1
|
Shin MK, Kim LJ, Davaanyam D, Amorim MR, Lee SM, Tang WY, Polotsky VY. Leptin receptor downregulation in the carotid body treats obesity-induced hypertension. J Neurophysiol 2025; 133:892-903. [PMID: 39903168 DOI: 10.1152/jn.00133.2024] [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: 04/01/2024] [Revised: 05/13/2024] [Accepted: 01/26/2025] [Indexed: 02/06/2025] Open
Abstract
Obesity and comorbid sleep disordered breathing (SDB) lead to high cardiovascular morbidity and mortality via multiple mechanisms including hypertension. Obesity also leads to high levels of leptin, which is produced in adipocytes. Increased leptin levels have also been implicated in increased sympathetic activity and the pathogenesis of hypertension in obesity. However, mechanisms for the effects of leptin on blood pressure are unclear. The carotid bodies (CB) express leptin receptor (Leprb), and diet-induced obesity (DIO) increases Leprb expression levels, but the mechanisms and consequences of leptin action in CB are poorly understood. We hypothesize that leptin signaling in CB in obesity leads to hypertension, which can be treated by Leprb knockdown specifically in CB. DIO male and female mice and lean male C57BL/6J mice were implanted with telemetry in the left femoral artery for continuous blood pressure monitoring. The adenoviral vectors carrying antisense RNA, Ad-LepR shRNA or Ad-scrambled control shRNA, were administered locally to the CB region. Blood pressure measurements were performed at baseline and 9-11 days after CB infection with the adenoviral vector. DIO male mice showed increased blood pressure compared with lean males and DIO females. Ad-LepR shRNA induced a twofold decrease in Leprb mRNA level in CB and abolished obesity-induced hypertension. Leprb knockdown was particularly effective during the light phase, when animals were predominantly asleep, decreasing mean arterial pressure by 8.5 mmHg. Control shRNA had no effect on DIO-induced hypertension. We conclude that inhibition of Leprb in the carotid bodies abolished obesity-induced hypertension.NEW & NOTEWORTHY Obesity and comorbid sleep apnea are key predisposing factors to hypertension. Obesity increases circulating leptin levels and hyperleptinemia may contribute to hypertension but mechanisms are not clear. Here, we have shown that knockdown of the leptin receptor LepRb in the carotid body decreased blood pressure and treated hypertension in diet-induced obese mice. Thus, we identified a novel mechanism of obesity hypertension and a novel drug target, LepRb in the carotid body.
Collapse
Affiliation(s)
- Mi-Kyung Shin
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Lenise J Kim
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Dashdulam Davaanyam
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Mateus R Amorim
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Sean M Lee
- Office of Clinical Research, George Washington University, Washington, District of Columbia, United States
| | - Wan-Yee Tang
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, United States
| | - Vsevolod Y Polotsky
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
- Department of Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
2
|
Leon-Mercado L, Menendez-Montes I, Tao J, Chen B, Olson DP, Mackaaij C, Cleypool CGJ, Gautron L. Hypoxia inducible factor-dependent upregulation of Agrp in glomus type I cells of the carotid body. Mol Metab 2025; 92:102095. [PMID: 39793758 PMCID: PMC11786784 DOI: 10.1016/j.molmet.2025.102095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Agouti-related peptide (AgRP) is a well-established potent orexigenic peptide primarily expressed in hypothalamic neurons. Nevertheless, the expression and functional significance of extrahypothalamic AgRP remain poorly understood. In this study, utilizing histological and molecular biology techniques, we have identified a significant expression of Agrp mRNA and AgRP peptide production in glomus type I cells within the mouse carotid body (CB). Furthermore, we have uncovered evidence supporting the expression of the AgRP receptor melanocortin receptor 3 (Mc3r) in adjacent sympathetic neurons, suggesting a potential local paracrine role for AgRP within the CB. Importantly, AgRP immunoreactivity was also identified in glomus type I cells of the human CB. Given the unexpected abundance of AgRP in glomus type I cells, a chemoreceptor cell specialized in oxygen sensing, we proceeded to investigate whether Agrp expression in the CB is regulated by hypoxemia and associated oxygen-sensing molecular mechanisms. In vitro luciferase assays reveal that hypoxia stimulates the human and mouse Agrp promoters in a Hypoxia Inducible Factor (HIF1/2)-dependent manner. Our in vivo experiments further demonstrate that exposure to environmental hypoxia (10%) robustly induces Agrp expression in type I glomus cells of mice. Furthermore, these findings collectively highlight the hitherto unknown source of AgRP in murine and human type I glomus cells and underscore the direct control of Agrp transcription by HIF signaling.
Collapse
Affiliation(s)
- Luis Leon-Mercado
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Ivan Menendez-Montes
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan Tao
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bandy Chen
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - David P Olson
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - C Mackaaij
- Division of Surgical Specialties, Department of Anatomy, University Medical Center, Utrecht, the Netherlands
| | - C G J Cleypool
- Division of Surgical Specialties, Department of Anatomy, University Medical Center, Utrecht, the Netherlands
| | - Laurent Gautron
- Center for Hypothalamic Research and Department of Internal medicine, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
3
|
Abstract
How oxygen is sensed by the heart and what mechanisms mediate its sensing remain poorly understood. Since recent reports show that low PO2 levels are detected by the cardiomyocytes in a few seconds, the rapid and short applications of low levels of oxygen (acute hypoxia), that avoid multiple effects of chronic hypoxia may be used to probe the oxygen sensing pathway of the heart. Here we explore the oxygen sensing pathway, focusing primarily on cellular surface membrane proteins that are first exposed to low PO2. Such studies suggest that acute hypoxia primarily targets the cardiac calcium channels, where either the channel itself or moieties closely associated with it, for instance, heme-oxygenase-2 (HO-2) interacting through kinase phosphorylation, signals the α-subunit of the channel as to the altered levels of PO2. Amino acids 1572-1651, the CaMKII phosphorylation sites (S1487 and S1545), CaM-binding site (I1624, Q1625) and Ser1928 of the carboxyl tail of the α-subunit appear to be critical residues that sense oxygen. Future studies in HO-2 knockout mice or CRISPR/Cas9 gene-edited hiPSC-CMs that reduce CaM-binding affinity are likely to provide deeper insights in the O2-sensinsing mechanisms.
