responses and highlighting the gaps or discrepancies in our knowledge. It is increasingly evident that in addition to hypoxia, the carotid body responds to a wide variety of blood-borne stimuli, including reduced glucose and immune-related cytokines and we therefore also consider the evidence for a polymodal function of the carotid body and its implications. It is clear that the sensory function of the carotid body exhibits considerable plasticity in response to the chronic perturbations in environmental O2 that is associated with many physiological and pathological conditions. The mechanisms and consequences of carotid body plasticity in health and disease are discussed in the final sections of this article.
Collapse
Affiliation(s)
- Prem Kumar
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, The University of Birmingham, Birmingham, United Kingdom.
| | | |
Collapse
|
34
|
Tsuchiya N, Iwase M, Izumizaki M, Homma I. Dopaminergic modulation of exercise hyperpnoea via D2receptors in mice. Exp Physiol 2011; 97:228-38. [DOI: 10.1113/expphysiol.2011.062703] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
35
|
Priming of hypoxia-inducible factor by neuronal nitric oxide synthase is essential for adaptive responses to severe anemia. Proc Natl Acad Sci U S A 2011; 108:17544-9. [PMID: 21976486 DOI: 10.1073/pnas.1114026108] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cells sense and respond to changes in oxygen concentration through gene regulatory processes that are fundamental to survival. Surprisingly, little is known about how anemia affects hypoxia signaling. Because nitric oxide synthases (NOSs) figure prominently in the cellular responses to acute hypoxia, we defined the effects of NOS deficiency in acute anemia. In contrast to endothelial NOS or inducible NOS deficiency, neuronal NOS (nNOS)(-/-) mice demonstrated increased mortality during anemia. Unlike wild-type (WT) animals, anemia did not increase cardiac output (CO) or reduce systemic vascular resistance (SVR) in nNOS(-/-) mice. At the cellular level, anemia increased expression of HIF-1α protein and HIF-responsive mRNA levels (EPO, VEGF, GLUT1, PDK1) in the brain of WT, but not nNOS(-/-) mice, despite comparable reductions in tissue PO(2). Paradoxically, nNOS(-/-) mice survived longer during hypoxia, retained the ability to regulate CO and SVR, and increased brain HIF-α protein levels and HIF-responsive mRNA transcripts. Real-time imaging of transgenic animals expressing a reporter HIF-α(ODD)-luciferase chimeric protein confirmed that nNOS was essential for anemia-mediated increases in HIF-α protein stability in vivo. S-nitrosylation effects the functional interaction between HIF and pVHL. We found that anemia led to nNOS-dependent S-nitrosylation of pVHL in vivo and, of interest, led to decreased expression of GSNO reductase. These findings identify nNOS effects on the HIF/pVHL signaling pathway as critically important in the physiological responses to anemia in vivo and provide essential mechanistic insight into the differences between anemia and hypoxia.
Collapse
|
36
|
Chai S, Gillombardo CB, Donovan L, Strohl KP. Morphological differences of the carotid body among C57/BL6 (B6), A/J, and CSS B6A1 mouse strains. Respir Physiol Neurobiol 2011; 177:265-72. [PMID: 21555000 DOI: 10.1016/j.resp.2011.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 04/20/2011] [Accepted: 04/21/2011] [Indexed: 11/26/2022]
Abstract
The C57/BL6 (B6) mouse strain exhibits post-hypoxic frequency decline and periodic breathing, as well as greater amount of irregular breathing during rest in comparison to the A/J and to the B6a1, a chromosomal substitution strain whereby the A/J chromosome 1 is bred onto the B6 background (Han et al., 2002; Yamauchi et al., 2008a,b). The hypothesis was that morphological differences in the carotid body would associate with such trait variations. After confirming strain differences in post-hypoxic ventilatory behavior, histological examination (n=8 in each group) using hematoxylin and eosin (H&E) staining revealed equivalent, well-defined tissue structure at the bifurcation of the carotid arteries, an active secretory parenchyma (type I cells) from the supportive stromal tissue, and clustering of type I cells in all three strains. Tyrosine hydroxylase (TH) immunohistochemical staining revealed a typical organization of type I cells and neurovascular components into glomeruli in all three strains. Image analysis from 5 μm sections from each strain generated a series of cytological metrics. The percent carotid body composition of TH+ type I cells in the A/J, B6 and B6a1 was 20±4%, 39±3%, and 44±3%, respectively (p=0.00004). However, cellular organization in terms of density and ultrastructure in the B6a1 is more similar to the B6 than to the A/J. These findings indicate that genetic mechanisms that produce strain differences in ventilatory function do not associate with carotid body structure or tyrosine hydroxylase morphology, and that A/J chromosome 1 does not contribute much to B6 carotid body morphology.
