1
|
Xinliang Z, Achkasov EE, Gavrikov LK, Yuchen L, Zhang C, Dudnik EN, Rumyantseva O, Beeraka NM, Glazachev OS. Assessing the importance and safety of hypoxia conditioning for patients with occupational pulmonary diseases: A recent clinical perspective. Biomed Pharmacother 2024; 178:117275. [PMID: 39126774 DOI: 10.1016/j.biopha.2024.117275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024] Open
Abstract
Occupational pulmonary diseases (OPDs) pose a significant global health challenge, contributing to high mortality rates. This review delves into the pathophysiology of hypoxia and the safety of intermittent hypoxic conditioning (IHC) in OPD patients. By examining sources such as PubMed, Relemed, NLM, Scopus, and Google Scholar, the review evaluates the efficacy of IHC in clinical outcomes for OPD patients. It highlights the complexities of cardiovascular and respiratory regulation dysfunctions in OPDs, focusing on respiratory control abnormalities and the impact of intermittent hypoxic exposures. Key areas include the physiological effects of hypoxia, the role of hypoxia-inducible factor-1 alpha (HIF-1α) in occupational lung diseases, and the links between brain ischemia, stroke, and OPDs. The review also explores the interaction between intermittent hypoxic exposures, mitochondrial energetics, and lung physiology. The potential of IHE to improve clinical manifestations and underlying pathophysiology in OPD patients is thoroughly examined. This comprehensive analysis aims to benefit molecular pathologists, pulmonologists, clinicians, and physicians by enhancing understanding of IHE's clinical benefits, from research to patient care, and improving clinical outcomes for OPD patients.
Collapse
Affiliation(s)
- Zhang Xinliang
- Chair of Sports Medicine and Rehabilitation, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Co-Chair of Normal Physiology, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Eugeny E Achkasov
- Chair of Sports Medicine and Rehabilitation, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Leonid K Gavrikov
- Volgograd State Medical University, 1, Pavshikh Bortsov Sq., Volgograd 400131, Russia.
| | - Li Yuchen
- Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Chen Zhang
- Chair of Epidemiology and Modern Technologies of Vaccination, Institute of Professional Education, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia
| | - Elena N Dudnik
- Co-Chair of Normal Physiology, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| | - Olga Rumyantseva
- Izmerov Research Institute of Occupational Health, 31 Budeynniy Avenye, Moscow 105275, Russia.
| | - Narasimha M Beeraka
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN 46202, USA; Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia; Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Chiyyedu, Anantapuramu, Andhra Pradesh 515721, India.
| | - Oleg S Glazachev
- Co-Chair of Normal Physiology, Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya Str., Moscow 119991, Russia.
| |
Collapse
|
2
|
Zhang N, Wei F, Ning S, Hu J, Shi H, Yao Z, Tang M, Zhang Y, Gong J, Ge J, Cui Z. PPARγ Agonist Rosiglitazone and Antagonist GW9662: Antihypertensive Effects on Chronic Intermittent Hypoxia-Induced Hypertension in Rats. J Cardiovasc Transl Res 2024; 17:803-815. [PMID: 38411834 DOI: 10.1007/s12265-024-10499-6] [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: 12/29/2023] [Accepted: 02/08/2024] [Indexed: 02/28/2024]
Abstract
The increased incidence of hypertension associated with obstructive sleep apnea (OSA) presents significant physical, psychological, and economic challenges. Peroxisome proliferator-activated receptor gamma (PPARγ) plays a role in both OSA and hypertension, yet the therapeutic potential of PPARγ agonists and antagonists for OSA-related hypertension remains unexplored. Therefore, we constructed a chronic intermittent hypoxia (CIH)-induced hypertension rat model that mimics the pathogenesis of OSA-related hypertension in humans. The model involved administering PPARγ agonist rosiglitazone (RSG), PPARγ antagonist GW9662, or normal saline, followed by regular monitoring of blood pressure and thoracic aorta analysis using staining and electron microscopy. Intriguingly, our results indicated that both RSG and GW9662 appeared to potently counteract CIH-induced hypertension. In silico study suggested that GW9662's antihypertensive effect might mediated through angiotensin II receptor type 1 (AGTR1). Our findings provide insights into the mechanisms of OSA-related hypertension and propose novel therapeutic targets.
Collapse
MESH Headings
- Animals
- PPAR gamma/agonists
- PPAR gamma/metabolism
- Hypertension/physiopathology
- Hypertension/drug therapy
- Hypertension/metabolism
- Rosiglitazone/pharmacology
- Disease Models, Animal
- Antihypertensive Agents/pharmacology
- Antihypertensive Agents/therapeutic use
- Male
- Hypoxia/complications
- Hypoxia/drug therapy
- Anilides/pharmacology
- Rats, Sprague-Dawley
- Blood Pressure/drug effects
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/physiopathology
- Aorta, Thoracic/pathology
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/drug effects
- Chronic Disease
- Signal Transduction
- Sleep Apnea, Obstructive/drug therapy
- Sleep Apnea, Obstructive/physiopathology
- Sleep Apnea, Obstructive/complications
- Sleep Apnea, Obstructive/metabolism
- Molecular Docking Simulation
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Ningzhi Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Feng Wei
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Sisi Ning
- Department of Cardiology, Shanghai Changning Tianshan Traditional Chinese Medicine Hospital, Shanghai, China
| | - Jialu Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Hongtao Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Zhifeng Yao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Minna Tang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Yongqiao Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Jiaxin Gong
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
| | - Zhaoqiang Cui
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
| |
Collapse
|
3
|
DeConne TM, Fancher IS, Edwards DG, Trott DW, Martens CR. CD8 + T-cell metabolism is related to cerebrovascular reactivity in middle-aged adults. Am J Physiol Regul Integr Comp Physiol 2024; 326:R416-R426. [PMID: 38406845 DOI: 10.1152/ajpregu.00267.2023] [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: 11/30/2023] [Revised: 02/13/2024] [Accepted: 02/22/2024] [Indexed: 02/27/2024]
Abstract
Cerebrovascular reactivity (CVR) decreases with advancing age, contributing to increased risk of cognitive impairment; however, the mechanisms underlying the age-related decrease in CVR are incompletely understood. Age-related changes to T cells, such as impaired mitochondrial respiration, increased inflammation, likely contribute to peripheral and cerebrovascular dysfunction in animals. However, whether T-cell mitochondrial respiration is related to cerebrovascular function in humans is not known. Therefore, we hypothesized that peripheral T-cell mitochondrial respiration would be positively associated with CVR and that T-cell glycolytic metabolism would be negatively associated with CVR. Twenty middle-aged adults (58 ± 5 yr) were recruited for this study. T cells were separated from peripheral blood mononuclear cells. Cellular oxygen consumption rate (OCR) and extracellular acidification rate (ECAR, a marker of glycolytic activity) were measured using extracellular flux analysis. CVR was quantified using the breath-hold index (BHI), which reflects the change in blood velocity in the middle-cerebral artery (MCAv) during a 30-s breath-hold. In contrast to our hypothesis, we found that basal OCR in CD8+ T cells (β = -0.59, R2 = 0.27, P = 0.019) was negatively associated with BHI. However, in accordance with our hypothesis, we found that basal ECAR (β = -2.20, R2 = 0.29, P = 0.015) and maximum ECAR (β = -50, R2 = 0.24, P = 0.029) were negatively associated with BHI in CD8+ T cells. There were no associations observed in CD4+ T cells. These associations appeared to be primarily mediated by an association with the pressor response to the breath-hold test. Overall, our findings suggest that CD8+ T-cell respiration and glycolytic activity may influence CVR in humans.NEW & NOTEWORTHY Peripheral T-cell metabolism is related to in vivo cerebrovascular reactivity in humans. Higher glycolytic metabolism in CD8+ T cells was associated with lower cerebrovascular reactivity to a breath-hold in middle-aged adults, which is possibly reflective of a more proinflammatory state in midlife.
Collapse
Affiliation(s)
- Theodore M DeConne
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States
| | - Ibra S Fancher
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| | - David G Edwards
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| | - Daniel W Trott
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, United States
| | - Christopher R Martens
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware, United States
| |
Collapse
|
4
|
Kaur J, Manokaran L, Thynne M, Subhan MMF. The effect of breathing hypoxic gas (15% FIO 2 ) on physiological and behavioral outcomes during simulated driving in healthy subjects. Physiol Rep 2024; 12:e15963. [PMID: 38439737 PMCID: PMC10912923 DOI: 10.14814/phy2.15963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/10/2024] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Hypoxia is mainly caused by cardiopulmonary disease or high-altitude exposure. We used a driving simulator to investigate whether breathing hypoxic gas influences driving behaviors in healthy subjects. Fifty-two healthy subjects were recruited in this study, approved by the Science and Engineering Ethical Committee. During simulated driving experiments, driving behaviors, breathing frequency, oxygen saturation (SpO2 ), and heart rate variability (HRV) were analyzed. Each subject had four driving sessions; a 10-min practice and three 20-min randomized interventions: normoxic room air (21% FIO2 ) and medical air (21% FIO2 ) and hypoxic air (equal to 15% FIO2 ), analyzed by repeated measures ANOVA. Driving behaviors and HRV frequency domains showed no significant change. Heart rate (HR; p < 0.0001), standard deviation of the RR interval (SDRR; p = 0.03), short-term HRV (SD1; p < 0.0001), breathing rate (p = 0.01), and SpO2 (p < 0.0001) were all significantly different over the three gas interventions. Pairwise comparisons showed HR increased during hypoxic gas exposure compared to both normoxic interventions, while SDRR, SD1, breathing rate, and SpO2 were lower. Breathing hypoxic gas (15% FiO2 , equivalent to 2710 m altitude) may not have a significant impact on driving behavior in healthy subjects. Furthermore, HRV was negatively affected by hypoxic gas exposure while driving suggesting further research to investigate the impact of breathing hypoxic gas on driving performance for patients with autonomic dysfunction.
Collapse
Affiliation(s)
- Jaspreet Kaur
- School of Biomedical Sciences, Faculty of HealthUniversity of PlymouthPlymouthUK
| | - Lebbathana Manokaran
- School of Biomedical Sciences, Faculty of HealthUniversity of PlymouthPlymouthUK
| | - Michael Thynne
- Chest ClinicUniversity Hospitals Plymouth NHS TrustPlymouthUK
| | - Mirza M. F. Subhan
- School of Biomedical Sciences, Faculty of HealthUniversity of PlymouthPlymouthUK
| |
Collapse
|
5
|
Wang N, Su X, Sams D, Prabhakar NR, Nanduri J. P300/CBP Regulates HIF-1-Dependent Sympathetic Activation and Hypertension by Intermittent Hypoxia. Am J Respir Cell Mol Biol 2024; 70:110-118. [PMID: 37874694 PMCID: PMC10848695 DOI: 10.1165/rcmb.2022-0481oc] [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: 12/15/2022] [Accepted: 10/23/2023] [Indexed: 10/26/2023] Open
Abstract
Obstructive sleep apnea (OSA), a widespread breathing disorder, leads to intermittent hypoxia (IH). Patients with OSA and IH-treated rodents exhibit heightened sympathetic nerve activity and hypertension. Previous studies reported transcriptional activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox) by HIF-1 (hypoxia-inducible factor-1) contribute to autonomic dysfunction in IH-treated rodents. Lysine acetylation, regulated by KATs (lysine acetyltransferases) and KDACs (lysine deacetylases), activates gene transcription and plays an important role in several physiological and pathological processes. This study tested the hypothesis that acetylation of HIF-1α by p300/CBP (CREB-binding protein) (KAT) activates Nox transcription, leading to sympathetic activation and hypertension. Experiments were performed on pheochromocytoma-12 cells and rats treated with IH. IH increased KAT activity, p300/CBP protein, HIF-1α lysine acetylation, HIF-1 transcription, and HIF-1 binding to the Nox4 gene promoter in pheochromocytoma-12 cells, and these responses were blocked by CTK7A, a selective p300/CBP inhibitor. Plasma norepinephrine (index of sympathetic activation) and blood pressures were elevated in IH-treated rats. These responses were associated with elevated p300/CBP protein, HIF-1α stabilization, transcriptional activation of Nox2 and Nox4 genes, and reactive oxygen species, and all these responses were absent in CTK7A-treated IH rats. These findings suggest lysine acetylation of HIF-1α by p300/CBP is an important contributor to sympathetic excitation and hypertension by IH.
Collapse
Affiliation(s)
- Ning Wang
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| | - Xiaoyu Su
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| | - David Sams
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| | - Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| | - Jayasri Nanduri
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, Illinois
| |
Collapse
|
6
|
Iturriaga R. Carotid body contribution to the physio-pathological consequences of intermittent hypoxia: role of nitro-oxidative stress and inflammation. J Physiol 2023; 601:5495-5507. [PMID: 37119020 DOI: 10.1113/jp284112] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
Obstructive sleep apnoea (OSA), characterized by chronic intermittent hypoxia (CIH), is considered to be an independent risk for hypertension. The pathological cardiorespiratory consequences of OSA have been attributed to systemic oxidative stress, inflammation and sympathetic overflow induced by CIH, but an emerging body of evidence indicates that a nitro-oxidative and pro-inflammatory milieu within the carotid body (CB) is involved in the potentiation of CB chemosensory responses to hypoxia, which contribute to enhance the sympathetic activity. Accordingly, autonomic and cardiovascular alterations induced by CIH are critically dependent on an abnormally heightened CB chemosensory input to the nucleus of tractus solitarius (NTS), where second-order neurons project onto the rostral ventrolateral medulla (RVLM), activating pre-sympathetic neurons that control pre-ganglionic sympathetic neurons. CIH produces oxidative stress and neuroinflammation in the NTS and RVLM, which may contribute to the long-term irreversibility of the CIH-induced alterations. This brief review is mainly focused on the contribution of nitro-oxidative stress and pro-inflammatory molecules on the hyperactivation of the hypoxic chemoreflex pathway including the CB and the brainstem centres, and whether the persistence of autonomic and cardiorespiratory alterations may depend on the glial-related neuroinflammation induced by the enhanced CB chemosensory afferent input.