Collapse
Affiliation(s)
| | - Martin Morad
- USC, MUSC, and Clemson University, Cardiac Signaling Center, Charleston, South Carolina, United States;
| |
Collapse
|
4
|
Stocco E, Sfriso MM, Borile G, Contran M, Barbon S, Romanato F, Macchi V, Guidolin D, De Caro R, Porzionato A. Experimental Evidence of A 2A-D 2 Receptor-Receptor Interactions in the Rat and Human Carotid Body. Front Physiol 2021; 12:645723. [PMID: 33935801 PMCID: PMC8082109 DOI: 10.3389/fphys.2021.645723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/09/2021] [Indexed: 12/26/2022] Open
Abstract
Adenosine A2A receptors (A2AR) and dopamine D2 receptors (D2R) are known to be involved in the physiological response to hypoxia, and their expression/activity may be modulated by chronic sustained or intermittent hypoxia. To date, A2AR and D2R can form transient physical receptor–receptor interactions (RRIs) giving rise to a dynamic equilibrium able to influence ligand binding and signaling, as demonstrated in different native tissues and transfected mammalian cell systems. Given the presence of A2AR and D2R in type I cells, type II cells, and afferent nerve terminals of the carotid body (CB), the aim of this work was to demonstrate here, for the first time, the existence of A2AR–D2R heterodimers by in situ proximity ligation assay (PLA). Our data by PLA analysis and tyrosine hydroxylase/S100 colocalization indicated the formation of A2AR–D2R heterodimers in type I and II cells of the CB; the presence of A2AR–D2R heterodimers also in afferent terminals is also suggested by PLA signal distribution. RRIs could play a role in CB dynamic modifications and plasticity in response to development/aging and environmental stimuli, including chronic intermittent/sustained hypoxia. Exploring other RRIs will allow for a broad comprehension of the regulative mechanisms these interactions preside over, with also possible clinical implications.
Collapse
Affiliation(s)
- Elena Stocco
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Maria Martina Sfriso
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Giulia Borile
- Department of Physics and Astronomy "G. Galilei," University of Padova, Padua, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Martina Contran
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Silvia Barbon
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Filippo Romanato
- Department of Physics and Astronomy "G. Galilei," University of Padova, Padua, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Veronica Macchi
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Diego Guidolin
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Raffaele De Caro
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Andrea Porzionato
- Department of Neuroscience, Institute of Human Anatomy, University of Padova, Padua, Italy
| |
Collapse
|
5
|
Pan W, Scott AL, Nurse CA, Jonz MG. Identification of oxygen-sensitive neuroepithelial cells through an endogenous reporter gene in larval and adult transgenic zebrafish. Cell Tissue Res 2021; 384:35-47. [PMID: 33404838 DOI: 10.1007/s00441-020-03307-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/17/2020] [Indexed: 11/28/2022]
Abstract
In teleost fish, specialized oxygen (O2) chemoreceptors, called neuroepithelial cells (NECs), are found in the gill epithelium in adults. During development, NECs are present in the skin before the formation of functional gills. NECs are known for retaining the monoamine neurotransmitter, serotonin (5-HT) and are conventionally identified through immunoreactivity with antibodies against 5-HT or synaptic vesicle protein (SV2). However, identification of NECs in live tissue and isolated cell preparations has been challenging due to the lack of a specific marker. The present study explored the use of the transgenic zebrafish, ETvmat2:GFP, which expresses green fluorescent protein (GFP) under the control of the vesicular monoamine transporter 2 (vmat2) regulatory element, to identify NECs. Using immunohistochemistry and confocal microscopy, we confirmed that the endogenous GFP in ETvmat2:GFP labelled serotonergic NECs in the skin of larvae and in the gills of adults. NECs of the gill filaments expressed a higher level of endogenous GFP compared with other cells. The endogenous GFP also labelled intrabranchial neurons of the gill filaments. Flow cytometric analysis demonstrated that filamental NECs could be distinguished from other dissociated gill cells based on high GFP expression alone. Acclimation to 2 weeks of severe hypoxia (PO2 = 35 mmHg) induced an increase in filamental NEC frequency, size and GFP gene expression. Here we present for the first time a transgenic tool that labels O2 chemoreceptors in an aquatic vertebrate and its use in high-throughput experimentation.
Collapse
Affiliation(s)
- Wen Pan
- Department of Biology, University of Ottawa, 30 Marie Curie Pvt., Ottawa, ON, K1N 6N5, Canada
| | - Angela L Scott
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Colin A Nurse
- Department of Biology, McMaster University, Hamilton, ON, L8S 4K1, Canada
| | - Michael G Jonz
- Department of Biology, University of Ottawa, 30 Marie Curie Pvt., Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
6
|
Baik AH, Jain IH. Turning the Oxygen Dial: Balancing the Highs and Lows. Trends Cell Biol 2020; 30:516-536. [PMID: 32386878 PMCID: PMC7391449 DOI: 10.1016/j.tcb.2020.04.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/02/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
Oxygen is both vital and toxic to life. Molecular oxygen is the most used substrate in the human body and is required for several hundred diverse biochemical reactions. The discovery of the PHD-HIF-pVHL system revolutionized our fundamental understanding of oxygen sensing and cellular adaptations to hypoxia. It deepened our knowledge of the biochemical underpinnings of numerous diseases, ranging from anemia to cancer. Cellular dysfunction and tissue pathology can result from a mismatch of oxygen supply and demand. Recent work has shown that mitochondrial disease models display tissue hyperoxia and that disease pathology can be reversed by normalization of excess oxygen, suggesting that certain disease states can potentially be treated by modulating oxygen levels. In this review, we describe cellular and organismal mechanisms of oxygen sensing and adaptation. We provide a revitalized framework for understanding pathologies of too little or too much oxygen.