Collapse
Affiliation(s)
- Sam Chai
- Department of Pulmonary, Critical Care, and Sleep Medicine, Case Western Reserve University, Cleveland, OH 44106, United States
| | | | | | | |
Collapse
|
37
|
Schwenke DO, Pearson JT, Kangawa K, Cragg PA, Shirai M. Exogenous ghrelin accentuates the acute hypoxic ventilatory response after two weeks of chronic hypoxia in conscious rats. Acta Physiol (Oxf) 2010; 200:279-87. [PMID: 20426772 DOI: 10.1111/j.1748-1716.2010.02142.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
AIM Ghrelin has been implicated as a modulator of numerous physiological pathways. To date, there have not been any studies describing the role of ghrelin in modulating the chemoreflex control of pulmonary ventilation. Yet the respiratory system impacts, at least to some degree, on virtually all homeostatic control systems. Chronic hypoxia (CH) can cause fundamental changes in ventilatory control, evident by alterations in the acute hypoxia ventilatory response (HVR). As ghrelin plays an important role in metabolic homeostasis, which is tightly linked to ventilatory control, we hypothesized that ghrelin may modulate HVR, especially following CH. METHODS Whole body plethysmography was used to measure the HVR (8% O(2) for 10 min) in male Sprague-Dawley rats (body wt ∼180-220 g) before and after 14 days of CH (CH=10% O(2)). During CH, rats received daily subcutaneous injections of either saline (control; n=5) or ghrelin (150 μg kg(-1) day(-1); n=5). The HVR was measured in another four rats that had received daily injections of ghrelin during normoxia for 7 days. RESULTS Ghrelin did not significantly alter basal ventilatory drive or acute HVR in normoxic rats. However, the acute HVR was accentuated following CH in ghrelin-treated rats compared with saline-treated rats. CONCLUSIONS These results describe the impact that ghrelin has in altering ventilatory control following CH and, although the mechanisms remain to be fully elucidated, provide guidance for future ghrelin-based studies interpreting physiological data indirectly related to the chemoreflex control of pulmonary ventilation.
Collapse
Affiliation(s)
- D O Schwenke
- Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, New Zealand.
| | | | | | | | | |
Collapse
|
38
|
Teppema LJ, Dahan A. The Ventilatory Response to Hypoxia in Mammals: Mechanisms, Measurement, and Analysis. Physiol Rev 2010; 90:675-754. [DOI: 10.1152/physrev.00012.2009] [Citation(s) in RCA: 257] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The respiratory response to hypoxia in mammals develops from an inhibition of breathing movements in utero into a sustained increase in ventilation in the adult. This ventilatory response to hypoxia (HVR) in mammals is the subject of this review. The period immediately after birth contains a critical time window in which environmental factors can cause long-term changes in the structural and functional properties of the respiratory system, resulting in an altered HVR phenotype. Both neonatal chronic and chronic intermittent hypoxia, but also chronic hyperoxia, can induce such plastic changes, the nature of which depends on the time pattern and duration of the exposure (acute or chronic, episodic or not, etc.). At adult age, exposure to chronic hypoxic paradigms induces adjustments in the HVR that seem reversible when the respiratory system is fully matured. These changes are orchestrated by transcription factors of which hypoxia-inducible factor 1 has been identified as the master regulator. We discuss the mechanisms underlying the HVR and its adaptations to chronic changes in ambient oxygen concentration, with emphasis on the carotid bodies that contain oxygen sensors and initiate the response, and on the contribution of central neurotransmitters and brain stem regions. We also briefly summarize the techniques used in small animals and in humans to measure the HVR and discuss the specific difficulties encountered in its measurement and analysis.