Collapse
Affiliation(s)
- Rodrigo Iturriaga
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Investigación en Fisiología y Medicina de Altura, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| |
Collapse
|
7
|
Cuéllar-Pérez R, Jauregui-Huerta F, Ruvalcaba-Delgadillo Y, Montero S, Lemus M, Roces de Álvarez-Buylla E, García-Estrada J, Luquín S. K252a Prevents Microglial Activation Induced by Anoxic Stimulation of Carotid Bodies in Rats. TOXICS 2023; 11:871. [PMID: 37888721 PMCID: PMC10610815 DOI: 10.3390/toxics11100871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/13/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023]
Abstract
Inducing carotid body anoxia through the administration of cyanide can result in oxygen deprivation. The lack of oxygen activates cellular responses in specific regions of the central nervous system, including the Nucleus Tractus Solitarius, hypothalamus, hippocampus, and amygdala, which are regulated by afferent pathways from chemosensitive receptors. These receptors are modulated by the brain-derived neurotrophic factor receptor TrkB. Oxygen deprivation can cause neuroinflammation in the brain regions that are activated by the afferent pathways from the chemosensitive carotid body. To investigate how microglia, a type of immune cell in the brain, respond to an anoxic environment resulting from the administration of NaCN, we studied the effects of blocking the TrkB receptor on this cell-type response. Male Wistar rats were anesthetized, and a dose of NaCN was injected into their carotid sinus to induce anoxia. Prior to the anoxic stimulus, the rats were given an intracerebroventricular (icv) infusion of either K252a, a TrkB receptor inhibitor, BDNF, or an artificial cerebrospinal fluid (aCSF). After the anoxic stimulus, the rats were perfused with paraformaldehyde, and their brains were processed for microglia immunohistochemistry. The results indicated that the anoxic stimulation caused an increase in the number of reactive microglial cells in the hypothalamic arcuate, basolateral amygdala, and dentate gyrus of the hippocampus. However, the infusion of the K252a TrkB receptor inhibitor prevented microglial activation in these regions.
Collapse
Affiliation(s)
- Ricardo Cuéllar-Pérez
- Microscopía de Alta Resolución, Depto, de Neurociencias, Universidad de Guadalajara, Guadalajara 44340, Mexico; (R.C.-P.)
| | - Fernando Jauregui-Huerta
- Microscopía de Alta Resolución, Depto, de Neurociencias, Universidad de Guadalajara, Guadalajara 44340, Mexico; (R.C.-P.)
| | - Yaveth Ruvalcaba-Delgadillo
- Microscopía de Alta Resolución, Depto, de Neurociencias, Universidad de Guadalajara, Guadalajara 44340, Mexico; (R.C.-P.)
| | - Sergio Montero
- Facultad de Medicina, Universidad de Colima, Colima 28040, Mexico
| | - Mónica Lemus
- Centro Universitario de Investigaciones Biomédicas, Universidad de Colima, Colima 28040, Mexico
| | | | - Joaquín García-Estrada
- División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social, Guadalajara 44340, Mexico
| | - Sonia Luquín
- Microscopía de Alta Resolución, Depto, de Neurociencias, Universidad de Guadalajara, Guadalajara 44340, Mexico; (R.C.-P.)
| |
Collapse
|
8
|
Yan Y, Mao Z, Jia Q, Zhao XJ, Yang SH. Changes in blood pressure, oxygen saturation, hemoglobin concentration, and heart rate among low-altitude migrants living at high altitude (5380 m) for 360 days. Am J Hum Biol 2023; 35:e23913. [PMID: 37200487 DOI: 10.1002/ajhb.23913] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND This article aimed to study the adjustment and adaptation of resting systolic blood pressure (SBP), diastolic blood pressure (DPB), oxygen saturation (SpO2 ), hemoglobin concentration ([Hb]), and heart rate (HR) in low-altitude migrants during a 1-year stay at high altitude. MATERIALS AND METHODS Our study enrolled 35 young migrants who were exposed to a hypoxia environment at 5380 m altitude on the Qinghai Tibetan Plateau between June 21, 2017, and June 16, 2018. We set 14-time points (the 1st-10th, 20th, 30th, 180th, and 360th day after arriving at 5380 m) for obtaining the measurements of resting SBP, DBP, HR, SpO2, and [Hb] and compared them with the control values recorded prior to migration. Variables with continuous data were summarized as means (SD). One-way repeated measures ANOVA without assuming sphericity was carried out to test whether the mean values (SBP, DBP, HR, SpO2 , and [Hb]) on different days were different significantly. Furthermore, Dunnett's multiple comparisons test was carried out to determine the time points whose values were significantly different from the control values. RESULTS SBP and DBP were continually increasing within d1-3 and peaked on the 3rd day, then steadily declined from d3 to d30. SBP fell back to the control values on d10 (p > 0.05), and DBP fell back to the control values on d20 (p > 0.05). A significant decline occurred on d180 (p < 0.05). Both SBP and DBP were lower than the control values on d180 (p < 0.05), and this trend was maintained to d360. There were similar characteristics of HR and BP in the time course at HA. HR on d1-3 was increasing (p < 0.05) compared to the control values, after which it fell back to the control values on d180 (p > 0.05), and this trend was maintained to d360. SpO2 was the lowest on d1 and lower than the control value throughout the study at HA (p < 0.05). [Hb] increased after long-term exposure (180 and 360 days) to HA (p < 0.05). CONCLUSIONS Our study continuously monitored lowlanders at 5380 m in Tibet, and is perhaps the only longitudinal study of migrants conducted at an altitude above 5000 m during a 1-year period. Our study provides new information on the adjustment and adaptation of [Hb], SpO2 , SBP, DBP, and HR in high-altitude plateau migrants during a 360-day stay at an altitude of 5380 m.
Collapse
Affiliation(s)
- Yan Yan
- Translational Medicine Research Center, Medical Innovation Research Division, The Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhong Mao
- Respiratory Cardiology and Nephrology, The 957th Chinese PLA Hospital, Xizang, People's Republic of China
| | - Qian Jia
- Translational Medicine Research Center, Medical Innovation Research Division, The Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiao-Jing Zhao
- Translational Medicine Research Center, Medical Innovation Research Division, The Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Sheng-Hong Yang
- The 949th Chinese PLA Hospital, Xinjiang, People's Republic of China
| |
Collapse
|
9
|
Hemingway HW, Richey RE, Moore AM, Saul BM, Shokraeifard AM, Cope HL, Olivencia-Yurvati AH, Cunningham RL, Smith ML, Romero SA. Effect of acute heat exposure on the pressor response to a voluntary hypoxic apnea. J Appl Physiol (1985) 2023; 135:542-548. [PMID: 37439242 PMCID: PMC10538993 DOI: 10.1152/japplphysiol.00245.2023] [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: 04/19/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/14/2023] Open
Abstract
The pressor response induced by a voluntary hypoxic apnea is mediated largely by increased sympathetic outflow. The neural control of blood pressure is altered in recovery from acute heat exposure, but its effect on the pressor response to a voluntary hypoxic apnea has never been explored. Therefore, we tested the hypothesis that prior heat exposure would attenuate the pressor response induced by a voluntary hypoxic apnea. Eleven healthy adults (five women) were exposed to whole body passive heating (water-perfused suit) sufficient to increase body core temperature by 1.2°C. Voluntary hypoxic apneas were performed at baseline and in recovery when body core temperature returned to ≤ 0.3°C of baseline. Participants breathed gas mixtures of varying [Formula: see text] (21%, 16%, and 12%; randomized) for 1 min followed by a 15-s end-expiratory apnea. The change in arterial oxygen saturation during each apnea did not differ from baseline to recovery (P = 0.6 for interaction), whereas the pressor response induced by a voluntary hypoxia apnea was reduced ([Formula: see text] 21%, baseline 17 ± 7 mmHg vs. recovery 14 ± 7 mmHg; [Formula: see text] 16%, baseline 24 ± 8 mmHg vs. recovery 18 ± 7 mmHg; [Formula: see text] 12%, baseline 28 ± 11 mmHg vs. recovery 24 ± 11 mmHg; P = 0.01 for main effect of time). These data suggest that prior heat exposure induces a cross-stressor effect such that the pressor response to a voluntary hypoxic apnea is attenuated.NEW & NOTEWORTHY The pressor response induced by a voluntary hypoxic apnea is mediated by increased sympathetic outflow. The neural control of blood pressure is altered in recovery from acute heat exposure, but its effect on the pressor response to a voluntary hypoxic apnea has never been explored. Our data suggest that prior heat exposure induces a cross-stressor effect such that the pressor response to a voluntary hypoxic apnea is attenuated.
Collapse
Affiliation(s)
- Holden W Hemingway
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Rauchelle E Richey
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Amy M Moore
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Benjamin M Saul
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Austin M Shokraeifard
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Heidi L Cope
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Albert H Olivencia-Yurvati
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
- Department of Surgery, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Michael L Smith
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Steven A Romero
- Human Vascular Physiology Laboratory, Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| |
Collapse
|
10
|
Atanasova DY, Dandov AD, Lazarov NE. Neurochemical plasticity of the carotid body in hypertension. Anat Rec (Hoboken) 2023; 306:2366-2377. [PMID: 37561329 DOI: 10.1002/ar.24997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/20/2022] [Accepted: 05/05/2022] [Indexed: 11/11/2022]
Abstract
The carotid body (CB), a main peripheral arterial chemoreceptor, has lately been implicated in the pathophysiology of various cardiovascular disorders. Emerging experimental evidence supports a causal relationship between CB dysfunction and augmented sympathetic outflow which is the common hallmark of human sympathetic-related diseases, including essential hypertension. To gain insight into the neurotransmitter profile of chemosensory cells in the hypertensive CB, we examined the expression and cellular localization of some classical neurotransmitters, neuropeptides, and gaseous signaling molecules as well as neurotrophic factors and their receptors in the CB of spontaneously hypertensive rats, a common animal model of hypertension. Our immunohistochemical experiments revealed an elevated catecholamine and serotonin content in the hypertensive CB compared to normotensive controls. GABA immunostaining was seen in some peripherally located glomus cells in the CB of SHR and it was significantly lower than in control animals. The density of substance P and vasoactive intestinal peptide-immunoreactive fibers was diminished whereas that of neuropeptide Y-immunostained nerve fibers was increased and that of calcitonin gene-related peptide-containing fibers remained almost unchanged in the hypertensive CB. We have further demonstrated that in the hypertensive state the production of nitric oxide is impaired and that the components of the neurotrophin signaling system display an abnormal enhanced expression. Our results provide immunohistochemical evidence that the altered transmitter phenotype of CB chemoreceptor cells and the elevated production of neurotrophic factors modulate the chemosensory processing in hypertensive animals which contributes to autonomic dysfunction and elicits sympathetic hyperactivity, consequently leading to elevated blood pressure.
Collapse
Affiliation(s)
- Dimitrinka Y Atanasova
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
- Department of Anatomy, Faculty of Medicine, Trakia University, Stara Zagora, Bulgaria
| | - Angel D Dandov
- Department of Anatomy and Histology, Medical University of Sofia, Sofia, Bulgaria
| | - Nikolai E Lazarov
- Institute of Neurobiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
- Department of Anatomy and Histology, Medical University of Sofia, Sofia, Bulgaria
| |
Collapse
|
11
|
Karim S, Chahal A, Khanji MY, Petersen SE, Somers V. Autonomic Cardiovascular Control in Health and Disease. Compr Physiol 2023; 13:4493-4511. [PMID: 36994768 PMCID: PMC10406398 DOI: 10.1002/cphy.c210037] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Autonomic neural control of the cardiovascular system is formed of complex and dynamic processes able to adjust rapidly to mitigate perturbations in hemodynamics and maintain homeostasis. Alterations in autonomic control feature in the development or progression of a multitude of diseases with wide-ranging physiological implications given the neural system's responsibility for controlling inotropy, chronotropy, lusitropy, and dromotropy. Imbalances in sympathetic and parasympathetic neural control are also implicated in the development of arrhythmia in several cardiovascular conditions sparking interest in autonomic modulation as a form of treatment. A number of measures of autonomic function have shown prognostic significance in health and in pathological states and have undergone varying degrees of refinement, yet adoption into clinical practice remains extremely limited. The focus of this contemporary narrative review is to summarize the anatomy, physiology, and pathophysiology of the cardiovascular autonomic nervous system and describe the merits and shortfalls of testing modalities available. © 2023 American Physiological Society. Compr Physiol 13:4493-4511, 2023.