Collapse
Affiliation(s)
- Alan H Baik
- Department of Physiology, University of California, San Francisco, CA 94158, USA; Department of Medicine, Division of Cardiology, University of California, San Francisco, CA 94143, USA.
| | - Isha H Jain
- Department of Physiology, University of California, San Francisco, CA 94158, USA.
| |
Collapse
|
7
|
Bittencourt‐Silva PG, Menezes MF, Mendonça‐Junior BA, Karlen‐Amarante M, Zoccal DB. Postnatal intermittent hypoxia enhances phrenic and reduces vagal upper airway motor activities in rats by epigenetic mechanisms. Exp Physiol 2019; 105:148-159. [DOI: 10.1113/ep087928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/08/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Paloma G. Bittencourt‐Silva
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Miguel Furtado Menezes
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Bolival A. Mendonça‐Junior
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Marlusa Karlen‐Amarante
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| | - Daniel B. Zoccal
- Department of Physiology and Pathology School of Dentistry of Araraquara São Paulo State University (UNESP) Araraquara Brazil
| |
Collapse
|
8
|
Caballero-Eraso C, Shin MK, Pho H, Kim LJ, Pichard LE, Wu ZJ, Gu C, Berger S, Pham L, Yeung HYB, Shirahata M, Schwartz AR, Tang WYW, Sham JSK, Polotsky VY. Leptin acts in the carotid bodies to increase minute ventilation during wakefulness and sleep and augment the hypoxic ventilatory response. J Physiol 2018; 597:151-172. [PMID: 30285278 DOI: 10.1113/jp276900] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/03/2018] [Indexed: 01/10/2023] Open
Abstract
KEY POINTS Leptin is a potent respiratory stimulant. A long functional isoform of leptin receptor, LepRb , was detected in the carotid body (CB), a key peripheral hypoxia sensor. However, the effect of leptin on minute ventilation (VE ) and the hypoxic ventilatory response (HVR) has not been sufficiently studied. We report that LepRb is present in approximately 74% of the CB glomus cells. Leptin increased carotid sinus nerve activity at baseline and in response to hypoxia in vivo. Subcutaneous infusion of leptin increased VE and HVR in C57BL/6J mice and this effect was abolished by CB denervation. Expression of LepRb in the carotid bodies of LepRb deficient obese db/db mice increased VE during wakefulness and sleep and augmented the HVR. We conclude that leptin acts on LepRb in the CBs to stimulate breathing and HVR, which may protect against sleep disordered breathing in obesity. ABSTRACT Leptin is a potent respiratory stimulant. The carotid bodies (CB) express the long functional isoform of leptin receptor, LepRb , but the role of leptin in CB has not been fully elucidated. The objectives of the current study were (1) to examine the effect of subcutaneous leptin infusion on minute ventilation (VE ) and the hypoxic ventilatory response to 10% O2 (HVR) in C57BL/6J mice before and after CB denervation; (2) to express LepRb in CB of LepRb -deficient obese db/db mice and examine its effects on breathing during sleep and wakefulness and on HVR. We found that leptin enhanced carotid sinus nerve activity at baseline and in response to 10% O2 in vivo. In C57BL/6J mice, leptin increased VE from 1.1 to 1.5 mL/min/g during normoxia (P < 0.01) and from 3.6 to 4.7 mL/min/g during hypoxia (P < 0.001), augmenting HVR from 0.23 to 0.31 mL/min/g/Δ F I O 2 (P < 0.001). The effects of leptin on VE and HVR were abolished by CB denervation. In db/db mice, LepRb expression in CB increased VE from 1.1 to 1.3 mL/min/g during normoxia (P < 0.05) and from 2.8 to 3.2 mL/min/g during hypoxia (P < 0.02), increasing HVR. Compared to control db/db mice, LepRb transfected mice showed significantly higher VE throughout non-rapid eye movement (20.1 vs. -27.7 mL/min respectively, P < 0.05) and rapid eye movement sleep (16.5 vs 23.4 mL/min, P < 0.05). We conclude that leptin acts in CB to augment VE and HVR, which may protect against sleep disordered breathing in obesity.
Collapse
Affiliation(s)
- Candela Caballero-Eraso
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Unidad Médico-Quirúrgica de Enfermedades Respiratorias, Instituto de Biomedicina de Sevilla (IBiS), Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Hospital Universitario Virgen del Rocío/Universidad de Sevilla, Sevilla, Spain
| | - Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huy Pho
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lenise J Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Luis E Pichard
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhi-Juan Wu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chenjuan Gu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Slava Berger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luu Pham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ho-Yee Bonnie Yeung
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Machiko Shirahata
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Alan R Schwartz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wan-Yee Winnie Tang
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - James S K Sham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Hernansanz-Agustín P, Ramos E, Navarro E, Parada E, Sánchez-López N, Peláez-Aguado L, Cabrera-García JD, Tello D, Buendia I, Marina A, Egea J, López MG, Bogdanova A, Martínez-Ruiz A. Mitochondrial complex I deactivation is related to superoxide production in acute hypoxia. Redox Biol 2017; 12:1040-1051. [PMID: 28511347 PMCID: PMC5430576 DOI: 10.1016/j.redox.2017.04.025] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/28/2017] [Accepted: 04/18/2017] [Indexed: 01/10/2023] Open
Abstract
Mitochondria use oxygen as the final acceptor of the respiratory chain, but its incomplete reduction can also produce reactive oxygen species (ROS), especially superoxide. Acute hypoxia produces a superoxide burst in different cell types, but the triggering mechanism is still unknown. Herein, we show that complex I is involved in this superoxide burst under acute hypoxia in endothelial cells. We have also studied the possible mechanisms by which complex I could be involved in this burst, discarding reverse electron transport in complex I and the implication of PTEN-induced putative kinase 1 (PINK1). We show that complex I transition from the active to ‘deactive’ form is enhanced by acute hypoxia in endothelial cells and brain tissue, and we suggest that it can trigger ROS production through its Na+/H+ antiporter activity. These results highlight the role of complex I as a key actor in redox signalling in acute hypoxia. Complex I is involved in the superoxide burst produced by cells in acute hypoxia. Complex I is deactivated in acute hypoxia. Deactive complex I is involved in superoxide production in acute hypoxia, probably through its Na+/H+ antiporter activity. Complex I deactivation occurs in brain tissue hypoxia ex vivo and in vivo.