Collapse
Affiliation(s)
- Luc J. Teppema
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Albert Dahan
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
39
|
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 2009; 168:125-32. [DOI: 10.1016/j.resp.2009.03.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 03/03/2009] [Accepted: 03/04/2009] [Indexed: 11/24/2022]
|
40
|
Moayeri M, Crown D, Dorward DW, Gardner D, Ward JM, Li Y, Cui X, Eichacker P, Leppla SH. The heart is an early target of anthrax lethal toxin in mice: a protective role for neuronal nitric oxide synthase (nNOS). PLoS Pathog 2009; 5:e1000456. [PMID: 19478875 PMCID: PMC2680977 DOI: 10.1371/journal.ppat.1000456] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 04/29/2009] [Indexed: 01/04/2023] Open
Abstract
Anthrax lethal toxin (LT) induces vascular insufficiency in experimental animals through unknown mechanisms. In this study, we show that neuronal nitric oxide synthase (nNOS) deficiency in mice causes strikingly increased sensitivity to LT, while deficiencies in the two other NOS enzymes (iNOS and eNOS) have no effect on LT-mediated mortality. The increased sensitivity of nNOS-/- mice was independent of macrophage sensitivity to toxin, or cytokine responses, and could be replicated in nNOS-sufficient wild-type (WT) mice through pharmacological inhibition of the enzyme with 7-nitroindazole. Histopathological analyses showed that LT induced architectural changes in heart morphology of nNOS-/- mice, with rapid appearance of novel inter-fiber spaces but no associated apoptosis of cardiomyocytes. LT-treated WT mice had no histopathology observed at the light microscopy level. Electron microscopic analyses of LT-treated mice, however, revealed striking pathological changes in the hearts of both nNOS-/- and WT mice, varying only in severity and timing. Endothelial/capillary necrosis and degeneration, inter-myocyte edema, myofilament and mitochondrial degeneration, and altered sarcoplasmic reticulum cisternae were observed in both LT-treated WT and nNOS-/- mice. Furthermore, multiple biomarkers of cardiac injury (myoglobin, cardiac troponin-I, and heart fatty acid binding protein) were elevated in LT-treated mice very rapidly (by 6 h after LT injection) and reached concentrations rarely reported in mice. Cardiac protective nitrite therapy and allopurinol therapy did not have beneficial effects in LT-treated mice. Surprisingly, the potent nitric oxide scavenger, carboxy-PTIO, showed some protective effect against LT. Echocardiography on LT-treated mice indicated an average reduction in ejection fraction following LT treatment in both nNOS-/- and WT mice, indicative of decreased contractile function in the heart. We report the heart as an early target of LT in mice and discuss a protective role for nNOS against LT-mediated cardiac damage.
Collapse
Affiliation(s)
- Mahtab Moayeri
- Bacterial Toxins and Therapeutics Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Devorah Crown
- Bacterial Toxins and Therapeutics Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - David W. Dorward
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Don Gardner
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America
| | - Jerrold M. Ward
- Infectious Diseases Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yan Li
- Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Xizhong Cui
- Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter Eichacker
- Critical Care Medicine Department, National Institutes of Health Clinical Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Stephen H. Leppla
- Bacterial Toxins and Therapeutics Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
41
|
Gallego J, Dauger S. PHOX2B mutations and ventilatory control. Respir Physiol Neurobiol 2009; 164:49-54. [PMID: 18675942 DOI: 10.1016/j.resp.2008.07.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2008] [Revised: 07/07/2008] [Accepted: 07/09/2008] [Indexed: 01/08/2023]
Abstract
The transcription factor PHOX2B is essential for the development of the autonomic nervous system. In humans, polyalanine expansion mutations in PHOX2B cause Congenital Central Hypoventilation Syndrome (CCHS), a rare life-threatening disorder characterized by hypoventilation during sleep and impaired chemosensitivity. CCHS is combined with comparatively less severe impairments of autonomic functions including thermoregulation, cardiac rhythm, and digestive motility. Respiratory phenotype analyses of mice carrying an invalidated Phox2b allele (Phox2b+/- mutant mice) or the Phox2b mutation (+7 alanine expansion) found in patients with CCHS (Phox2b(27Ala/+) mice) have shed light on the role for PHOX2B in breathing control and on the pathophysiological mechanisms underlying CCHS. Newborn mice that lacked one Phox2b allele (Phox2b+/-) had sleep apneas and depressed sensitivity to hypercapnia. However, these impairments resolved rapidly, whereas the CCHS phenotype is irreversible. Heterozygous Phox2b(27Ala/+) pups exhibited a lack of responsiveness to hypercapnia and unstable breathing; they died within the first few postnatal hours. The generation of mouse models of CCHS provides tools for evaluating treatments aimed at alleviating both the respiratory symptoms and all other autonomic symptoms of CCHS.