Collapse
Affiliation(s)
- Shahid Karim
- Mayo Clinic, Rochester, Minnesota, USA
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University London, UK
| | - Anwar Chahal
- Mayo Clinic, Rochester, Minnesota, USA
- University of Pennsylvania, Pennsylvania, USA
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University London, UK
| | - Mohammed Y. Khanji
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Newham University Hospital, Barts Health NHS Trust, London, UK
| | - Steffen E. Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Centre, Queen Mary University London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Health Data Research UK, London, UK
- Alan Turing Institute, London, UK
| | | |
Collapse
|
12
|
Ou Y, Zong D, Ouyang R. Role of epigenetic abnormalities and intervention in obstructive sleep apnea target organs. Chin Med J (Engl) 2023; 136:631-644. [PMID: 35245923 PMCID: PMC10129098 DOI: 10.1097/cm9.0000000000002080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Obstructive sleep apnea (OSA) is a common condition that has considerable impacts on human health. Epigenetics has become a rapidly developing and exciting area in biology, and it is defined as heritable alterations in gene expression and has regulatory effects on disease progression. However, the published literature that is integrating both of them is not sufficient. The purpose of this article is to explore the relationship between OSA and epigenetics and to offer better diagnostic methods and treatment options. Epigenetic modifications mainly manifest as post-translational modifications in DNA and histone proteins and regulation of non-coding RNAs. Chronic intermittent hypoxia-mediated epigenetic alterations are involved in the progression of OSA and diverse multiorgan injuries, including cardiovascular disease, metabolic disorders, pulmonary hypertension, neural dysfunction, and even tumors. This article provides deeper insights into the disease mechanism of OSA and potential applications of targeted diagnosis, treatment, and prognosis in OSA complications.
Collapse
Affiliation(s)
- Yanru Ou
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Dandan Zong
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| | - Ruoyun Ouyang
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Unit of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
- Diagnosis and Treatment Center of Respiratory Disease, Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
13
|
Felippe ISA, Zera T, da Silva MP, Moraes DJA, McBryde F, Paton JFR. The sympathetic nervous system exacerbates carotid body sensitivity in hypertension. Cardiovasc Res 2023; 119:316-331. [PMID: 35048948 PMCID: PMC10022867 DOI: 10.1093/cvr/cvac008] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/24/2021] [Accepted: 01/14/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS The carotid bodies (CBs) of spontaneously hypertensive (SH) rats exhibit hypertonicity and hyperreflexia contributing to heightened peripheral sympathetic outflow. We hypothesized that CB hyperexcitability is driven by its own sympathetic innervation. METHODS AND RESULTS To test this, the chemoreflex was activated (NaCN 50-100 µL, 0.4 µg/µL) in SH and Wistar rats in situ before and after: (i) electrical stimulation (ES; 30 Hz, 2 ms, 10 V) of the superior cervical ganglion (SCG), which innervates the CB; (ii) unilateral resection of the SCG (SCGx); (iii) CB injections of an α1-adrenergic receptor agonist (phenylephrine, 50 µL, 1 mmol/L), and (iv) α1-adrenergic receptor antagonist prazosin (40 µL, 1 mmol/L) or tamsulosin (50 µL, 1 mmol/L). ES of the SCG enhanced CB-evoked sympathoexcitation by 40-50% (P < 0.05) with no difference between rat strains. Unilateral SCGx attenuated the CB-evoked sympathoexcitation in SH (62%; P < 0.01) but was without effect in Wistar rats; it also abolished the ongoing firing of chemoreceptive petrosal neurones of SH rats, which became hyperpolarized. In Wistar rats, CB injections of phenylephrine enhanced CB-evoked sympathoexcitation (33%; P < 0.05), which was prevented by prazosin (26%; P < 0.05) in SH rats. Tamsulosin alone reproduced the effects of prazosin in SH rats and prevented the sensitizing effect of the SCG following ES. Within the CB, α1A- and α1B-adrenoreceptors were co-localized on both glomus cells and blood vessels. In conscious SH rats instrumented for recording blood pressure (BP), the CB-evoked pressor response was attenuated after SCGx, and systolic BP fell by 16 ± 4.85 mmHg. CONCLUSIONS The sympathetic innervation of the CB is tonically activated and sensitizes the CB of SH but not Wistar rats. Furthermore, sensitization of CB-evoked reflex sympathoexcitation appears to be mediated by α1-adrenoceptors located either on the vasculature and/or glomus cells. The SCG is novel target for controlling CB pathophysiology in hypertension.
Collapse
Affiliation(s)
- Igor S A Felippe
- Department of Physiology, Faculty of Health & Medical Sciences, Manaaki Mānawa—The Centre for Heart Research, University of Auckland, 85 Park Road, Grafton Campus, Auckland 1023, New Zealand
| | - Tymoteusz Zera
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw 02-091, Poland
| | - Melina P da Silva
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil
| | - Davi J A Moraes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil
| | - Fiona McBryde
- Department of Physiology, Faculty of Health & Medical Sciences, Manaaki Mānawa—The Centre for Heart Research, University of Auckland, 85 Park Road, Grafton Campus, Auckland 1023, New Zealand
| | | |
Collapse
|
14
|
Caballero-Eraso C, Colinas O, Sobrino V, González-Montelongo R, Cabeza JM, Gao L, Pardal R, López-Barneo J, Ortega-Sáenz P. Rearrangement of cell types in the rat carotid body neurogenic niche induced by chronic intermittent hypoxia. J Physiol 2023; 601:1017-1036. [PMID: 36647759 DOI: 10.1113/jp283897] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
The carotid body (CB) is a prototypical acute oxygen (O2 )-sensing organ that mediates reflex hyperventilation and increased cardiac output in response to hypoxaemia. CB overactivation, secondary to the repeated stimulation produced by the recurrent episodes of intermittent hypoxia, is believed to contribute to the pathogenesis of sympathetic hyperactivity present in sleep apnoea patients. Although CB functional plasticity induced by chronic intermittent hypoxia (CIH) has been demonstrated, the underlying mechanisms are not fully elucidated. Here, we show that CIH induces a small increase in CB volume and rearrangement of cell types in the CB, characterized by a mobilization of immature quiescent neuroblasts, which enter a process of differentiation into mature, O2 -sensing and neuron-like, chemoreceptor glomus cells. Prospective isolation of individual cell classes has allowed us to show that maturation of CB neuroblasts is paralleled by an upregulation in the expression of specific glomus cell genes involved in acute O2 -sensing. CIH enhances mitochondrial responsiveness to hypoxia in maturing neuroblasts as well as in glomus cells. These data provide novel perspectives on the pathogenesis of CB-mediated sympathetic overflow that may lead to the development of new pharmacological strategies of potential applicability in sleep apnoea patients. KEY POINTS: Obstructive sleep apnoea is a frequent condition in the human population that predisposes to severe cardiovascular and metabolic alterations. Activation of the carotid body, the main arterial oxygen-sensing chemoreceptor, by repeated episodes of hypoxaemia induces exacerbation of the carotid body-mediated chemoreflex and contributes to sympathetic overflow characteristic of sleep apnoea patients. In rats, chronic intermittent hypoxaemia induces fast neurogenesis in the carotid body with rapid activation of neuroblasts, which enter a process of proliferation and maturation into O2 -sensing chemoreceptor glomus cells. Maturing carotid body neuroblasts and glomus cells exposed to chronic intermittent hypoxia upregulate genes involved in acute O2 sensing and enhance mitochondrial responsiveness to hypoxia. These findings provide novel perspectives on the pathogenesis of carotid body-mediated sympathetic hyperactivation. Pharmacological modulation of carotid body fast neurogenesis could help to ameliorate the deleterious effects of chronic intermittent hypoxaemia in sleep apnoea patients.
Collapse
Affiliation(s)
- Candela Caballero-Eraso
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Unidad Médico Quirúrgica de Enfermedades Respiratorias, Hospital Universitario Virgen del Rocío/IBIS, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Olaia Colinas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Verónica Sobrino
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Rafaela González-Montelongo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José María Cabeza
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Lin Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Ricardo Pardal
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Patricia Ortega-Sáenz
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
15
|
Gagnon M, Fournier S, Marcouiller F, Guay L, Joseph V, Michael NJ, Kinkead R. Intermittent Hypoxia and Weight Loss: Insights into the Etiology of the Sleep Apnea Phenotype. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1427:61-71. [PMID: 37322336 DOI: 10.1007/978-3-031-32371-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Sleep apnea (SA) is a major respiratory disorder with increased risk for hypertension and obesity; however, our understanding of the origins of this complex disorder remains limited. Because apneas lead to recurrent drops in O2 during sleep, intermittent hypoxia (IH) is the main animal model to explore the pathophysiology of SA. Here, we assessed the impacts of IH on metabolic function and related signals. Adult male rats were exposed to 1 week of moderate IH (FiO2 = 0.10-30 s, ten cycles/hour, 8 h/day). Using whole-body plethysmography, we measured respiratory variability and apnea index during sleep. Blood pressure and heart rate were measured by the tail-cuff method; blood samples were taken for multiplex assay. At rest, IH augmented arterial blood pressure, respiratory instability, but not apnea index. IH induced weight, fat, and fluid loss. IH also reduced food intake and plasma leptin, adrenocorticotropic hormone (ACTH), and testosterone levels but increased inflammatory cytokines. We conclude that IH does not replicate the metabolic clinical features of SA patient, thus raising our awareness of the limitations of the IH model. The fact that the risk for hypertension occurs before the appearance of apneas provides new insights into the progression of the disease.
Collapse
Affiliation(s)
- Marianne Gagnon
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada.
- Faculty of Pharmacy, Université Laval, Québec City, QC, Canada.
| | - Stéphanie Fournier
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
| | - Loralie Guay
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
| | - Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
- Department of Pediatrics, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Natalie J Michael
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
- Faculty of Pharmacy, Université Laval, Québec City, QC, Canada
| | - Richard Kinkead
- Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec City, QC, Canada
- Department of Pediatrics, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
16
|
Lazarov NE, Atanasova DY. Neurochemical Plasticity of the Carotid Body. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 237:105-122. [PMID: 37946079 DOI: 10.1007/978-3-031-44757-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
A striking feature of the carotid body (CB) is its remarkable degree of plasticity in a variety of neurotransmitter/modulator systems in response to environmental stimuli, particularly following hypoxic exposure of animals and during ascent to high altitude. Current evidence suggests that acetylcholine and adenosine triphosphate are two major excitatory neurotransmitter candidates in the hypoxic CB, and they may also be involved as co-transmitters in hypoxic signaling. Conversely, dopamine, histamine and nitric oxide have recently been considered inhibitory transmitters/modulators of hypoxic chemosensitivity. It has also been revealed that interactions between excitatory and inhibitory messenger molecules occur during hypoxia. On the other hand, alterations in purinergic neurotransmitter mechanisms have been implicated in ventilatory acclimatization to hypoxia. Chronic hypoxia also induces profound changes in other neurochemical systems within the CB such as the catecholaminergic, peptidergic and nitrergic, which in turn may contribute to increased ventilatory and chemoreceptor responsiveness to hypoxia at high altitude. Taken together, current data suggest that complex interactions among transmitters markedly influence hypoxia-induced transmitter release from the CB. In addition, the expression of a wide variety of growth factors, proinflammatory cytokines and their receptors have been identified in CB parenchymal cells in response to hypoxia and their upregulated expression could mediate the local inflammation and functional alteration of the CB under hypoxic conditions.
Collapse
Affiliation(s)
- Nikolai E Lazarov
- Department of Anatomy and Histology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria.
| | | |
Collapse
|
17
|
Lazarov NE, Atanasova DY. Structural Plasticity of the Carotid Body. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 237:37-48. [PMID: 37946076 DOI: 10.1007/978-3-031-44757-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
The mammalian carotid body (CB) exhibits considerable plasticity of its structure during development and aging and as a consequence of environmental, metabolic and inflammatory stimuli. The structural changes during maturation include an enlargement of the total and vascular volume of the CB. Conversely, aging results in a reduction in the number and volume of glomus cells with progressive cellular degeneration and an apparent increase in the surrounding connective tissue. Age-related structural alterations are similar to those during chronic hypoxia. Long-term hypoxic exposure and sodium nitrate treatment enlarge several-fold the size of the rat CB causing glomus cell hypertrophy and hyperplasia, and evoke changes in its vascular structure, inducing marked vasodilation and neovascularization. In humans, such structural CB adaptation responses to prolonged hypoxia occur during acclimatization to high altitudes. On the other hand, the hyperoxic CB is significantly smaller than those of age-matched normoxic controls. Morphological alterations in the CB in both hypertensive animals and humans are characterized by a slightly enlarged parenchyma without apparent vascular expansion and/or dilation. The CB structural plasticity depends on the existence of a population of multipotent neural crest-derived stem cells, which are activated during hypoxia to proliferate and differentiate into new both neuronal (glomus) and vascular cell types.
Collapse
Affiliation(s)
- Nikolai E Lazarov
- Department of Anatomy and Histology, Faculty of Medicine, Medical University of Sofia, Sofia, Bulgaria.
| | | |
Collapse
|
18
|
Yoshizawa M, Fukushi I, Takeda K, Kono Y, Hasebe Y, Koizumi K, Ikeda K, Pokorski M, Toda T, Okada Y. Role of microglia in blood pressure and respiratory responses to acute hypoxic exposure in rats. J Physiol Sci 2022; 72:26. [PMID: 36229778 DOI: 10.1186/s12576-022-00848-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 09/02/2022] [Indexed: 11/10/2022]
Abstract
Microglia modulate cardiorespiratory activities during chronic hypoxia. It has not been clarified whether microglia are involved in the cardiorespiratory responses to acute hypoxia. Here we investigated this issue by comparing cardiorespiratory responses to two levels of acute hypoxia (13% O2 for 4 min and 7% O2 for 5 min) in conscious unrestrained rats before and after systemic injection of minocycline (MINO), an inhibitor of microglia activation. MINO increased blood pressure but not lung ventilation in the control normoxic condition. Acute hypoxia stimulated cardiorespiratory responses in MINO-untreated rats. MINO failed to significantly affect the magnitude of hypoxia-induced blood pressure elevation. In contrast, MINO tended to suppress the ventilatory responses to hypoxia. We conclude that microglia differentially affect cardiorespiratory regulation depending on the level of blood oxygenation. Microglia suppressively contribute to blood pressure regulation in normoxia but help maintain ventilatory augmentation in hypoxia, which underscores the dichotomy of central regulatory pathways for both systems.