Collapse
Affiliation(s)
- Pablo Hernansanz-Agustín
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28006 Madrid, Spain; Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas Alberto Sols, E-28029 Madrid, Spain
| | - Elena Ramos
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28006 Madrid, Spain
| | - Elisa Navarro
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28029 Madrid, Spain
| | - Esther Parada
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28029 Madrid, Spain
| | - Nuria Sánchez-López
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28006 Madrid, Spain; Servicio de Proteómica, Centro de Biología Molecular "Severo Ochoa (CBSMO), Consejo Superior de Investigaciones Científicas (CSIC) - UAM, E-28049 Madrid, Spain
| | - Laura Peláez-Aguado
- Servicio de Proteómica, Centro de Biología Molecular "Severo Ochoa (CBSMO), Consejo Superior de Investigaciones Científicas (CSIC) - UAM, E-28049 Madrid, Spain
| | - J Daniel Cabrera-García
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28006 Madrid, Spain
| | - Daniel Tello
- Unidad de Investigación, Hospital Santa Cristina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IP), E-28009 Madrid, Spain
| | - Izaskun Buendia
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28029 Madrid, Spain
| | - Anabel Marina
- Servicio de Proteómica, Centro de Biología Molecular "Severo Ochoa (CBSMO), Consejo Superior de Investigaciones Científicas (CSIC) - UAM, E-28049 Madrid, Spain
| | - Javier Egea
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28029 Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28029 Madrid, Spain
| | - Anna Bogdanova
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, CH-8057 Zurich, Switzerland
| | - Antonio Martínez-Ruiz
- Servicio de Inmunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), E-28006 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.
| |
Collapse
|
10
|
Wang J, Hogan JO, Kim D. Voltage- and receptor-mediated activation of a non-selective cation channel in rat carotid body glomus cells. Respir Physiol Neurobiol 2016; 237:13-21. [PMID: 28013061 DOI: 10.1016/j.resp.2016.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/16/2016] [Accepted: 12/08/2016] [Indexed: 10/20/2022]
Abstract
A recent study showed that hypoxia activates a Ca2+-sensitive, Na+-permeable non-selective cation channel (NSC) in carotid body glomus cells. We studied the effects of mitochondrial inhibitors that increase Ca2+ influx via Ca2+ channel (Cav), and receptor agonists that release Ca2+ from endoplasmic reticulum (ER) on NSC. Mitochondrial inhibitors (NaCN, FCCP, H2S, NO) elevated [Ca2+]i and activated NSC. Angiotensin II and acetylcholine that elevate [Ca2+]i via the Gq-IP3 pathway activated NSC. However, endothelin-1 (Gq) and 5-HT (Gq) showed little or no effect on [Ca2+]i and did not activate NSC. Adenosine (Gs) caused a weak rise in [Ca2+]i but did not activate NSC. Dopamine (Gs) and γ-aminobytyric acid (Gi) were ineffective in raising [Ca2+]i and failed to activate NSC. Store-operated Ca2+ entry (SOCE) produced by depletion of Ca2+ stores with cyclopiazonic acid activated NSC. Our results show that Ca2+ entry via Cav, ER Ca2+ release and SOCE can activate NSC. Thus, NSC contributes to both voltage- and receptor-mediated excitation of glomus cells.
Collapse
Affiliation(s)
- Jiaju Wang
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - James O Hogan
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Donghee Kim
- Department of Physiology and Biophysics, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.
| |
Collapse
|
11
|
Abstract
From birth, animals should possess functional machinery to appropriately regulate its respiration. This machinery has to detect the available oxygen quantity in order to efficiently modulate breathing movements in accordance with body requirements. The chemosensitivity process responsible for this detection is known to be mainly performed by carotid bodies. However, pulmonary neuroendocrine cells, which are mainly gathered in neuroepithelial bodies, also present the capability to exert chemosensitivity. The goal of this article is to put in perspective the potential complementarity in the activity of these two peripheral chemosensors in the context of neonatal oxygen chemosensitivity.
Collapse
Affiliation(s)
- Céline Caravagna
- Institut de Neurosciences de la Timone-Equipe IMAPATH, CERIMED, UMR 7289 CNRS & Aix-Marseille Université, 27 Boulevard Jean Moulin,13385, Marseille Cedex 05, France.
| | - Tommy Seaborn
- Faculté de Médecine, Université Laval, Pavillon Ferdinand-Vandry, Room 4645-A,1050, Avenue de la Médecine, Quebec, QC, G1V 0A6, Canada
| |
Collapse
|
12
|
Zhou T, Chien MS, Kaleem S, Matsunami H. Single cell transcriptome analysis of mouse carotid body glomus cells. J Physiol 2016; 594:4225-51. [PMID: 26940531 DOI: 10.1113/jp271936] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 02/24/2016] [Indexed: 01/12/2023] Open
Abstract
KEY POINTS Carotid body (CB) glomus cells mediate acute oxygen sensing and the initiation of the hypoxic ventilatory response, yet the gene expression profile of these cells is not available. We demonstrate that the single cell RNA-Seq method is a powerful tool for identifying highly expressed genes in CB glomus cells. Our single cell RNA-Seq results characterized novel CB glomus cell genes, including members of the G protein-coupled receptor signalling pathway, ion channels and atypical mitochondrial electron transport chain subunits. A heterologous cell-based screening identified acetate (which is known to affect CB glomus cell activity) as an agonist for the most highly abundant G protein-coupled receptor (Olfr78) in CB glomus cells. These data established the first transcriptome profile of CB glomus cells, highlighting genes with potential implications in CB chemosensory function. ABSTRACT The carotid body (CB) is a major arterial chemoreceptor containing glomus cells whose activities are regulated by changes in arterial blood content, including oxygen. Despite significant advancements in the characterization of their physiological properties, our understanding of the underlying molecular machinery and signalling pathway in CB glomus cells is still limited. To overcome this, we employed the single cell RNA-Seq method by performing next-generation sequencing on single glomus cell-derived cDNAs to eliminate contamination of genes derived from other cell types present in the CB. Using this method, we identified a set of genes abundantly expressed in glomus cells, which contained novel glomus cell-specific genes. Transcriptome and subsequent in situ hybridization and immunohistochemistry analyses identified abundant G protein-coupled receptor signalling pathway components and various types of ion channels, as well as members of the hypoxia-inducible factors pathway. A short-chain fatty acid olfactory receptor Olfr78, recently implicated in CB function, was the most abundant G protein-coupled receptor. Two atypical mitochondrial electron transport chain subunits (Ndufa4l2 and Cox4i2) were among the most specifically expressed genes in CB glomus cells, highlighting their potential roles in mitochondria-mediated oxygen sensing. The wealth of information provided by the present study offers a valuable foundation for identifying molecules functioning in the CB.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA
| | - Ming-Shan Chien
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA
| | - Safa Kaleem
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Duke University Medical Centre, Durham, NC, USA.,Department of Neurobiology and Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| |
Collapse
|
13
|
Jonz MG, Zachar PC, Da Fonte DF, Mierzwa AS. Peripheral chemoreceptors in fish: A brief history and a look ahead. Comp Biochem Physiol A Mol Integr Physiol 2015; 186:27-38. [DOI: 10.1016/j.cbpa.2014.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/03/2014] [Accepted: 09/03/2014] [Indexed: 11/24/2022]
|
14
|
Mori Y, Takahashi N, Polat OK, Kurokawa T, Takeda N, Inoue M. Redox-sensitive transient receptor potential channels in oxygen sensing and adaptation. Pflugers Arch 2015; 468:85-97. [PMID: 26149285 PMCID: PMC4700073 DOI: 10.1007/s00424-015-1716-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 06/22/2015] [Indexed: 12/14/2022]
Abstract
Regulation of ion channels is central to the mechanisms that underlie immediate acute physiological responses to changes in the availability of molecular oxygen (O2). A group of cation-permeable channels that are formed by transient receptor potential (TRP) proteins have been characterized as exquisite sensors of redox reactive species and as efficient actuators of electric/ionic signals in vivo. In this review, we first discuss how redox-sensitive TRP channels such as TRPA1 have recently emerged as sensors of the relatively inert oxidant O2. With regard to the physiological significance of O2 sensor TRP channels, vagal TRPA1 channels are mainly discussed with respect to their role in respiratory regulation in comparison with canonical pathways in glomus cells of the carotid body, which is a well-established O2-sensing organ. TRPM7 channels are discussed regarding hypoxia-sensing function in ischemic cell death. Also, ubiquitous expression of TRPA1 and TRPM7 together with their physiological relevance in the body is examined. Finally, based upon these studies on TRP channels, we propose a hypothesis of “O2 remodeling.” The hypothesis is that cells detect deviation of O2 availability from appropriate levels via sensors and adjust local O2 environments in vivo by controlling supply and consumption of O2 via pathways comprising cellular signals and transcription factors downstream of sensors, which consequently optimize physiological functions. This new insight into O2 adaptation through ion channels, particularly TRPs, may foster a paradigm shift in our understanding in the biological significance of O2.
Collapse
Affiliation(s)
- Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan.
- Laboratory of Environmental Systems Biology, Department of Technology and Ecology, Hall of Global Environmental Studies, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan.
| | - Nobuaki Takahashi
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Onur Kerem Polat
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Tatsuki Kurokawa
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto, 615-8510, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masahiro Inoue
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases, Higashinari-ku, Osaka, 537-8511, Japan
| |
Collapse
|
15
|
Jonz MG, Buck LT, Perry SF, Schwerte T, Zaccone G. Sensing and surviving hypoxia in vertebrates. Ann N Y Acad Sci 2015; 1365:43-58. [PMID: 25959851 DOI: 10.1111/nyas.12780] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/15/2022]
Abstract
Surviving hypoxia is one of the most critical challenges faced by vertebrates. Most species have adapted to changing levels of oxygen in their environment with specialized organs that sense hypoxia, while only few have been uniquely adapted to survive prolonged periods of anoxia. The goal of this review is to present the most recent research on oxygen sensing, adaptation to hypoxia, and mechanisms of anoxia tolerance in nonmammalian vertebrates. We discuss the respiratory structures in fish, including the skin, gills, and air-breathing organs, and recent evidence for chemosensory neuroepithelial cells (NECs) in these tissues that initiate reflex responses to hypoxia. The use of the zebrafish as a genetic and developmental model has allowed observation of the ontogenesis of respiratory and chemosensory systems, demonstration of a putative intracellular O2 sensor in chemoreceptors that may initiate transduction of the hypoxia signal, and investigation into the effects of extreme hypoxia on cardiorespiratory development. Other organisms, such as goldfish and freshwater turtles, display a high degree of anoxia tolerance, and these models are revealing important adaptations at the cellular level, such as the regulation of glutamatergic and GABAergic neurotransmission in defense of homeostasis in central neurons.
Collapse
Affiliation(s)
- Michael G Jonz
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Leslie T Buck
- Cell and Systems Biology, and Ecology and Evolutionary Biology, University of Toronto, Toronto, Ontario, Canada
| | - Steve F Perry
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | | | - Giacomo Zaccone
- Department of Environmental Sciences, Territorial, Food and Health Security (S.A.S.T.A.S.), University of Messina, Messina, Italy
| |
Collapse
|
16
|
Stuth EAE, Stucke AG, Zuperku EJ. Effects of anesthetics, sedatives, and opioids on ventilatory control. Compr Physiol 2013; 2:2281-367. [PMID: 23720250 DOI: 10.1002/cphy.c100061] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
This article provides a comprehensive, up to date summary of the effects of volatile, gaseous, and intravenous anesthetics and opioid agonists on ventilatory control. Emphasis is placed on data from human studies. Further mechanistic insights are provided by in vivo and in vitro data from other mammalian species. The focus is on the effects of clinically relevant agonist concentrations and studies using pharmacological, that is, supraclinical agonist concentrations are de-emphasized or excluded.