Collapse
Affiliation(s)
- Jorge Gallego
- INSERM, U676, Hôpital Robert Debré, 48 Bd Sérurier, 75019 Paris, France.
| | | |
Collapse
|
42
|
Soliz J, Thomsen JJ, Soulage C, Lundby C, Gassmann M. Sex-dependent regulation of hypoxic ventilation in mice and humans is mediated by erythropoietin. Am J Physiol Regul Integr Comp Physiol 2009; 296:R1837-46. [PMID: 19321698 DOI: 10.1152/ajpregu.90967.2008] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Acclimatization to hypoxic exposure relies on an elevated ventilation and erythropoietic activity. We recently proposed that erythropoietin (Epo) links both responses: apart from red blood cell production, cerebral and plasma Epo interact with the central and peripheral respiratory centers. Knowing that women cope better than men with reduced oxygen supply (as observed at high altitude), we analyzed the hypoxic ventilatory response in Epo-overexpressing transgenic male and female mice with high Epo levels in brain and plasma (Tg6) or in wild-type animals injected with recombinant human Epo (rhEpo). Exposure to moderate and severe hypoxia as well as to hyperoxia and injection of domperidone, a potent peripheral ventilatory stimulant, revealed that the presence of transgenic or rhEpo extensively increased the hypoxic ventilatory response in female mice compared with their corresponding male siblings. Alterations of catecholamines in the brain stem's respiratory centers were also sex dependent. In a proof-of-concept study, human volunteers were intravenously injected with 5,000 units rhEpo and subsequently exposed to 10% oxygen. Compared with men, the hypoxic ventilatory response was significantly increased in women. We conclude that Epo exerts a sex-dependent impact on hypoxic ventilation improving the response in female mice and in women that most probably involves sexual hormones. Our data provides an explanation as to why women are less susceptible to hypoxia-associated syndromes than men.
Collapse
Affiliation(s)
- Jorge Soliz
- nstitute of Veterinary Physiology, Vetsuisse Faculty, and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich CH-8057, Switzerland
| | | | | | | | | |
Collapse
|
43
|
Granjeiro ÉM, Machado BH. NO in the caudal NTS modulates the increase in respiratory frequency in response to chemoreflex activation in awake rats. Respir Physiol Neurobiol 2009; 166:32-40. [DOI: 10.1016/j.resp.2009.01.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2008] [Revised: 12/16/2008] [Accepted: 01/13/2009] [Indexed: 10/21/2022]
|
44
|
Reeves SR, Simakajornboon N, Gozal D. The role of nitric oxide in the neural control of breathing. Respir Physiol Neurobiol 2008; 164:143-50. [DOI: 10.1016/j.resp.2008.08.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Revised: 08/10/2008] [Accepted: 08/11/2008] [Indexed: 11/16/2022]
|
45
|
Soliz J, Soulage C, Borter E, van Patot MT, Gassmann M. Ventilatory responses to acute and chronic hypoxia are altered in female but not male Paskin-deficient mice. Am J Physiol Regul Integr Comp Physiol 2008; 295:R649-58. [DOI: 10.1152/ajpregu.00876.2007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Proteins harboring a Per-Arnt-Sim (PAS) domain are versatile and allow archaea, bacteria, and plants to sense oxygen partial pressure, as well as light intensity and redox potential. A PAS domain associated with a histidine kinase domain is found in FixL, the oxygen sensor molecule of Rhizobium species. PASKIN is the mammalian homolog of FixL, but its function is far from being understood. Using whole body plethysmography, we evaluated the ventilatory response to acute and chronic hypoxia of homozygous deficient male and female PASKIN mice ( Paskin −/−). Although only slight ventilatory differences were found in males, female Paskin −/− mice increased ventilatory response to acute hypoxia. Unexpectedly, females had an impaired ability to reach ventilatory acclimatization in response to chronic hypoxia. Central control of ventilation occurs in the brain stem respiratory centers and is modulated by catecholamines via tyrosine hydroxylase (TH) activity. We observed that TH activity was altered in male and female Paskin −/− mice. Peripheral chemoreceptor effects on ventilation were evaluated by exposing animals to hyperoxia (Dejours test) and domperidone, a peripheral ventilatory stimulant drug directly affecting the carotid sinus nerve discharge. Male and female Paskin −/− had normal peripheral chemosensory (carotid bodies) responses. In summary, our observations suggest that PASKIN is involved in the central control of hypoxic ventilation, modulating ventilation in a gender-dependent manner.