Collapse
Affiliation(s)
- Masashi Yoshizawa
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.,Clinical Research Center, Murayama Medical Center, Tokyo, Japan
| | - Isato Fukushi
- Clinical Research Center, Murayama Medical Center, Tokyo, Japan.,Faculty of Health Sciences, Aomori University of Health and Welfare, Aomori, Japan
| | - Kotaro Takeda
- Clinical Research Center, Murayama Medical Center, Tokyo, Japan.,Faculty of Rehabilitation, School of Health Sciences, Fujita Health University, Toyoake, Japan
| | - Yosuke Kono
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.,Clinical Research Center, Murayama Medical Center, Tokyo, Japan
| | - Yohei Hasebe
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.,Clinical Research Center, Murayama Medical Center, Tokyo, Japan
| | - Keiichi Koizumi
- Department of Pediatrics, Fujiyoshida Municipal Hospital, Yamanashi, Japan
| | - Keiko Ikeda
- Institute of Innovative Research, Homeostatic Mechanism Research Unit, Tokyo Institute of Technology, Yokohama, Japan
| | | | - Takako Toda
- Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yasumasa Okada
- Clinical Research Center, Murayama Medical Center, Tokyo, Japan.
| |
Collapse
|
19
|
Oxygen regulation of breathing is abolished in mitochondrial complex III-deficient arterial chemoreceptors. Proc Natl Acad Sci U S A 2022; 119:e2202178119. [PMID: 36122208 PMCID: PMC9522341 DOI: 10.1073/pnas.2202178119] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Oxygen sensing by chemoreceptor glomus cells in the carotid body plays an essential adaptive function in health and disease; however, the underlying mechanisms are not fully understood. Glomus cells survive genetic disruption of mitochondrial complex III, although this results in a functional disconnection between the distal and proximal components of the mitochondrial electron transport chain (ETC). These cells exhibit selective abolition of mitochondrial and cellular responsiveness to hypoxia, as well as altered systemic hyperventilation and acclimatization to hypoxia, indicating that acute oxygen-sensing and -signaling during hypoxia result from the integrated action of mitochondrial ETC components. The mitochondrial ETC emerges as a complex oxygen-sensing and -signaling system of potential pathophysiological relevance in maladaptive responses to hypoxia. Acute oxygen (O2) sensing is essential for adaptation of organisms to hypoxic environments or medical conditions with restricted exchange of gases in the lung. The main acute O2-sensing organ is the carotid body (CB), which contains neurosecretory chemoreceptor (glomus) cells innervated by sensory fibers whose activation by hypoxia elicits hyperventilation and increased cardiac output. Glomus cells have mitochondria with specialized metabolic and electron transport chain (ETC) properties. Reduced mitochondrial complex (MC) IV activity by hypoxia leads to production of signaling molecules (NADH and reactive O2 species) in MCI and MCIII that modulate membrane ion channel activity. We studied mice with conditional genetic ablation of MCIII that disrupts the ETC in the CB and other catecholaminergic tissues. Glomus cells survived MCIII dysfunction but showed selective abolition of responsiveness to hypoxia (increased [Ca2+] and transmitter release) with normal responses to other stimuli. Mitochondrial hypoxic NADH and reactive O2 species signals were also suppressed. MCIII-deficient mice exhibited strong inhibition of the hypoxic ventilatory response and altered acclimatization to sustained hypoxia. These data indicate that a functional ETC, with coupling between MCI and MCIV, is required for acute O2 sensing. O2 regulation of breathing results from the integrated action of mitochondrial ETC complexes in arterial chemoreceptors.
Collapse
|
20
|
Shafer BM, Incognito AV, Vermeulen TD, Nardone M, Teixeira AL, Klassen SA, Millar PJ, Foster GE. Action potential amplitude and baroreflex resetting of action potential clusters mediate hypoxia-induced sympathetic long-term facilitation. J Physiol 2022; 600:3127-3147. [PMID: 35661360 DOI: 10.1113/jp282933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/03/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Acute isocapnic hypoxia resets the arterial baroreflex and permits long-lasting sympathoexcitation called sympathetic long-term facilitation. Our understanding of sympathetic long-term facilitation following hypoxia in humans is based on multiunit muscle sympathetic nerve activity and does not fully characterize the underlying baroreflex control of sympathetic neuronal subpopulations or their discharge/recruitment strategies. We show that sympathetic long-term facilitation is mediated by baroreflex resetting of sympathetic action potential clusters to higher arterial pressure operating points, a reduction in the percentage of action potentials firing asynchronously, and a shift toward larger amplitude action potential activity. The results advance our fundamental understanding of how the sympathetic nervous system mediates sympathetic long-term facilitation following exposure to acute isocapnic hypoxia in humans. ABSTRACT Baroreflex resetting permits sympathetic long-term facilitation (sLTF) following hypoxia; however, baroreflex control of action potential (AP) clusters and AP recruitment patterns facilitating sLTF is unknown. We hypothesized that baroreflex resetting of arterial pressure operating points (OPs) of AP clusters and recruitment of large-amplitude APs would mediate sLTF following hypoxia. Eight men (age: 24 (3) yrs; BMI: 24 (3) kg/m2 ) underwent 20-min isocapnic hypoxia (PET O2 : 47 (2) mmHg) and 30-min recovery. Multi-unit microneurography (muscle sympathetic nerve activity; MSNA) and a continuous wavelet transform with matched mother wavelet was used to detect sympathetic APs during baseline, hypoxia, early (first 5-min), and late recovery (last 5-min). AP amplitude (normalized to largest baseline AP amplitude), percent APs occurring outside a MSNA burst (% asynchronous APs), and proportion of APs firing in small (1-3), medium (4-6), and large (7-10) normalized cluster sizes was calculated. Normalized clusters were used to assess baroreflex OPs and sensitivity. Hypoxia increased total MSNA activity, which remained elevated during recovery (P<0.0001). Baroreflex OPs were shifted rightward for all clusters in recovery, with no effect on slope. Compared to baseline, AP amplitude was elevated by 3 (2) % and 4 (2) % while asynchronous APs were reduced by 9 (5) % and 7 (6) % in early and late recovery, respectively. In early recovery, the proportion of APs firing in large clusters was increased compared to baseline. Hypoxia-induced sLTF is mediated by baroreflex resetting of AP clusters to higher OPs, reduced asynchronous AP firing, and increased contribution from large-amplitude APs. Abstract figure legend Eight healthy men underwent 20-min isocapnic hypoxia and 30-min recovery. The study tested the hypothesis that baroreflex resetting of arterial pressure operating points (OPs) of action potential (AP) clusters and recruitment of large-amplitude APs would mediate sympathetic long-term facilitation (sLTF) following acute hypoxic exposure. Hypoxia increased multi-unit muscle sympathetic nerve activity (MSNA; measured via microneurography), which remained elevated throughout recovery. Sympathetic APs were detected in the filtered MSNA neurogram using a continuous wavelet transform with matched mother wavelet. An effect of condition revealed that compared to baseline, AP amplitude was elevated while asynchronous APs were reduced in early and late recovery, respectively. Our findings show that AP amplitude distributions are shifting towards larger AP amplitudes in all subjects following hypoxia. Our findings indicate that hypoxia-induced sLTF is mediated by baroreflex resetting of AP clusters to higher OPs, reduced asynchronous AP firing, and increased contribution from large-amplitude APs. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Brooke M Shafer
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Kelowna, Canada
| | - Anthony V Incognito
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| | - Tyler D Vermeulen
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Kelowna, Canada
| | - Massimo Nardone
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| | - André L Teixeira
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| | | | - Philip J Millar
- Department of Human Health and Nutritional Sciences, University of Guelph, Canada
| | - Glen E Foster
- Centre for Heart, Lung, and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Kelowna, Canada
| |
Collapse
|
21
|
Argent LP, Bose A, Paton JFR. Intra-carotid body inter-cellular communication. J R Soc N Z 2022; 53:332-361. [PMID: 39439480 PMCID: PMC11459819 DOI: 10.1080/03036758.2022.2079681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/14/2022] [Indexed: 10/18/2022]
Abstract
The classic peripheral chemoreflex response is a critical homeostatic mechanism. In healthy individuals, appropriate chemoreflex responses are triggered by acute activation of the carotid body - the principal chemosensory organ in mammals. However, the aberrant chronic activation of the carotid body can drive the elevated sympathetic activity underlying cardio-respiratory diseases such as hypertension, diabetes and heart failure. Carotid body resection induces intolerable side effects and so understanding how to modulate carotid body output without removing it, and whilst maintaining the physiological chemoreflex response, represents the next logical next step in the development of effective clinical interventions. By definition, excessive carotid body output must result from altered intra-carotid body inter-cellular communication. Alongside the canonical synaptic transmission from glomus cells to petrosal afferents, many other modes of information exchange in the carotid body have been identified, for example bidirectional signalling between type I and type II cells via ATP-induced ATP release, as well as electrical communication via gap junctions. Thus, herein we review the carotid body as an integrated circuit, discussing a variety of different inter-cellular signalling mechanisms and highlighting those that are potentially relevant to its pathological hyperactivity in disease with the aim of identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Liam P. Argent
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Aabharika Bose
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| | - Julian F. R. Paton
- Manaaki Manawa – the Centre for Heart Research, Department of Physiology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
Park HY, Kim SW, Jung WS, Kim J, Lim K. Hypoxic Therapy as a New Therapeutic Modality for Cardiovascular Benefit: A Mini Review. Rev Cardiovasc Med 2022; 23:161. [PMID: 39077598 PMCID: PMC11273974 DOI: 10.31083/j.rcm2305161] [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: 02/09/2022] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 07/31/2024] Open
Abstract
Cardiovascular diseases (CVDs) are recognized as one of the major causes of morbidity and mortality worldwide. Generally, most CVDs can be prevented by addressing behavioral risk factors, including smoking, unhealthy diet and obesity, lack of physical activity, and alcohol abuse. Therefore, it is important to have a healthy lifestyle by performing regular physical activity to improve cardiovascular health and diseases. However, a majority of adults worldwide do not meet the minimum recommendations for regular aerobic exercise, and overweight and obesity ratio continues to rise. In addition, obese individuals, with a high prevalence of CVDs, have a lower participation rate for exercise because of the strain on the musculoskeletal system. Hypoxic therapy, including exposure or exercise intervention under hypoxia, has been utilized as a new therapeutic modality for cardiovascular benefit and amelioration of CVDs. Hypoxic therapy shows various physiological and pathophysiological properties, including increased appetite suppression and dietary intake reduction, increased energy consumption, improved glycogen storage, enhanced fatty acid oxidation, improved myocardial angiogenesis or ventricular remodeling, augmentation of blood flow within the skeletal muscle vascular beds, and reduction of the burden on the musculoskeletal system making it applicable to patients with CVDs and obesity with attenuated cardiovascular function. In particular, hypoxic therapy is very effective in improving cardiovascular benefits and preventing CVDs by enhancing arterial function, vascular endothelial function, and hemorheological properties. These observations indicate that hypoxic therapy may be an important and essential strategy for improving cardiovascular health and reducing cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Hun-Young Park
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 05029 Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, 05029 Seoul, Republic of Korea
| | - Sung-Woo Kim
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 05029 Seoul, Republic of Korea
| | - Won-Sang Jung
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 05029 Seoul, Republic of Korea
| | - Jisu Kim
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 05029 Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, 05029 Seoul, Republic of Korea
| | - Kiwon Lim
- Department of Sports Medicine and Science, Graduate School, Konkuk University, 05029 Seoul, Republic of Korea
- Physical Activity and Performance Institute (PAPI), Konkuk University, 05029 Seoul, Republic of Korea
- Department of Physical Education, Konkuk University, 05029 Seoul, Republic of Korea
| |
Collapse
|
23
|
Adaptive cardiorespiratory changes to chronic continuous and intermittent hypoxia. HANDBOOK OF CLINICAL NEUROLOGY 2022; 188:103-123. [PMID: 35965023 PMCID: PMC9906984 DOI: 10.1016/b978-0-323-91534-2.00009-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This chapter reviews cardiorespiratory adaptations to chronic hypoxia (CH) experienced at high altitude and cardiorespiratory pathologies elicited by chronic intermittent hypoxia (CIH) occurring with obstructive sleep apnea (OSA). Short-term CH increases breathing (ventilatory acclimatization to hypoxia) and blood pressure (BP) through carotid body (CB) chemo reflex. Hyperplasia of glomus cells, alterations in ion channels, and recruitment of additional excitatory molecules are implicated in the heightened CB chemo reflex by CH. Transcriptional activation of hypoxia-inducible factors (HIF-1 and 2) is a major molecular mechanism underlying respiratory adaptations to short-term CH. High-altitude natives experiencing long-term CH exhibit blunted hypoxic ventilatory response (HVR) and reduced BP due to desensitization of CB response to hypoxia and impaired processing of CB sensory information at the central nervous system. Ventilatory changes evoked by long-term CH are not readily reversed after return to sea level. OSA patients and rodents subjected to CIH exhibit heightened CB chemo reflex, increased hypoxic ventilatory response, and hypertension. Increased generation of reactive oxygen species (ROS) is a major cellular mechanism underlying CIH-induced enhanced CB chemo reflex and the ensuing cardiorespiratory pathologies. ROS generation by CIH is mediated by nontranscriptional, disrupted HIF-1 and HIF-2-dependent transcriptions as well as epigenetic mechanisms.