Collapse
Affiliation(s)
- Eckehard A E Stuth
- Medical College of Wisconsin, Anesthesia Research Service, Zablocki VA Medical Center, Milwaukee, Wisconsin, USA.
| | | | | |
Collapse
|
17
|
Abstract
The main functions of the respiratory neural network are to produce a coordinated, efficient, rhythmic motor behavior and maintain homeostatic control over blood oxygen and CO2/pH levels. Purinergic (ATP) signaling features prominently in these homeostatic reflexes. The signaling actions of ATP are produced through its binding to a diversity of ionotropic P2X and metabotropic P2Y receptors. However, its net effect on neuronal and network excitability is determined by the interaction between the three limbs of a complex system comprising the signaling actions of ATP at P2Rs, the distribution of multiple ectonucleotidases that differentially metabolize ATP into ADP, AMP, and adenosine (ADO), and the signaling actions of ATP metabolites, especially ADP at P2YRs and ADO at P1Rs. Understanding the significance of purinergic signaling is further complicated by the fact that neurons, glia, and the vasculature differentially express P2 and P1Rs, and that both neurons and glia release ATP. This article reviews at cellular, synaptic, and network levels, current understanding and emerging concepts about the diverse roles played by this three-part signaling system in: mediating the chemosensitivity of respiratory networks to hypoxia and CO2/pH; modulating the activity of rhythm generating networks and inspiratory motoneurons, and; controlling blood flow through the cerebral vasculature.
Collapse
Affiliation(s)
- Gregory D Funk
- Department of Physiology, Centre for Neuroscience, Women & Children's Health Research Institute (WCHRI), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
18
|
Huang X, Sun J, Rong W, Zhao T, Li DH, Ding X, Wu LY, Wu K, Schachner M, Xiao ZC, Zhu LL, Fan M. Loss of cell adhesion molecule CHL1 improves homeostatic adaptation and survival in hypoxic stress. Cell Death Dis 2013; 4:e768. [PMID: 23949217 PMCID: PMC3763446 DOI: 10.1038/cddis.2013.284] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 07/03/2013] [Accepted: 07/04/2013] [Indexed: 12/26/2022]
Abstract
Close homologue of L1 (CHL1) is a transmembrane cell adhesion molecule that is critical for brain development and for the maintenance of neural circuits in adults. Recent studies revealed that CHL1 has diverse roles and is involved in the regulation of recovery after spinal cord injury. CHL1 expression was downregulated in the cerebral cortex, hypothalamus, and brain stem after the induction of acute hypoxia (AH). In the current study, we sought to address the role of CHL1 in regulating homeostasis responses to hypoxia using CHL1-knockout (CHL1−/−) mice. We found that, compared with wild-type littermates, CHL1−/− mice showed a dramatically lower mortality rate and an augmented ventilatory response after they were subjected to AH. Immunofluorescence staining revealed that CHL1 was expressed in the carotid body (CB), the key oxygen sensor in rodents, and CHL1 expression level in the CB as assayed by western blot was decreased after hypoxic exposure. The number of glomus cells and the expression of tyrosine hydroxylase (a marker for glomus cells) in the CB of CHL1−/− mice appeared to be increased compared with CHL1+/+ mice. In addition, in the ex vivo CB preparation, hypoxia induced a significantly greater afferent nerve discharge in CHL1−/− mice compared with CHL1+/+ mice. Furthermore, the arterial blood pressure and plasma catecholamine levels of CHL1−/− mice were also significantly higher than those of CHL1+/+ mice. Our findings first demonstrate that CHL1 is a novel intrinsic factor that is involved in CB function and in the ventilatory response to AH.
Collapse
Affiliation(s)
- X Huang
- Department of Cognitive Sciences, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kim MJ, Ainsley JA, Carder JW, Johnson WA. Hyperoxia-triggered aversion behavior in Drosophila foraging larvae is mediated by sensory detection of hydrogen peroxide. J Neurogenet 2013; 27:151-62. [PMID: 23927496 DOI: 10.3109/01677063.2013.804920] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Reactive oxygen species (ROS) in excess have been implicated in numerous chronic illnesses, including asthma, diabetes, aging, cardiovascular disease, and neurodegenerative illness. However, at lower concentrations, ROS can also serve essential routine functions as part of cellular signal transduction pathways. As products of atmospheric oxygen, ROS-mediated signals can function to coordinate external environmental conditions with growth and development. A central challenge has been a mechanistic distinction between the toxic effects of oxidative stress and endogenous ROS functions occurring at much lower concentrations. Drosophila larval aerotactic behavioral assays revealed strong developmentally regulated aversion to mild hyperoxia mediated by H2O2-dependent activation of class IV multidendritic (mdIV) sensory neurons expressing the Degenerin/epithelial Na(+) channel subunit, Pickpocket1 (PPK1). Electrophysiological recordings in foraging-stage larvae (78-84 h after egg laying [AEL]) demonstrated PPK1-dependent activation of mdIV neurons by nanomolar levels of H2O2 well below levels normally associated with oxidative stress. Acute sensitivity was reduced > 100-fold during the larval developmental transition to wandering stage (> 96 h AEL), corresponding to a loss of hyperoxia aversion behavior during the same period. Degradation of endogenous H2O2 by transgenic overexpression of catalase in larval epidermis caused a suppression of hyperoxia aversion behavior. Conversely, disruption of endogenous catalase activity using a UAS-CatRNAi transposon resulted in an enhanced hyperoxia-aversive response. These results demonstrate an essential role for low-level endogenous H2O2 as an environment-derived signal coordinating developmental behavioral transitions.
Collapse
Affiliation(s)
- Myung Jun Kim
- Department of Molecular Physiology and Biophysics, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa , USA
| | | | | | | |
Collapse
|
20
|
EGL-13/SoxD specifies distinct O2 and CO2 sensory neuron fates in Caenorhabditis elegans. PLoS Genet 2013; 9:e1003511. [PMID: 23671427 PMCID: PMC3650002 DOI: 10.1371/journal.pgen.1003511] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Accepted: 04/03/2013] [Indexed: 11/19/2022] Open
Abstract
Animals harbor specialized neuronal systems that are used for sensing and coordinating responses to changes in oxygen (O2) and carbon dioxide (CO2). In Caenorhabditis elegans, the O2/CO2 sensory system comprises functionally and morphologically distinct sensory neurons that mediate rapid behavioral responses to exquisite changes in O2 or CO2 levels via different sensory receptors. How the diversification of the O2- and CO2-sensing neurons is established is poorly understood. We show here that the molecular identity of both the BAG (O2/CO2-sensing) and the URX (O2-sensing) neurons is controlled by the phylogenetically conserved SoxD transcription factor homolog EGL-13. egl-13 mutant animals fail to fully express the distinct terminal gene batteries of the BAG and URX neurons and, as such, are unable to mount behavioral responses to changes in O2 and CO2. We found that the expression of egl-13 is regulated in the BAG and URX neurons by two conserved transcription factors-ETS-5(Ets factor) in the BAG neurons and AHR-1(bHLH factor) in the URX neurons. In addition, we found that EGL-13 acts in partially parallel pathways with both ETS-5 and AHR-1 to direct BAG and URX neuronal fate respectively. Finally, we found that EGL-13 is sufficient to induce O2- and CO2-sensing cell fates in some cellular contexts. Thus, the same core regulatory factor, egl-13, is required and sufficient to specify the distinct fates of O2- and CO2-sensing neurons in C. elegans. These findings extend our understanding of mechanisms of neuronal diversification and the regulation of molecular factors that may be conserved in higher organisms.