Collapse
|
46
|
Ding Y, Li YL, Schultz HD. Downregulation of carbon monoxide as well as nitric oxide contributes to peripheral chemoreflex hypersensitivity in heart failure rabbits. J Appl Physiol (1985) 2008; 105:14-23. [PMID: 18356479 DOI: 10.1152/japplphysiol.01345.2007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Peripheral chemoreflex sensitivity is potentiated in clinical and experimental chronic heart failure (CHF). Downregulation of nitric oxide (NO) synthase (NOS) in the carotid body (CB) is involved in this effect. However, it remains poorly understood whether carbon monoxide (CO) also contributes to the altered peripheral chemoreflex sensitivity in CHF. This work highlights the effect of NO and CO on renal sympathetic nerve activity (RSNA) in response to graded hypoxia in conscious rabbits. Renal sympathetic nerve responses to graded hypoxia were enhanced in CHF rabbits compared with sham rabbits. The NO donor S-nitroso-N-acetylpenicillamine (SNAP, 1.2 microg x kg(-1) x min(-1)) and the CO-releasing molecule tricarbonyldichlororuthenium (II) dimer {[Ru(CO)(3)Cl(2)](2), 3.0 microg x kg(-1) x min(-1)} each attenuated hypoxia-induced RSNA increases in CHF rabbits (P < 0.05), but the degree of attenuation of RSNA induced by SNAP or [Ru(CO)(3)Cl(2)](2) was smaller than that induced by SNAP + [Ru(CO)(3)Cl(2)](2). Conversely, treatment with the NOS inhibitor N(omega)-nitro-L-arginine (30 mg/kg) + the heme oxygenase (HO) inhibitor Cr (III) mesoporphyrin IX chloride (0.5 mg/kg) augmented the renal sympathetic nerve response to hypoxia in sham rabbits to a greater extent than treatment with either inhibitor alone and was without effect in CHF rabbits. In addition, using immunostaining and Western blot analyses, we found that expression of neuronal NOS, endothelial NOS, and HO-2 protein (expressed as the ratio of NOS or HO-2 expression to beta-tubulin protein expression) was lower in CBs from CHF (0.19 +/- 0.04, 0.17 +/- 0.06, and 0.15 +/- 0.02, respectively) than sham (0.63 +/- 0.04, 0.56 +/- 0.06, and 0.27 +/- 0.03, respectively) rabbits (P < 0.05). These results suggest that a deficiency of NO and CO in the CBs augments peripheral chemoreflex sensitivity to hypoxia in CHF.