Collapse
|
24
|
Iturriaga R, Del Rio R, Alcayaga J. Carotid Body Inflammation: Role in Hypoxia and in the Anti-inflammatory Reflex. Physiology (Bethesda) 2021; 37:128-140. [PMID: 34866399 DOI: 10.1152/physiol.00031.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Emergent evidence indicates that the carotid body (CB) chemoreceptors may sense systemic inflammatory molecules, and is an afferent-arm of the anti-inflammatory reflex. Moreover, a pro-inflammatory milieu within the CB is involved in the enhanced CB chemosensory responsiveness to oxygen following sustained and intermittent hypoxia. In this review, we focus on the physio-pathological participation of CBs in inflammatory diseases, such as sepsis and intermittent hypoxia.
Collapse
Affiliation(s)
- Rodrigo Iturriaga
- Laboratorio de Neurobiologia. Departamento de Fisiologia. Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Pontificia Universidad Catolica de Chile, Santiago-1, Región, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Santiago, Chile
| | - Rodrigo Del Rio
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Santiago, Chile.,Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Julio Alcayaga
- Laboratorio de Fisiología Celular, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
25
|
Ganouna-Cohen G, Khadangi F, Marcouiller F, Bossé Y, Joseph V. Additive effects of orchiectomy and intermittent hypoxia on lung mechanics and inflammation in C57BL/6J male mice. Exp Physiol 2021; 107:68-81. [PMID: 34761830 DOI: 10.1113/ep090050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/05/2021] [Indexed: 01/01/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does endogenous testosterone modulate the consequences of intermittent hypoxia (IH) in the lungs of male mice? What is the main finding and its importance? Orchiectomized mice exposed to IH develop a pattern that is similar to emphysema or obstructive lung disease with elevated lung volumes, low pulmonary elastance during a methacholine challenge test and high counts of lymphocytes in bronchoalveolar lavages. Since low testosterone levels and other respiratory diseases are common in sleep apnoea, there is a clear clinical relevance to these results. ABSTRACT We tested the hypothesis that low testosterone levels modulate the pulmonary responses to intermittent hypoxia (IH; used as a model of sleep apnoea (SA)) in male mice. We used intact (SHAM) or orchiectomized (ORX) mice exposed to IH for 14 days (12 h/day, 10 cycles/h, 6% oxygen) or to normoxia (Nx). We first measured ventilation and metabolic rates in freely behaving mice (whole-body plethysmography) and then respiratory mechanics in tracheotomized mice (flexiVent). We assessed the respiratory system resistance and elastance (Ers ), Newtonian resistance (resistance of the large airways), tissue damping and tissue elastance (H) under baseline conditions and during a methacholine challenge test. We also measured the quasi-static compliance and inspiratory capacity with partial pressure-volume loops. Finally, inflammatory cells were counted in the broncho-alveolar lavage (BAL) and we measured lung volume by water displacement. ORX-IH mice had higher tidal volume, inspiratory capacity and lung volume compared to the other groups, but showed signs of low efficiency of O2 exchange rate relative to minute ventilation. During the methacholine challenge, orchiectomy decreased the values of most mechanical parameters and IH reduced Ers and H leading to very low values in ORX-IH mice. Finally, the total number of cells and the number of lymphocytes in BAL were both increased by IH in ORX mice. Since reduced lung elasticity, low O2 extraction, increased lung volumes and inflammation are signs of emphysematous lung disease, we conclude that testosterone might prevent lung emphysema during IH exposures.
Collapse
Affiliation(s)
- Gauthier Ganouna-Cohen
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie du Québec, Université Laval, Québec, QC, Canada
| | - Fatemeh Khadangi
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie du Québec, Université Laval, Québec, QC, Canada
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie du Québec, Université Laval, Québec, QC, Canada
| | - Ynuk Bossé
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie du Québec, Université Laval, Québec, QC, Canada
| | - Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie du Québec, Université Laval, Québec, QC, Canada
| |
Collapse
|
26
|
Stewart JM, Pianosi PT. Postural orthostatic tachycardia syndrome: A respiratory disorder? Curr Res Physiol 2021; 4:1-6. [PMID: 34746821 PMCID: PMC8562237 DOI: 10.1016/j.crphys.2021.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 11/28/2022] Open
Abstract
Postural orthostatic tachycardia syndrome (POTS) is a disorder epitomized by the story of the blind men and the elephant. Patients may see primary care internists or pediatricians due to fatigue, be referred to neurologists for “spells”, to cardiologists for evaluation of pre-syncope or chest pain, to gastroenterologists for nausea or dyspepsia, and even pulmonologists for dyspnea. Adoption of a more systematic approach to their evaluation and better characterization of patients has led to greater understanding of comorbidities, hypotheses prompting mechanistic investigations, and pharmacologic trials. Recent work has implicated disordered sympathetic nervous system activation in response to central (thoracic) hypovolemia. It is this pathway that leads one zero in on a putative focal point from which many of the clinical manifestations can be explained – specifically the carotid body. Despite heterogeneity in etiopathogenesis of a POTS phenotype, we propose that aberrant activation and response of the carotid body represents one potential common pathway in evolution. To understand this postulate, one must jettison isolationist or reductionist ideas of chemoreceptor and baroreceptor functions of the carotid body or sinus, respectively, and consider their interaction and interdependence both locally and centrally where some of its efferents merge. Doing so enables one to connect the dots and appreciate origins of diverse manifestations of POTS, including dyspnea for which the concept of neuro-mechanical uncoupling is wanting, thereby expanding our construct of this symptom. This perspective expounds our premise that POTS has a prominent respiratory component. Dyspnea affects ~⅓ patients with postural orthostatic tachycardia syndrome (POTS). POTS is characterized by thoracic hypovolemia and compromised cephalad perfusion when upright. Carotid body and adjacent carotid sinus mediate chemo- and baro- reflexes, respectively. These are not independent and stimulation of either activates sympathetic discharge. We speculate that carotid body mediates hyperventilation and dyspnea in POTS.
Collapse
Affiliation(s)
- Julian M Stewart
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Physiology, New York Medical College, Valhalla, NY, USA
| | - Paolo T Pianosi
- Department of Pediatrics, Division of Pulmonary & Sleep Medicine, University of Minnesota, VCRC, 401 E River Parkway Rm 413, Minneapolis, UK
| |
Collapse
|
27
|
Fukushi I, Takeda K, Pokorski M, Kono Y, Yoshizawa M, Hasebe Y, Nakao A, Mori Y, Onimaru H, Okada Y. Activation of Astrocytes in the Persistence of Post-hypoxic Respiratory Augmentation. Front Physiol 2021; 12:757731. [PMID: 34690820 PMCID: PMC8531090 DOI: 10.3389/fphys.2021.757731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 09/15/2021] [Indexed: 11/16/2022] Open
Abstract
Acute hypoxia increases ventilation. After cessation of hypoxia loading, ventilation decreases but remains above the pre-exposure baseline level for a time. However, the mechanism of this post-hypoxic persistent respiratory augmentation (PHRA), which is a short-term potentiation of breathing, has not been elucidated. We aimed to test the hypothesis that astrocytes are involved in PHRA. To this end, we investigated hypoxic ventilatory responses by whole-body plethysmography in unanesthetized adult mice. The animals breathed room air, hypoxic gas mixture (7% O2, 93% N2) for 2min, and again room air for 10min before and after i.p. administration of low (100mg/kg) and high (300mg/kg) doses of arundic acid (AA), an astrocyte inhibitor. AA suppressed PHRA, with the high dose decreasing ventilation below the pre-hypoxic level. Further, we investigated the role of the astrocytic TRPA1 channel, a putative ventilatory hypoxia sensor, in PHRA using astrocyte-specific Trpa1 knockout (asTrpa1−/−) and floxed Trpa1 (Trpa1f/f) mice. In both Trpa1f/f and asTrpa1−/− mice, PHRA was noticeable, indicating that the astrocyte TRPA1 channel was not directly involved in PHRA. Taken together, these results indicate that astrocytes mediate the PHRA by mechanisms other than TRPA1 channels that are engaged in hypoxia sensing.
Collapse
Affiliation(s)
- Isato Fukushi
- Faculty of Health Sciences, Uekusa Gakuen University, Chiba, Japan.,Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan
| | - Kotaro Takeda
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan.,Faculty of Rehabilitation, School of Healthcare, Fujita Health University, Toyoake, Japan
| | - Mieczyslaw Pokorski
- Institute of Health Sciences, University of Opole, Opole, Poland.,Faculty of Health Sciences, The Jan Dlugosz University in Czestochowa, Czestochowa, Poland
| | - Yosuke Kono
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan.,Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Masashi Yoshizawa
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan.,Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yohei Hasebe
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan.,Department of Pediatrics, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Akito Nakao
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Hiroshi Onimaru
- Department of Physiology, Showa University School of Medicine, Tokyo, Japan
| | - Yasumasa Okada
- Clinical Research Center, Murayama Medical Center, Musashimurayama, Japan
| |
Collapse
|
28
|
Csizmadia S, Fodor GH, Palkó A, Vörös E. Size of the Carotid Body in Patients with Cardiovascular and Respiratory Diseases Measured by Computed Tomography Angiography: A Case-Control Study. Radiol Res Pract 2021; 2021:9499420. [PMID: 34697571 PMCID: PMC8538397 DOI: 10.1155/2021/9499420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Carotid bodies (CBs) play an important role in regulating sympathetic nervous system activity. Thus, they are likely to be enlarged in patients with certain cardiovascular and respiratory diseases. The aim of this case-control study was to verify this hypothesis using computed tomography angiography (CTA). METHODS We retrospectively analysed 141 CTAs including 16 controls, 96 patients with only hypertension (HT), 12 with HT and previous acute myocardial infarction (AMI), 9 with HT and heart failure (HF), and 8 with HT and chronic obstructive pulmonary disease (COPD). We assessed the data using analysis of variance, with p < 0.05 indicating significance. RESULTS CB average areas in the controls were 2.31 mm2 (right side (RS)) vs. 2.34 mm2 (left side (LS)). CB size was significantly enlarged in patients with HT: 3.07 mm2 (RS) (p=0.019) vs. 2.91 mm2 (LS) (p=0.002). If AMI (RS: 3.5 mm2; LS: 3.44 mm2) or HF (RS: 4.01 mm2; LS: 4.55 mm2) was associated with HT, the CB size was even more enlarged. COPD did not affect CB size (RS: 2.40 mm2; LS: 2.29 mm2). CONCLUSIONS Our data showed that certain diseases with increased activity of the sympathetic nervous system were associated with significantly enlarged CBs.
Collapse
Affiliation(s)
- Sándor Csizmadia
- Affidea Hungary Ltd. Budapest, 44-46 Bókay János Street, Budapest H-1083, Hungary
| | - Gergely H. Fodor
- Department of Medical Physics and Informatics, University of Szeged, Faculty of General Medicine, 9 Korányi Alley, Szeged H-6725, Hungary
| | - András Palkó
- Department of Radiology, University of Szeged, Faculty of General Medicine, 6 Semmelweis Street, Szeged H-6725, Hungary
| | - Erika Vörös
- Department of Radiology, University of Szeged, Faculty of General Medicine, 6 Semmelweis Street, Szeged H-6725, Hungary
| |
Collapse
|
29
|
Investigation of Hemodynamic Receptors of the Internal Carotid Artery Segments. Artery Res 2021. [DOI: 10.1007/s44200-021-00005-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Abstract
Objectives
Internal carotid artery (ICA), the main artery of the brain, passes through the cavernous sinus (CS) which forms one of these venous pools. During this transition, while there is arterial blood in the lumen of ICA, its outer surface is in contact with venous blood from the brain. Herein, we aimed to detect the receptor differences of ICA in this highly specialized anatomical region of the skull base.
Methods
We performed the study on 10 human cadavers and searched CGRPR, TRP12, ASIC3 and ACTHR receptors via immunostaining using laser scanning confocal microscopy.
Results
We determined TRP12 receptor positive in the tunica media and tunica adventitia layers of the cavernous segment of ICA. We did not detect similar positivity in the cervical part of the ICA. In the receptor scan we made in terms of CGRPR, while we detected positivity in the tunica media layer of the cavernous segment, we found positivity in the tunica intima layer of the cervicalis segment of the ICA. We did not detect any positivity for ASIC3 and ACTHR receptors in both parts of the ICA.
Conclusions
As a result, we observed various differences in receptors between ICA segments. While the outer surface of the ICA in the cervical region did not show any receptor positivity, we detected TRP12 receptor positivity along the tissue contour of vessel in the CS. We assume that it may provide a new perspective on pathologies of the CS/ICA and preservation of brain hemodynamics for clinicians.
Collapse
|
30
|
Kinkead R, Gagnon M, Joseph V, Sériès F, Ambrozio-Marques D. Stress and Loss of Ovarian Function: Novel Insights into the Origins of Sex-Based Differences in the Manifestations of Respiratory Control Disorders During Sleep. Clin Chest Med 2021; 42:391-405. [PMID: 34353446 DOI: 10.1016/j.ccm.2021.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The respiratory system of women and men develops and functions in distinct neuroendocrine milieus. Despite differences in anatomy and neural control, homeostasis of arterial blood gases is ensured in healthy individuals regardless of sex. This convergence in function differs from the sex-based differences observed in many respiratory diseases. Sleep-disordered breathing (SDB) results mainly from episodes of upper airway closure. This complex and multifactorial respiratory disorder shows significant sexual dimorphism in its clinical manifestations and comorbidities. Guided by recent progress from basic research, this review discusses the hypothesis that stress is necessary to reveal the sexual dimorphism of SDB.