Collapse
|
21
|
Palmer LA, May WJ, deRonde K, Brown-Steinke K, Gaston B, Lewis SJ. Hypoxia-induced ventilatory responses in conscious mice: gender differences in ventilatory roll-off and facilitation. Respir Physiol Neurobiol 2012. [PMID: 23183420 DOI: 10.1016/j.resp.2012.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The aim of this study was to compare the ventilatory responses of C57BL6 female and male mice during a 15 min exposure to a hypoxic-hypercapnic (H-H) or a hypoxic (10% O(2), 90% N(2)) challenge and subsequent return to room air. The ventilatory responses to H-H were similar in males and females whereas there were pronounced gender differences in the ventilatory responses during and following hypoxic challenge. In males, the hypoxic response included initial increases in minute volume via increases in tidal volume and frequency of breathing. These responses declined substantially (roll-off) during hypoxic exposure. Upon return to room-air, relatively sustained increases in these ventilatory parameters (short-term potentiation) were observed. In females, the initial responses to hypoxia were similar to those in males whereas roll-off was greater and post-hypoxia facilitation was smaller than in males. The marked differences in ventilatory roll-off and post-hypoxia facilitation between female and male C57BL6 mice provide evidence that gender is of vital importance to ventilatory control.
Collapse
Affiliation(s)
- Lisa A Palmer
- Pediatric Respiratory Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | | | | | | | | |
Collapse
|
22
|
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
|
23
|
Ca2+ homeostasis and exocytosis in carotid glomus cells: role of mitochondria. Cell Calcium 2011; 51:155-63. [PMID: 22209034 DOI: 10.1016/j.ceca.2011.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 11/07/2011] [Accepted: 12/04/2011] [Indexed: 12/30/2022]
Abstract
In oxygen sensing carotid glomus (type 1) cells, the hypoxia-triggered depolarization can be mimicked by mitochondrial inhibitors. We examined the possibility that, other than causing glomus cell depolarization, mitochondrial inhibition can regulate transmitter release via changes in Ca(2+) dynamics. Under whole-cell voltage clamp conditions, application of the mitochondrial inhibitors, carbonyl cyanide m-chlorophenylhydrazone (CCCP) or cyanide caused a dramatic slowing in the decay of the depolarization-triggered Ca(2+) signal in glomus cells. In contrast, inhibition of the Na(+)/Ca(2+) exchanger (NCX), plasma membrane Ca(2+)-ATPase (PMCA) pump or sarco-endoplasmic reticulum Ca(2+)-ATPase (SERCA) pump had much smaller effects. Consistent with the notion that mitochondrial Ca(2+) uptake is the dominant mechanism in cytosolic Ca(2+) removal, inhibition of the mitochondrial uniporter with ruthenium red slowed the decay of the depolarization-triggered Ca(2+) signal. Hypoxia also slowed cytosolic Ca(2+) removal, suggesting a partial impairment of mitochondrial Ca(2+) uptake. Using membrane capacitance measurement, we found that the increase in the duration of the depolarization-triggered Ca(2+) signal after mitochondrial inhibition was associated with an enhancement of the exocytotic response. The role of mitochondria in the regulation of Ca(2+) signal and transmitter release from glomus cells highlights the importance of mitochondria in hypoxic chemotransduction in the carotid bodies.
Collapse
|
24
|
Haouzi P, Bell H, Van de Louw A. Hypoxia-induced arterial chemoreceptor stimulation and hydrogen sulfide: too much or too little? Respir Physiol Neurobiol 2011; 179:97-102. [PMID: 22001444 DOI: 10.1016/j.resp.2011.09.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 09/16/2011] [Accepted: 09/19/2011] [Indexed: 11/28/2022]
Abstract
This brief review presents and discusses some of the important issues surrounding the theory which asserts that endogenous hydrogen sulfide (H(2)S) is the mediator of, or at least an important contributor to, hypoxia-induced arterial chemorereceptor stimulation. The view presented here is that before H(2)S can seriously be considered as a candidate for transducing the O(2)-signal in the carotid bodies (CB), fundamental contradictions need to be resolved. One of these major contradictions is certainly the discrepancy between the levels of H(2)S endogenously present in the CB during hypoxia compared to the levels required to stimulate the arterial chemoreceptors in vitro. Very small amounts of H(2)S are thought to be produced endogenously during a given level of hypoxia, yet the partial pressure of tissue H(2)S which is needed to produce an effect commensurate with that of hypoxia is thousands to millions of times higher. This review discusses this and other contradictions in light of what is known about H(2)S concentration and production in various tissues, the lessons we have learnt from the response to exogenous sulfide and the ability of the blood and the mitochondria to oxidize very large amounts of sulfide. These considerations suggest that the increased production of H(2)S in hypoxia and exogenous sulfide cannot produce the same effect on the carotid bodies and breathing. While the effects of the endogenous H(2)S on breathing remains to be established, the effects exogenous sulfide can be accounted for by its long established toxicity on cytochrome C oxidase.