Collapse
Affiliation(s)
- Yanfeng Ding
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | | | | |
Collapse
|
47
|
Nitric oxide and respiratory rhythm in mammals: a new modulator of phase transition? Biochem Soc Trans 2008; 35:1258-63. [PMID: 17956325 DOI: 10.1042/bst0351258] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
NO (nitric oxide) modulates several central pattern generators, but its role in respiratory rhythmogenesis and its mode of action on medullary respiratory neurons during normoxia are unknown. We analysed the actions of NO on the mammalian respiratory network at the system and cellular levels. Given systemically, the NO donor diethylamine NONOate increased post-inspiratory duration in vagus, phrenic and hypoglossal nerves, whereas blockade of NO generation with L-NAME (N(G)-nitro-L-arginine methyl ester) produced the opposite response. At the cellular level, we pressure-ejected the NO donor on to respiratory neurons. NO had both inhibitory and excitatory effects on all types of respiratory neurons. Inhibitory effects involved soluble guanylate cyclase, as they were blocked with ODQ (1H-[1,2,4]oxadiazolo[4,3a]quinoxalin-1-one), whereas excitations were antagonized by uric acid and possibly mediated via peroxynitrite. Importantly, NO facilitated both GABA (gamma-aminobutyric acid)- and NMDA (N-methyl-D-aspartate)-induced neuronal responses, but this was restricted to post-inspiratory and pre-inspiratory neurons; other neuron types showed additive effects only. Our results support NO as modulator of centrally generated respiratory activity and specifically of ligand-mediated responses in respiratory neuron types involved in respiratory phase transition.
Collapse
|
48
|
Effect of Systemic Administration of the Nitric Oxide Synthase Inhibitor L-NMMA on the Human Ventilatory Response to Hypoxia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008. [DOI: 10.1007/978-0-387-73693-8_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
49
|
Abstract
The role of nitric oxide (NO) as a highly diffusible free radical gaseous vasodilator is intrinsically linked to the control of blood flow and oxygen (O(2)) delivery to tissue. NO also is involved in regulating mitochondrial O(2) metabolism, growth of new blood vessels, and blood oxygenation through control of respiratory ventilation. Hemoglobin and myoglobin may help to conserve NO for subsequent release of a NO-related vasoactive species under hypoxic conditions. NO has a major role in regulating microvascular O(2), and dysfunctional NO signaling is associated with the pathogenesis of metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Donald G Buerk
- Departments of Physiology and Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
50
|
Schultz HD, Li YL. Carotid body function in heart failure. Respir Physiol Neurobiol 2007; 157:171-85. [PMID: 17374517 PMCID: PMC1965591 DOI: 10.1016/j.resp.2007.02.011] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2007] [Revised: 02/07/2007] [Accepted: 02/08/2007] [Indexed: 11/16/2022]
Abstract
In this review, we summarize the present state of knowledge of the functional characteristics of the carotid body (CB) chemoreflex with respect to control of sympathetic nerve activity (SNA) in chronic heart failure (CHF). Evidence from both CHF patients and animal models of CHF has clearly established that the CB chemoreflex is enhanced in CHF and contributes to the tonic elevation in SNA. This adaptive change derives from altered function at the level of both the afferent and central nervous system (CNS) pathways of the reflex arc. At the level of the CB, an elevation in basal afferent discharge occurs under normoxic conditions in CHF rabbits, and the discharge responsiveness to hypoxia is enhanced. Outward voltage-gated K(+) currents (I(K)) are suppressed in CB glomus cells from CHF rabbits, and their sensitivity to hypoxic inhibition is enhanced. These changes in I(K) derive partly from downregulation of nitric oxide synthase (NOS)/NO signaling and upregulation of angiotensin II (Ang II)/Ang II receptor (AT(1)R) signaling in glomus cells. At the level of the CNS, interactions of the enhanced input from CB chemoreceptors with altered input from baroreceptor and cardiac afferent pathways and from central Ang II further enhance sympathetic drive. In addition, impaired function of NO in the paraventricular nucleus of the hypothalamus participates in the increased SNA response to CB chemoreceptor activation. These results underscore the principle that multiple mechanisms involving Ang II and NO at the level of both the CB and CNS represent complementary and perhaps redundant adaptive mechanisms to enhance CB chemoreflex function in CHF.
Collapse
Affiliation(s)
- Harold D Schultz
- Department of Cellular and Integrative Physiology, University of Nebraska College of Medicine, 985850 Nebraska Medical Center, Omaha, NE 68198-5850, USA.
| | | |
Collapse
|