Collapse
Affiliation(s)
- Richard Kinkead
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada.
| | - Marianne Gagnon
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada
| | - Vincent Joseph
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada
| | - Frédéric Sériès
- Department of Medicine, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, Québec, Québec, Canada
| | - Danuzia Ambrozio-Marques
- Department of Pediatrics, Université Laval, Centre de Recherche de l'Institut Universitaire de Cardiologie et Pneumologie de Québec, 2725 Chemin Ste-Foy, Québec, Québec G1V 4G5, Canada
| |
Collapse
|
31
|
Iturriaga R, Alcayaga J, Chapleau MW, Somers VK. Carotid body chemoreceptors: physiology, pathology, and implications for health and disease. Physiol Rev 2021; 101:1177-1235. [PMID: 33570461 PMCID: PMC8526340 DOI: 10.1152/physrev.00039.2019] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The carotid body (CB) is the main peripheral chemoreceptor for arterial respiratory gases O2 and CO2 and pH, eliciting reflex ventilatory, cardiovascular, and humoral responses to maintain homeostasis. This review examines the fundamental biology underlying CB chemoreceptor function, its contribution to integrated physiological responses, and its role in maintaining health and potentiating disease. Emphasis is placed on 1) transduction mechanisms in chemoreceptor (type I) cells, highlighting the role played by the hypoxic inhibition of O2-dependent K+ channels and mitochondrial oxidative metabolism, and their modification by intracellular molecules and other ion channels; 2) synaptic mechanisms linking type I cells and petrosal nerve terminals, focusing on the role played by the main proposed transmitters and modulatory gases, and the participation of glial cells in regulation of the chemosensory process; 3) integrated reflex responses to CB activation, emphasizing that the responses differ dramatically depending on the nature of the physiological, pathological, or environmental challenges, and the interactions of the chemoreceptor reflex with other reflexes in optimizing oxygen delivery to the tissues; and 4) the contribution of enhanced CB chemosensory discharge to autonomic and cardiorespiratory pathophysiology in obstructive sleep apnea, congestive heart failure, resistant hypertension, and metabolic diseases and how modulation of enhanced CB reactivity in disease conditions may attenuate pathophysiology.
Collapse
Affiliation(s)
- Rodrigo Iturriaga
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile, and Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
| | - Julio Alcayaga
- Laboratorio de Fisiología Celular, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Mark W Chapleau
- Department of Internal Medicine, University of Iowa and Department of Veterans Affairs Medical Center, Iowa City, Iowa
| | - Virend K Somers
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
32
|
Tenorio-Lopes L, Kinkead R. Sex-Specific Effects of Stress on Respiratory Control: Plasticity, Adaptation, and Dysfunction. Compr Physiol 2021; 11:2097-2134. [PMID: 34107062 DOI: 10.1002/cphy.c200022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As our understanding of respiratory control evolves, we appreciate how the basic neurobiological principles of plasticity discovered in other systems shape the development and function of the respiratory control system. While breathing is a robust homeostatic function, there is growing evidence that stress disrupts respiratory control in ways that predispose to disease. Neonatal stress (in the form of maternal separation) affects "classical" respiratory control structures such as the peripheral O2 sensors (carotid bodies) and the medulla (e.g., nucleus of the solitary tract). Furthermore, early life stress disrupts the paraventricular nucleus of the hypothalamus (PVH), a structure that has emerged as a primary determinant of the intensity of the ventilatory response to hypoxia. Although underestimated, the PVH's influence on respiratory function is a logical extension of the hypothalamic control of metabolic demand and supply. In this article, we review the functional and anatomical links between the stress neuroendocrine axis and the medullary network regulating breathing. We then present the persistent and sex-specific effects of neonatal stress on respiratory control in adult rats. The similarities between the respiratory phenotype of stressed rats and clinical manifestations of respiratory control disorders such as sleep-disordered breathing and panic attacks are remarkable. These observations are in line with the scientific consensus that the origins of adult disease are often found among developmental and biological disruptions occurring during early life. These observations bring a different perspective on the structural hierarchy of respiratory homeostasis and point to new directions in our understanding of the etiology of respiratory control disorders. © 2021 American Physiological Society. Compr Physiol 11:1-38, 2021.
Collapse
Affiliation(s)
- Luana Tenorio-Lopes
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, The University of Calgary, Calgary, Alberta, Canada
| | - Richard Kinkead
- Département de Pédiatrie, Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
33
|
Drummond SE, Burns DP, O'Connor KM, Clarke G, O'Halloran KD. The role of NADPH oxidase in chronic intermittent hypoxia-induced respiratory plasticity in adult male mice. Respir Physiol Neurobiol 2021; 292:103713. [PMID: 34116239 DOI: 10.1016/j.resp.2021.103713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/18/2021] [Accepted: 06/06/2021] [Indexed: 10/21/2022]
Abstract
Reactive oxygen species (ROS) are proposed as mediators of chronic intermittent hypoxia (CIH)-induced respiratory plasticity. We sought to determine if NADPH oxidase 2 (NOX2)-derived ROS underpin CIH-induced maladaptive changes in respiratory control. Adult male mice (C57BL/6 J) were assigned to one of three groups: normoxic controls (sham); chronic intermittent hypoxia-exposed (CIH, 12 cycles/hour, 8 h/day for 14 days); and CIH + apocynin (NOX2 inhibitor, 2 mM) given in the drinking water throughout exposure to CIH. In addition, we studied sham and CIH-exposed NOX2-null mice (B6.129S-CybbTM1Din/J). Whole-body plethysmography was used to measure breathing and metabolic parameters. Ventilation (V̇I/V̇CO2) during normoxia was unaffected by CIH, but apnoea index was increased, which was prevented by apocynin, but not by NOX2 deletion. The ventilatory response to hypercapnia following exposure to CIH was potentiated in NOX2-null mice. Our results reveal ROS-dependent influences on the control of breathing and point to antioxidant intervention as a potential adjunctive therapeutic strategy in respiratory control disorders.
Collapse
Affiliation(s)
- Sarah E Drummond
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
34
|
Wang N, Peng YJ, Su X, Prabhakar NR, Nanduri J. Histone Deacetylase 5 Is an Early Epigenetic Regulator of Intermittent Hypoxia Induced Sympathetic Nerve Activation and Blood Pressure. Front Physiol 2021; 12:688322. [PMID: 34079475 PMCID: PMC8165245 DOI: 10.3389/fphys.2021.688322] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/26/2021] [Indexed: 12/29/2022] Open
Abstract
Intermittent hypoxia (IH) is a hallmark manifestation of obstructive sleep apnea (OSA). Long term IH (LT-IH) triggers epigenetic reprogramming of the redox state involving DNA hypermethylation in the carotid body chemo reflex pathway resulting in persistent sympathetic activation and hypertension. Present study examined whether IH also activates epigenetic mechanism(s) other than DNA methylation. Histone modification by lysine acetylation is another major epigenetic mechanism associated with gene regulation. Equilibrium between the activities of histone acetyltransferases (HATs) and histone deacetylases (HDACs) determine the level of lysine acetylation. Here we report that exposure of rat pheochromocytoma (PC)-12 cells to IH in vitro exhibited reduced HDAC enzyme activity due to proteasomal degradation of HDAC3 and HDAC5 proteins. Mechanistic investigations showed that IH-evoked decrease in HDAC activity increases lysine acetylation of α subunit of hypoxia inducible factor (HIF)-1α as well as Histone (H3) protein resulting in increased HIF-1 transcriptional activity. Trichostatin A (TSA), an inhibitor of HDACs, mimicked the effects of IH. Studies on rats treated with 10 days of IH or TSA showed reduced HDAC activity, HDAC5 protein, and increased HIF-1 dependent NADPH oxidase (NOX)-4 transcription in adrenal medullae (AM) resulting in elevated plasma catecholamines and blood pressure. Likewise, heme oxygenase (HO)-2 null mice, which exhibit IH because of high incidence of spontaneous apneas (apnea index 72 ± 1.2 apnea/h), also showed decreased HDAC activity and HDAC5 protein in the AM along with elevated circulating norepinephrine levels. These findings demonstrate that lysine acetylation of histone and non-histone proteins is an early epigenetic mechanism associated with sympathetic nerve activation and hypertension in rodent models of IH.
Collapse
Affiliation(s)
- Ning Wang
- Institute for Integrative Physiology, Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL, United States
| | - Ying-Jie Peng
- Institute for Integrative Physiology, Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL, United States
| | - Xiaoyu Su
- Institute for Integrative Physiology, Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL, United States
| | - Nanduri R Prabhakar
- Institute for Integrative Physiology, Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL, United States
| | - Jayasri Nanduri
- Institute for Integrative Physiology, Center for Systems Biology of O2 Sensing, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
35
|
Peng YJ, Su X, Wang B, Matthews T, Nanduri J, Prabhakar NR. Role of olfactory receptor78 in carotid body-dependent sympathetic activation and hypertension in murine models of chronic intermittent hypoxia. J Neurophysiol 2021; 125:2054-2067. [PMID: 33909496 DOI: 10.1152/jn.00067.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Chronic intermittent hypoxia (CIH) is a hallmark manifestation of obstructive sleep apnea (OSA), a widespread breathing disorder. CIH-treated rodents exhibit activation of the sympathetic nervous system and hypertension. Heightened carotid body (CB) activity has been implicated in CIH-induced hypertension. CB expresses high abundance of olfactory receptor (Olfr) 78, a G-protein coupled receptor. Olfr 78 null mice exhibit impaired CB sensory nerve response to acute hypoxia. Present study examined whether Olfr78 participates in CB-dependent activation of the sympathetic nervous system and hypertension in CIH-treated mice and in hemeoxygenase (HO)-2 null mice experiencing CIH as a consequence of naturally occurring OSA. CIH-treated wild-type (WT) mice showed hypertension, biomarkers of sympathetic nerve activation, and enhanced CB sensory nerve response to hypoxia and sensory long-term facilitation (sLTF), and these responses were absent in CIH-treated Olfr78 null mice. HO-2 null mice showed higher apnea index (AI) (58 ± 1.2 apneas/h) than WT mice (AI = 8 ± 0.8 apneas/h) and exhibited elevated blood pressure (BP), elevated plasma norepinephrine (NE) levels, and heightened CB sensory nerve response to hypoxia and sLTF. The magnitude of hypertension correlated with AI in HO-2 null mice. In contrast, HO-2/Olfr78 double null mice showed absence of elevated BP and plasma NE levels and augmented CB response to hypoxia and sLTF. These results demonstrate that Olfr78 participates in sympathetic nerve activation and hypertension and heightened CB activity in two murine models of CIH.NEW & NOTEWORTHY Carotid body (CB) sensory nerve activation is essential for sympathetic nerve excitation and hypertension in rodents treated with chronic intermittent hypoxia (CIH) simulating blood O2 profiles during obstructive sleep apnea (OSA). Here, we report that CIH-treated mice and hemeoxygenase (HO)-2-deficient mice, which show OSA phenotype, exhibit sympathetic excitation, hypertension, and CB activation. These effects are absent in Olfr78 null and Olfr78/HO-2 double null mice.
Collapse
Affiliation(s)
- Ying-Jie Peng
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| | - Xiaoyu Su
- 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
| | - Timothy Matthews
- 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
| | - Nanduri R Prabhakar
- Institute for Integrative Physiology and Center for Systems Biology of O2 Sensing, University of Chicago, Chicago, Illinois
| |
Collapse
|
36
|
Kim SW, Jung WS, Chung S, Park HY. Exercise intervention under hypoxic condition as a new therapeutic paradigm for type 2 diabetes mellitus: A narrative review. World J Diabetes 2021; 12:331-343. [PMID: 33889283 PMCID: PMC8040082 DOI: 10.4239/wjd.v12.i4.331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 01/25/2021] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
This review aims to summarize the health benefits of exposure to hypoxic conditions during exercise in patients with type 2 diabetes mellitus (T2DM). Exposure to hypoxic conditions during exercise training positively changes the physiological response in healthy subjects. Exposure to hypoxic conditions during exercise could markedly increase skeletal muscle glucose uptake compared to that in normoxic conditions. Furthermore, post-exercise insulin sensitivity of T2DM patients increases more when exercising under hypoxic than under normoxic conditions. Regular exercise under short-term hypoxic conditions can improve blood glucose control at lower workloads than in normoxic conditions. Additionally, exercise training under short-term hypoxic conditions can maximize weight loss in overweight and obese patients. Previous studies on healthy subjects have reported that regular exercise under hypoxic conditions had a more positive effect on vascular health than exercising under normoxic conditions. However, currently, evidence indicating that exposure to hypoxic conditions could positively affect T2DM patients in the long-term is lacking. Therefore, further evaluations of the beneficial effects of exercise under hypoxic conditions on the human body, considering different cycle lengths, duration of exposures, sessions per day, and the number of days, are necessary. In this review, we conclude that there is evidence that exercise under hypoxic conditions can yield health benefits, which is potentially valuable in terms of clinical care as a new intervention for T2DM patients.