Collapse
Affiliation(s)
- Philippe Haouzi
- Division of Pulmonary and Critical Care Medicine, Pennsylvania State University, College of Medicine, Hershey, PA 17033, United States.
| | | | | |
Collapse
|
25
|
Koos BJ. Adenosine A₂a receptors and O₂ sensing in development. Am J Physiol Regul Integr Comp Physiol 2011; 301:R601-22. [PMID: 21677265 DOI: 10.1152/ajpregu.00664.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reduced mitochondrial oxidative phosphorylation, via activation of adenylate kinase and the resulting exponential rise in the cellular AMP/ATP ratio, appears to be a critical factor underlying O₂ sensing in many chemoreceptive tissues in mammals. The elevated AMP/ATP ratio, in turn, activates key enzymes that are involved in physiologic adjustments that tend to balance ATP supply and demand. An example is the conversion of AMP to adenosine via 5'-nucleotidase and the resulting activation of adenosine A(₂A) receptors, which are involved in acute oxygen sensing by both carotid bodies and the brain. In fetal sheep, A(₂A) receptors associated with carotid bodies trigger hypoxic cardiovascular chemoreflexes, while central A(₂A) receptors mediate hypoxic inhibition of breathing and rapid eye movements. A(₂A) receptors are also involved in hypoxic regulation of fetal endocrine systems, metabolism, and vascular tone. In developing lambs, A(₂A) receptors play virtually no role in O₂ sensing by the carotid bodies, but brain A(₂A) receptors remain critically involved in the roll-off ventilatory response to hypoxia. In adult mammals, A(₂A) receptors have been implicated in O₂ sensing by carotid glomus cells, while central A(₂A) receptors likely blunt hypoxic hyperventilation. In conclusion, A(₂A) receptors are crucially involved in the transduction mechanisms of O₂ sensing in fetal carotid bodies and brains. Postnatally, central A(₂A) receptors remain key mediators of hypoxic respiratory depression, but they are less critical for O₂ sensing in carotid chemoreceptors, particularly in developing lambs.
Collapse
Affiliation(s)
- Brian J Koos
- Department of Obstetrics and Gynecology; Brain Research Institute, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
26
|
Abstract
Carbon dioxide (CO(2)) is a physiological gas found at low levels in the atmosphere and produced in cells during the process of aerobic respiration. Consequently, the levels of CO(2) within tissues are usually significantly higher than those found externally. Shifts in tissue levels of CO(2) (leading to either hypercapnia or hypocapnia) are associated with a number of pathophysiological conditions in humans and can occur naturally in niche habitats such as those of burrowing animals. Clinical studies have indicated that such altered CO(2) levels can impact upon disease progression. Recent advances in our understanding of the biology of CO(2) has shown that like other physiological gases such as molecular oxygen (O(2)) and nitric oxide (NO), CO(2) levels can be sensed by cells resulting in the initiation of physiological and pathophysiological responses. Acute CO(2) sensing in neurons and peripheral and central chemoreceptors is important in rapidly activated responses including olfactory signalling, taste sensation and cardiorespiratory control. Furthermore, a role for CO(2) in the regulation of gene transcription has recently been identified with exposure of cells and model organisms to high CO(2) leading to suppression of genes involved in the regulation of innate immunity and inflammation. This latter, transcriptional regulatory role for CO(2), has been largely attributed to altered activity of the NF-B family of transcription factors. Here, we review our evolving understanding of how CO(2) impacts upon gene transcription.
Collapse
Affiliation(s)
- Cormac T Taylor
- UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| | | |
Collapse
|
27
|
Ding Y, Li YL, Schultz HD. Role of blood flow in carotid body chemoreflex function in heart failure. J Physiol 2010; 589:245-58. [PMID: 21078591 DOI: 10.1113/jphysiol.2010.200584] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Peripheral chemoreflex sensitivity is potentiated in clinical and experimental chronic heart failure (CHF). Blood supply to tissues is inevitably reduced in CHF. However, it remains poorly understood whether the reduced blood flow is the cause of increased peripheral chemoreflex sensitivity in CHF. This work highlights the effect of chronically reduced blood flow to the carotid body (CB) on peripheral chemoreflex function in rabbits. In pacing-induced CHF rabbits, blood flow in the carotid artery was reduced by 36.4 ± 5.2% after 3 weeks of pacing. For comparison, a similar level of blood flow reduction was induced by carotid artery occlusion (CAO) over a similar 3 week time course without pacing. CB blood supply was reduced by similar levels in both CHF and CAO rabbits as measured with fluorescent microspheres. Compared with sham rabbits, CAO enhanced peripheral chemoreflex sensitivity in vivo, increased CB chemoreceptor activity in an isolated CB preparation and decreased outward potassium current (Ik) in CB glomus cells to levels similar to those that were observed in CHF rabbits. In CAO CB compared to sham, neural nitric oxide (NO) synthase (nNOS) expression and NO levels were suppressed, and angiotensin II (Ang II) type 1 receptor (AT1-R) protein expression and Ang II concentration were elevated; these changes were similar to those seen in the CB from CHF rabbits. A NO donor and AT1-R antagonist reversed CAO-enhanced chemoreflex sensitivity. These results suggest that a reduction of blood flow to the CB is involved in the augmentation of peripheral chemoreflex sensitivity in CHF.
Collapse
Affiliation(s)
- Yanfeng Ding
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE 68198-5850, USA
| | | | | |
Collapse
|
28
|
The role of the central chemoreceptors: a modeling perspective. Respir Physiol Neurobiol 2010; 173:230-43. [PMID: 20227528 DOI: 10.1016/j.resp.2010.03.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 03/06/2010] [Accepted: 03/08/2010] [Indexed: 11/22/2022]
Abstract
After introducing the respiratory control system, a previously developed model of the respiratory chemoreflexes, based on rebreathing test data, is briefly described. This model is used to gain insights into the respiratory chemoreflex characteristics of a selection of individuals, and so discover the role of their central chemoreceptors. The chemoreflex model characteristics for each individual were estimated by adjusting the model parameters so that its predictions fit their rebreathing test results. To gain a steady state description of the control of breathing at rest the chemoreflex model is combined with a model of the cerebrovascular reactivity and converted from P(CO)₂ to [H(+)] chemoreceptor inputs. This description is used to illustrate how acid-base and cerebrovascular reactivity factors affect the environment of the central chemoreceptors and determine their role in breathing control. Finally, a dynamic model incorporating the chemoreflex model, acid-base and cerebrovascular reactivity is used to show the role of the central chemoreceptors in stabilizing breathing during sleep at altitude.
Collapse
|