Collapse
Affiliation(s)
- Sung-Woo Kim
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul 05029, South Korea
| | - Won-Sang Jung
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul 05029, South Korea
| | - Sochung Chung
- Department of Pediatrics, Konkuk University Medical Center, Research Institute of Medical Science, Konkuk University, School of Medicine, Seoul 05029, South Korea
| | - Hun-Young Park
- Physical Activity and Performance Institute (PAPI), Konkuk University, Seoul 05029, South Korea
- Department of Sports Science and Medicine, Konkuk University, Seoul 05029, South Korea
| |
Collapse
|
37
|
Marciante AB, Shell B, Farmer GE, Cunningham JT. Role of angiotensin II in chronic intermittent hypoxia-induced hypertension and cognitive decline. Am J Physiol Regul Integr Comp Physiol 2021; 320:R519-R525. [PMID: 33595364 PMCID: PMC8238144 DOI: 10.1152/ajpregu.00222.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 01/19/2021] [Accepted: 02/13/2021] [Indexed: 02/03/2023]
Abstract
Sleep apnea is characterized by momentary interruptions in normal respiration and leads to periods of decreased oxygen, or intermittent hypoxia. Chronic intermittent hypoxia is a model of the hypoxemia associated with sleep apnea and results in a sustained hypertension that is maintained during normoxia. Adaptations of the carotid body and activation of the renin-angiotensin system may contribute to the development of hypertension associated with chronic intermittent hypoxia. The subsequent activation of the brain renin-angiotensin system may produce changes in sympathetic regulatory neural networks that support the maintenance of the hypertension associated with intermittent hypoxia. Hypertension and sleep apnea not only increase risk for cardiovascular disease but are also risk factors for cognitive decline and Alzheimer's disease. Activation of the angiotensin system could be a common mechanism that links these disorders.
Collapse
Affiliation(s)
- Alexandria B Marciante
- Breathing REsearch And THErapeutics (BREATHE) Center, University of Florida, Gainesville, Florida
- Department of Physical Therapy, University of Florida, Gainesville, Florida
- McKnight Brain Institute, University of Florida, Gainesville, Florida
| | - Brent Shell
- Zuckerberg College of Health Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts
- Department of Biomedical and Nutritional Sciences, University of Massachusetts-Lowell, Lowell, Massachusetts
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
38
|
Prabhakar NR, Peng YJ, Nanduri J. Hypoxia-inducible factors and obstructive sleep apnea. J Clin Invest 2021; 130:5042-5051. [PMID: 32730232 DOI: 10.1172/jci137560] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Intermittent hypoxia (IH) is a hallmark manifestation of obstructive sleep apnea (OSA), a widespread disorder of breathing. This Review focuses on the role of hypoxia-inducible factors (HIFs) in hypertension, type 2 diabetes (T2D), and cognitive decline in experimental models of IH patterned after O2 profiles seen in OSA. IH increases HIF-1α and decreases HIF-2α protein levels. Dysregulated HIFs increase reactive oxygen species (ROS) through HIF-1-dependent activation of pro-oxidant enzyme genes in addition to reduced transcription of antioxidant genes by HIF-2. ROS in turn activate chemoreflex and suppress baroreflex, thereby stimulating the sympathetic nervous system and causing hypertension. We also discuss how increased ROS generation by HIF-1 contributes to IH-induced insulin resistance and T2D as well as disrupted NMDA receptor signaling in the hippocampus, resulting in cognitive decline.
Collapse
|
39
|
Steiner AR, Rousseau-Blass F, Schroeter A, Hartnack S, Bettschart-Wolfensberger R. Systematic Review: Anaesthetic Protocols and Management as Confounders in Rodent Blood Oxygen Level Dependent Functional Magnetic Resonance Imaging (BOLD fMRI)-Part A: Effects of Changes in Physiological Parameters. Front Neurosci 2020; 14:577119. [PMID: 33192261 PMCID: PMC7646331 DOI: 10.3389/fnins.2020.577119] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Accepted: 09/10/2020] [Indexed: 12/09/2022] Open
Abstract
Background: To understand brain function in health and disease, functional magnetic resonance imaging (fMRI) is widely used in rodent models. Because animals need to be immobilised for image acquisition, fMRI is commonly performed under anaesthesia. The choice of anaesthetic protocols and may affect fMRI readouts, either directly or via changing physiological balance, and thereby threaten the scientific validity of fMRI in rodents. Methods: The present study systematically reviewed the literature investigating the influence of different anaesthesia regimes and changes in physiological parameters as confounders of blood oxygen level dependent (BOLD) fMRI in rats and mice. Four databases were searched, studies selected according to pre-defined criteria, and risk of bias assessed for each study. Results are reported in two separate articles; this part of the review focuses on effects of changes in physiological parameters. Results: A total of 121 publications was included, of which 49 addressed effects of changes in physiological parameters. Risk of bias was high in all included studies. Blood oxygenation [arterial partial pressure of oxygen (paO2)], ventilation [arterial partial pressure of carbon dioxide (paCO2)] and arterial blood pressure affected BOLD fMRI readouts across various experimental paradigms. Conclusions: Blood oxygenation, ventilation and arterial blood pressure should be monitored and maintained at stable physiological levels throughout experiments. Appropriate anaesthetic management and monitoring are crucial to obtain scientifically valid, reproducible results from fMRI studies in rodent models.
Collapse
Affiliation(s)
- Aline R. Steiner
- Section of Anaesthesiology, Department of Clinical and Diagnostic Services, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Frédérik Rousseau-Blass
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Aileen Schroeter
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Sonja Hartnack
- Section of Epidemiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Regula Bettschart-Wolfensberger
- Section of Anaesthesiology, Department of Clinical and Diagnostic Services, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Joseph V, Laouafa S, Marcouiller F, Roussel D, Pialoux V, Bairam A. Progesterone decreases apnoea and reduces oxidative stress induced by chronic intermittent hypoxia in ovariectomized female rats. Exp Physiol 2020; 105:1025-1034. [PMID: 32196792 DOI: 10.1113/ep088430] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/12/2020] [Indexed: 12/15/2022]
Abstract
NEW FINDINGS What is the central question of this study? Does progesterone reduce the effect of chronic intermittent hypoxia (CIH) on arterial blood pressure, respiratory control and oxidative stress in the central nervous system in ovariectomized rats? What is the main finding and its importance? Progesterone does not prevent the elevation of arterial blood pressure in rats exposed to CIH, but normalizes respiratory control, and reduces cerebral oxidative stress. This study draws focus to a potential role of progesterone and the consequences of sleep apnoea in menopausal women. ABSTRACT We tested the hypothesis that progesterone (Prog) reduces the effect of chronic intermittent hypoxia (CIH) on arterial blood pressure, respiratory chemoreflexes and oxidative stress in the central nervous system. Ovariectomized female rats were implanted with osmotic pumps delivering vehicle (Veh) or Prog (4 mg kg-1 day-1 ). Two weeks following the surgery, rats were exposed to room air (Air) or CIH (7 days, 10% O2 , 10 cycles h-1 , 8 h day-1 ). We studied three groups: Veh-Air, Veh-CIH and Prog-CIH. After the CIH exposures, we measured the mean arterial pressure (MAP; tail cuff) and assessed the frequency of apnoeas at rest and ventilatory responses to hypoxia and hypercapnia (whole body plethysmography). The activities of the pro-oxidant enzyme NADPH oxidase (NOX) and antioxidant enzymes superoxide dismutase (SOD; in mitochondrial and cytosolic fractions) and glutathione peroxidase (GPx), as well as the concentration of malondialdehyde (MDA), a marker of lipid peroxidation, were measured in brain cortex and brainstem samples. CIH exposure increased the MAP, the frequency of apnoeas, and the respiratory frequency response to hypoxia and hypercapnia. Prog did not prevent the CIH-induced elevation in MAP, but it reduced the CIH-induced frequency of apnoeas and increased hypoxic and hypercapnic ventilatory responses. In the brain cortex, CIH increased NOX activity, and decreased the cytosolic and mitochondrial SOD activities. These effects were prevented by Prog. NOX activity was increased by CIH in the brainstem, and this was also blocked by Prog. The study draws focus to the links between ovarian hormones and the consequences of sleep apnoea in women.
Collapse
Affiliation(s)
- Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Sofien Laouafa
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada.,University of Lyon, Université Claude Bernard Lyon 1, CNRS, ENTPE, UMR5023 LEHNA, Villeurbanne, F-69622, France
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Damien Roussel
- University of Lyon, Université Claude Bernard Lyon 1, CNRS, ENTPE, UMR5023 LEHNA, Villeurbanne, F-69622, France
| | - Vincent Pialoux
- University of Lyon, Université Claude Bernard Lyon 1, LIBM EA 7424, Villeurbanne, 69622, France.,Institut Universitaire de France, Paris, France
| | - Aida Bairam
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
41
|
Abstract
Air-breathing animals do not experience hyperoxia (inspired O2 > 21%) in nature, but preterm and full-term infants often experience hyperoxia/hyperoxemia in clinical settings. This article focuses on the effects of normobaric hyperoxia during the perinatal period on breathing in humans and other mammals, with an emphasis on the neural control of breathing during hyperoxia, after return to normoxia, and in response to subsequent hypoxic and hypercapnic challenges. Acute hyperoxia typically evokes an immediate ventilatory depression that is often, but not always, followed by hyperpnea. The hypoxic ventilatory response (HVR) is enhanced by brief periods of hyperoxia in adult mammals, but the limited data available suggest that this may not be the case for newborns. Chronic exposure to mild-to-moderate levels of hyperoxia (e.g., 30-60% O2 for several days to a few weeks) elicits several changes in breathing in nonhuman animals, some of which are unique to perinatal exposures (i.e., developmental plasticity). Examples of this developmental plasticity include hypoventilation after return to normoxia and long-lasting attenuation of the HVR. Although both peripheral and CNS mechanisms are implicated in hyperoxia-induced plasticity, it is particularly clear that perinatal hyperoxia affects carotid body development. Some of these effects may be transient (e.g., decreased O2 sensitivity of carotid body glomus cells) while others may be permanent (e.g., carotid body hypoplasia, loss of chemoafferent neurons). Whether the hyperoxic exposures routinely experienced by human infants in clinical settings are sufficient to alter respiratory control development remains an open question and requires further research. © 2020 American Physiological Society. Compr Physiol 10:597-636, 2020.
Collapse
Affiliation(s)
- Ryan W Bavis
- Department of Biology, Bates College, Lewiston, Maine, USA
| |
Collapse
|
42
|
Maternal physical activity prevents the overexpression of hypoxia-inducible factor 1-α and cardiorespiratory dysfunction in protein malnourished rats. Sci Rep 2019; 9:14406. [PMID: 31594995 PMCID: PMC6783408 DOI: 10.1038/s41598-019-50967-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022] Open
Abstract
Maternal physical activity attenuates cardiorespiratory dysfunctions and transcriptional alterations presented by the carotid body (CB) of rats. Rats performed physical activity and were classified as inactive/active. During gestation and lactation, mothers received either normoprotein (NP-17% protein) or low-protein diet (LP-8% protein). In offspring, biochemical serum levels, respiratory parameters, cardiovascular parameters and the mRNA expression of hypoxia-inducible factor 1-alpha (HIF-1α), tyrosine hydroxylase (TH) and purinergic receptors were evaluate. LP-inactive pups presented lower RF from 1st to 14th days old, and higher RF at 30 days than did NP-inactive and NP-active pups. LP-inactive pups presented with reduced serum protein, albumin, cholesterol and triglycerides levels and an increased fasting glucose level compared to those of NP-inactive and NP-active groups. LP and LP-inactive animals showed an increase in the cardiac variability at the Low-Frequency bands, suggesting a major influence of sympathetic nervous activity. In mRNA analyses, LP-inactive animals showed increased HIF-1α expression and similar expression of TH and purinergic receptors in the CB compared to those of NP groups. All these changes observed in LP-inactive pups were reversed in the pups of active mothers (LP-active). Maternal physical activity is able to attenuate the metabolic, cardiorespiratory and HIF-1α transcription changes induced by protein malnutrition.
Collapse
|
43
|
Christensson E, Ebberyd A, Hårdemark Cedborg A, Lodenius Å, Österlund Modalen Å, Franklin KA, Eriksson LI, Jonsson Fagerlund M. Hypoxic ventilatory response after rocuronium‐induced partial neuromuscular blockade in men with obstructive sleep apnoea. Anaesthesia 2019; 75:338-347. [DOI: 10.1111/anae.14806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2019] [Indexed: 12/12/2022]
Affiliation(s)
- E. Christensson
- Function Peri‐operative Medicine and Intensive Care Karolinska University Hospital Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
- Department of Physiology and Pharmacology Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
| | - A. Ebberyd
- Department of Physiology and Pharmacology Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
| | - A. Hårdemark Cedborg
- Department of Physiology and Pharmacology Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
| | - Å. Lodenius
- Department of Physiology and Pharmacology Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
| | | | - K. A. Franklin
- Department of Surgical and Perioperative Sciences, Surgery Umeå University Umeå Sweden
| | - L. I. Eriksson
- Function Peri‐operative Medicine and Intensive Care Karolinska University Hospital Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
- Department of Physiology and Pharmacology Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
| | - M. Jonsson Fagerlund
- Function Peri‐operative Medicine and Intensive Care Karolinska University Hospital Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
- Department of Physiology and Pharmacology Section for Anaesthesiology and Intensive Care Medicine Karolinska Institute StockholmSweden
| |
Collapse
|
44
|
Laouafa S, Iturri P, Arias-Reyes C, Marcouiller F, Gonzales M, Joseph V, Bairam A, Soliz J. Erythropoietin and caffeine exert similar protective impact against neonatal intermittent hypoxia: Apnea of prematurity and sex dimorphism. Exp Neurol 2019; 320:112985. [PMID: 31254520 DOI: 10.1016/j.expneurol.2019.112985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 05/21/2019] [Accepted: 06/25/2019] [Indexed: 12/30/2022]
Abstract
Apnea of prematurity (AoP) is associated with severe and repeated episodes of arterial oxygen desaturation (intermittent hypoxia - IH), which in turn increases the number of apneas. So far, there is no data addressing whether IH leads to sex-specific respiratory consequences, neither if drugs targeting AoP are more effective in males or females. We used rat pups for investigating whether IH-mediated increase of apneas is sex-specific. We also tested whether caffeine (treatment of choice of AoP), erythropoietin (Epo - a neuroprotective factor and potent respiratory stimulant), and combination of both (caffeine+Epo) prevent the IH-mediated formation of apneas in a sex-dependent manner. Newborn rats exposed to IH (21% - 10% FIO2-8 h a day - 10 cycles per hour) during postnatal days (P) 3-10 were used in this work. Animals were administered drug vehicle, Epo, caffeine and Epo + caffeine (daily from P3 to P10) gavage. At P10 the frequency of apneas at rest (as an index of respiratory dysfunction induced by IH), and respiratory parameters were measured by plethysmography. Our results showed that IH significantly increases the number of apneas in male but not in female rat pups. Moreover, caffeine and Epo in males similarly prevented the increase of apneas induced by IH, and the administration of both drugs together did not provide a cumulative beneficial effect. No impact of drugs was evidenced in females. Apart from apneas, IH increased the normoxic basal ventilation (ventilation at rest) of male animals, and treatments did not prevent such alteration. Besides, no IH- nor treatment-mediated modulation of basal ventilation was found in the basal ventilation of female animals. Analysis of the activity of pro- and antioxidative molecules revealed that IH induces oxidative stress in the brainstem of male and female animals and that all tested treatments similarly prevented such oxidative imbalance in pups of both sexes. We concluded that neonatal IH and the treatments tested to prevent its respiratory consequences are sex-specific. The mechanics associated with such prevention are directly linked with the prevention of oxidative stress and the maturation of the brain. These findings are relevant to understanding better the AoP disorder and for proposing Epo as a new therapeutical tool.
Collapse
Affiliation(s)
- Sofien Laouafa
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Pablo Iturri
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Christian Arias-Reyes
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada; Departamento de Biologia Celular y Molecular, Facultad de Ciencias Puras y Naturales, Universidad Mayor de San Andres, La Paz, Bolivia
| | - François Marcouiller
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Marcelino Gonzales
- Instituto Boliviano de Biologia de la Altura, Facultad de Medicina, Universidad Mayor de San Andres, La Paz, Bolivia
| | - Vincent Joseph
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Aida Bairam
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada
| | - Jorge Soliz
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Québec City, Québec, Canada; Departamento de Biologia Celular y Molecular, Facultad de Ciencias Puras y Naturales, Universidad Mayor de San Andres, La Paz, Bolivia.
| |
Collapse
|
45
|
Shell B, Farmer GE, Nedungadi TP, Wang LA, Marciante AB, Snyder B, Cunningham RL, Cunningham JT. Angiotensin type 1a receptors in the median preoptic nucleus support intermittent hypoxia-induced hypertension. Am J Physiol Regul Integr Comp Physiol 2019; 316:R651-R665. [PMID: 30892911 PMCID: PMC6589598 DOI: 10.1152/ajpregu.00393.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/25/2019] [Accepted: 03/13/2019] [Indexed: 01/19/2023]
Abstract
Chronic intermittent hypoxia (CIH) is a model of the hypoxemia from sleep apnea that causes a sustained increase in blood pressure. Inhibition of the central renin-angiotensin system or FosB in the median preoptic nucleus (MnPO) prevents the sustained hypertensive response to CIH. We tested the hypothesis that angiotensin type 1a (AT1a) receptors in the MnPO, which are upregulated by CIH, contribute to this hypertension. In preliminary experiments, retrograde tract tracing studies showed AT1a receptor expression in MnPO neurons projecting to the paraventricular nucleus. Adult male rats were exposed to 7 days of intermittent hypoxia (cycling between 21% and 10% O2 every 6 min, 8 h/day during light phase). Seven days of CIH was associated with a FosB-dependent increase in AT1a receptor mRNA without changes in the permeability of the blood-brain barrier in the MnPO. Separate groups of rats were injected in the MnPO with an adeno-associated virus containing short hairpin (sh)RNA against AT1a receptors to test their role in intermittent hypoxia hypertension. Injections of shRNA against AT1a in MnPO blocked the increase in mRNA associated with CIH, prevented the sustained component of the hypertension during normoxia, and reduced circulating advanced oxidation protein products, an indicator of oxidative stress. Rats injected with shRNA against AT1a and exposed to CIH had less FosB staining in MnPO and the rostral ventrolateral medulla after intermittent hypoxia than rats injected with the control vector that were exposed to CIH. Our results indicate AT1a receptors in the MnPO contribute to the sustained blood pressure increase to intermittent hypoxia.
Collapse
MESH Headings
- Angiotensin II/administration & dosage
- Animals
- Blood Pressure/drug effects
- Disease Models, Animal
- Hypertension/etiology
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Hypoxia/complications
- Hypoxia/genetics
- Hypoxia/metabolism
- Hypoxia/physiopathology
- Injections, Intraventricular
- Male
- Oxidative Stress
- Preoptic Area/drug effects
- Preoptic Area/metabolism
- Preoptic Area/physiopathology
- Proto-Oncogene Proteins c-fos/genetics
- Proto-Oncogene Proteins c-fos/metabolism
- RNA Interference
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats, Sprague-Dawley
- Receptor, Angiotensin, Type 1/agonists
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Signal Transduction
- Up-Regulation
Collapse
Affiliation(s)
- Brent Shell
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - T Prashant Nedungadi
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Lei A Wang
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Alexandria B Marciante
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Brina Snyder
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Rebecca L Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
46
|
Li J, Yang S, Yu F, Ji E, Woodrow Weiss J. Endothelin-1 enhanced carotid body chemosensory activity in chronic intermittent hypoxia through PLC, PKC and p38MAPK signaling pathways. Neuropeptides 2019; 74:44-51. [PMID: 30579678 DOI: 10.1016/j.npep.2018.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/24/2018] [Accepted: 12/17/2018] [Indexed: 10/27/2022]
Abstract
Endothelin-1 (ET-1), as it functions as a neuromodulator, has been associated with hypertension in chronic intermittent hypoxia (CIH) which attribute to enhanced carotid body sensibility to hypoxia. However, the molecular mechanism of ET-1 on carotid body sensibility in CIH is still not clear. Here, effect of ET-1 on carotid body chemosensory stimulation in rats exposed to either CIH or room air (Normoxia) was explored. Furthermore, Phospholipase C (PLC), Protein kinase C (PKC) or p38 MAPK antagonists were adopted to clarify the signalling pathways involved. Results showed that ET-1 induced a higher increase of carotid sinus nerve activity (CSNA) in animals exposed to CIH. Both ETA and ETB receptor expression were up-regulated by CIH exposure, but only ETA is responsible for ET-1 induced CSNA increase. Additional, the increase was inhibited by PLC, PKC, p38 MAPK antagonists and calcium channel blocker. Our findings support that ETA receptor mediates ET-1-induced CSNA increase through PLC, PKC and p38 MAPK signalling pathways in chronic intermittent hypoxia. Also, our study indicated that calcium influx was necessary for enhancing effect of ET-1 on CSNA.
Collapse
Affiliation(s)
- Jieru Li
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Shengchang Yang
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - Fuyang Yu
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China
| | - EnSheng Ji
- Department of Physiology, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People's Republic of China.
| | - J Woodrow Weiss
- Division of Pulmonary, Critical Care & Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
47
|
Lindsey BG, Nuding SC, Segers LS, Morris KF. Carotid Bodies and the Integrated Cardiorespiratory Response to Hypoxia. Physiology (Bethesda) 2019; 33:281-297. [PMID: 29897299 DOI: 10.1152/physiol.00014.2018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Advances in our understanding of brain mechanisms for the hypoxic ventilatory response, coordinated changes in blood pressure, and the long-term consequences of chronic intermittent hypoxia as in sleep apnea, such as hypertension and heart failure, are giving impetus to the search for therapies to "erase" dysfunctional memories distributed in the carotid bodies and central nervous system. We review current network models, open questions, sex differences, and implications for translational research.
Collapse
Affiliation(s)
- Bruce G Lindsey
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Sarah C Nuding
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Lauren S Segers
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida , Tampa, Florida
| | - Kendall F Morris
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida , Tampa, Florida
| |
Collapse
|
48
|
Endothelin-1-Mediated Mechanisms in the Carotid Body Modulates Cardiovascular Responses in Rats Exposed to Chronic Intermittent Hypoxia. Int J Pept Res Ther 2018. [DOI: 10.1007/s10989-018-9794-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
49
|
Leonard EM, Zhang M, Nurse CA. Evidence for protein kinase involvement in the 5-HT-[Ca 2+ ] i -pannexin-1 signalling pathway in type II glial cells of the rat carotid body. Exp Physiol 2018; 104:244-253. [PMID: 30456914 DOI: 10.1113/ep087411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/19/2018] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? The mammalian carotid body (CB) is a peripheral chemoreceptor organ involved in O2 and CO2 /H+ homeostasis. Recent studies suggest that 5-HT, released from CB receptor cells, can stimulate adjacent glial-like type II cells, leading to an increase in intracellular Ca2+ (Δ[Ca2+ ]i ) and activation of ATP-permeable pannexin-1 (Panx-1) channels. The aim of this study was to elucidate the role of protein kinases in the 5-HT-[Ca2+ ]i -Panx-1 signalling pathway. What is the main finding and its importance? Src family kinase and protein kinase A, acting downstream from Δ[Ca2+ ]i , played central roles in 5-HT-mediated Panx-1 channel activation. This provides new insight into mechanisms regulating CB excitation, especially in pathophysiological conditions. ABSTRACT Chemoreceptor (type I) cells of the rodent carotid body (CB) synthesize and release several neurotransmitters/neuromodulators, including 5-hydroxytryptamine (5-HT), implicated in enhanced CB excitation after exposure to chronic intermittent hypoxia, e.g. sleep apnoea. However, recent studies suggest that 5-HT can robustly stimulate adjacent glial-like type II cells via ketanserin-sensitive 5-HT2 receptors, leading to intracellular Ca2+ elevation (Δ[Ca2+ ]i ) and activation of ATP-permeable pannexin-1 (Panx-1) channels. Using dissociated rat CB cultures, we investigated the role of protein kinases in the intracellular signalling pathways in type II cells. In isolated type II cells, 5-HT activated a Panx-1-like inward current (I5-HT ) that was reversibly inhibited by the Src family kinase inhibitor PP2 (1 μm), but not by its inactive analogue, PP3 (1 μm). Moreover, I5-HT was reversibly inhibited (>90%) by H89 (1 μm), a protein kinase A blocker, whereas the protein kinase C blocker GF109203X (2 μm) was largely ineffective. In contrast, the P2Y2R agonist UTP (100 μm) activated Panx-1-like currents that were reversibly inhibited (∼60%) by either H89 or GF109203X. Using fura-2 spectrofluorimetry, the 5-HT-induced Δ[Ca2+ ]i was unaffected by PP2, H89 and GF109293X, suggesting that the kinases acted downstream of the Ca2+ rise. Given that intracellular Ca2+ chelation was previously shown to block receptor-mediated Panx-1 current activation in type II cells, these data suggest that CB neuromodulators use overlapping, but not necessarily identical, signalling pathways to activate Panx-1 channels and release ATP, a CB excitatory neurotransmitter. In conclusion, these studies provide new mechanistic insight into 5-HT signalling in the CB that has pathophysiological relevance.
Collapse
Affiliation(s)
- Erin M Leonard
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Min Zhang
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Colin A Nurse
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
50
|
Lucking EF, O'Connor KM, Strain CR, Fouhy F, Bastiaanssen TFS, Burns DP, Golubeva AV, Stanton C, Clarke G, Cryan JF, O'Halloran KD. Chronic intermittent hypoxia disrupts cardiorespiratory homeostasis and gut microbiota composition in adult male guinea-pigs. EBioMedicine 2018; 38:191-205. [PMID: 30446434 PMCID: PMC6306383 DOI: 10.1016/j.ebiom.2018.11.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/26/2018] [Accepted: 11/05/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Carotid body (peripheral oxygen sensor) sensitisation is pivotal in the development of chronic intermittent hypoxia (CIH)-induced hypertension. We sought to determine if exposure to CIH, modelling human sleep apnoea, adversely affects cardiorespiratory control in guinea-pigs, a species with hypoxia-insensitive carotid bodies. We reasoned that CIH-induced disruption of gut microbiota would evoke cardiorespiratory morbidity. METHODS Adult male guinea-pigs were exposed to CIH (6.5% O2 at nadir, 6 cycles.hour-1) for 8 h.day-1 for 12 consecutive days. FINDINGS CIH-exposed animals established reduced faecal microbiota species richness, with increased relative abundance of Bacteroidetes and reduced relative abundance of Firmicutes bacteria. Urinary corticosterone and noradrenaline levels were unchanged in CIH-exposed animals, but brainstem noradrenaline concentrations were lower compared with sham. Baseline ventilation was equivalent in CIH-exposed and sham animals; however, respiratory timing variability, sigh frequency and ventilation during hypoxic breathing were all lower in CIH-exposed animals. Baseline arterial blood pressure was unaffected by exposure to CIH, but β-adrenoceptor-dependent tachycardia and blunted bradycardia during phenylephrine-induced pressor responses was evident compared with sham controls. INTERPRETATION Increased carotid body chemo-afferent signalling appears obligatory for the development of CIH-induced hypertension and elevated chemoreflex control of breathing commonly reported in mammals, with hypoxia-sensitive carotid bodies. However, we reveal that exposure to modest CIH alters gut microbiota richness and composition, brainstem neurochemistry, and autonomic control of heart rate, independent of carotid body sensitisation, suggesting modulation of breathing and autonomic homeostasis via the microbiota-gut-brainstem axis. The findings have relevance to human sleep-disordered breathing. FUNDING The Department of Physiology, and APC Microbiome Ireland, UCC.
Collapse
Affiliation(s)
- Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Conall R Strain
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Fiona Fouhy
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Anna V Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|