1
|
Polecka A, Olszewska N, Pulido M, Olszewska E. Rostral fluid shifts and other mechanisms of interaction between obstructive sleep apnea and heart failure - a systematic review. OTOLARYNGOLOGIA POLSKA 2024; 78:10-17. [PMID: 39417262 DOI: 10.5604/01.3001.0054.6742] [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: 10/19/2024]
Abstract
<b>Introduction:</b> Obstructive sleep apnea (OSA) is a chronic inflammatory disorder characterized by episodes of total or partial upper airway obstruction during sleep. Untreated OSA leads to various cardiovascular complications, including heart failure (HF), both involving complex and detrimental pathophysiological processes.<b>Aim:</b> The aim of this study is to describe the role of rostral fluid shifts and other mechanisms responsible for the co-existence of OSA and HF, providing insight into potential diagnostic and therapeutic strategies.<b>Materials and methods:</b> Two authors independently searched the literature and assessed articles following PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analytics) guidelines.<b>Results:</b> Rostral fluid shifts, characterized by nocturnal redistribution from the lower limbs to the neck tissues, exacerbate upper airway obstruction by increasing neck circumference and predisposing individuals to respiratory events. This phenomenon is particularly significant in patients with HF due to impaired cardiovascular function leading to fluid retention. The repetitive collapse of the upper airway during sleep triggers abrupt changes in intrathoracic pressure negatively impacting cardiac tissue remodeling by promoting inflammation and fibrosis. Moreover, sleep fragmentation and arousals activate the sympathetic nervous system (SNS), imposing additional strain on the cardiovascular system. Accumulated data suggest that rostral fluid shifts are a clinically significant pathomechanism in the coexistence of OSA and HF. Therapeutic strategies, including the benefits of continuous positive airway pressure (CPAP) therapy and lifestyle modifications, have been discussed. This systematic review highlights the need for integrated treatment approaches to manage both OSA and HF effectively.<b>Conclusions:</b> Understanding and addressing these interconnected mechanisms is essential to offer an integrated diagnostic and therapeutic management of patients, highlighting the importance of multidisciplinary care to optimize patient health and quality of life.
Collapse
Affiliation(s)
- Agnieszka Polecka
- Doctoral School of Medical University of Bialystok, Department of Cardiology and Internal Medicine with Cardiac Intensive Care Unit, University Clinical Hospital in Bialystok, Poland
| | - Natalia Olszewska
- Student Research Group, Department of Otolaryngology, Medical University of Bialystok, Poland
| | - Maria Pulido
- Whipps Cross&Royal London Hospital, Barts Health NHS Trust, United Kingdom
| | - Ewa Olszewska
- Sleep Apnea Surgery Center, Department of Otolaryngology, Medical University of Bialystok, Poland
| |
Collapse
|
2
|
Huff AD, Karlen-Amarante M, Oliveira LM, Ramirez JM. Chronic intermittent hypoxia reveals role of the Postinspiratory Complex in the mediation of normal swallow production. eLife 2024; 12:RP92175. [PMID: 38655918 PMCID: PMC11042803 DOI: 10.7554/elife.92175] [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] [Indexed: 04/26/2024] Open
Abstract
Obstructive sleep apnea (OSA) is a prevalent sleep-related breathing disorder that results in multiple bouts of intermittent hypoxia. OSA has many neurological and systemic comorbidities, including dysphagia, or disordered swallow, and discoordination with breathing. However, the mechanism in which chronic intermittent hypoxia (CIH) causes dysphagia is unknown. Recently, we showed the postinspiratory complex (PiCo) acts as an interface between the swallow pattern generator (SPG) and the inspiratory rhythm generator, the preBötzinger complex, to regulate proper swallow-breathing coordination (Huff et al., 2023). PiCo is characterized by interneurons co-expressing transporters for glutamate (Vglut2) and acetylcholine (ChAT). Here we show that optogenetic stimulation of ChATcre:Ai32, Vglut2cre:Ai32, and ChATcre:Vglut2FlpO:ChR2 mice exposed to CIH does not alter swallow-breathing coordination, but unexpectedly disrupts swallow behavior via triggering variable swallow motor patterns. This suggests that glutamatergic-cholinergic neurons in PiCo are not only critical for the regulation of swallow-breathing coordination, but also play an important role in the modulation of swallow motor patterning. Our study also suggests that swallow disruption, as seen in OSA, involves central nervous mechanisms interfering with swallow motor patterning and laryngeal activation. These findings are crucial for understanding the mechanisms underlying dysphagia, both in OSA and other breathing and neurological disorders.
Collapse
Affiliation(s)
- Alyssa D Huff
- Center for Integrative Brain Research, Seattle Children’s Research InstituteSeattleUnited States
| | - Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children’s Research InstituteSeattleUnited States
| | - Luiz M Oliveira
- Center for Integrative Brain Research, Seattle Children’s Research InstituteSeattleUnited States
| | - Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children’s Research InstituteSeattleUnited States
- Department of Neurological Surgery, University of Washington School of MedicineSeattleUnited States
| |
Collapse
|
3
|
Huff A, Karlen-Amarante M, Oliveira LM, Ramirez JM. Chronic Intermittent Hypoxia reveals role of the Postinspiratory Complex in the mediation of normal swallow production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.26.559560. [PMID: 37808787 PMCID: PMC10557756 DOI: 10.1101/2023.09.26.559560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Obstructive sleep apnea (OSA) is a prevalent sleep-related breathing disorder that results in multiple bouts of intermittent hypoxia. OSA has many neurologic and systemic comorbidities including dysphagia, or disordered swallow, and discoordination with breathing. However, the mechanism in which chronic intermittent hypoxia (CIH) causes dysphagia is unknown. Recently we showed the Postinspiratory complex (PiCo) acts as an interface between the swallow pattern generator (SPG) and the inspiratory rhythm generator, the preBötzinger Complex, to regulate proper swallow-breathing coordination (Huff et al., 2023). PiCo is characterized by interneurons co-expressing transporters for glutamate (Vglut2) and acetylcholine (ChAT). Here we show that optogenetic stimulation of ChATcre:Ai32, Vglut2cre:Ai32, and ChATcre:Vglut2FlpO:ChR2 mice exposed to CIH does not alter swallow-breathing coordination, but unexpectedly disrupts swallow behavior via triggering variable swallow motor patterns. This suggests, glutamatergic-cholinergic neurons in PiCo are not only critical for the regulation of swallow-breathing coordination, but also play an important role in the modulation of swallow motor patterning. Our study also suggests that swallow disruption, as seen in OSA, involves central nervous mechanisms interfering with swallow motor patterning and laryngeal activation. These findings are crucial for understanding the mechanisms underlying dysphagia, both in OSA and other breathing and neurological disorders.
Collapse
Affiliation(s)
- Alyssa Huff
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
| | - Marlusa Karlen-Amarante
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
| | - Luiz Marcelo Oliveira
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
| | - Jan Marino Ramirez
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, WA, 98101
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, WA, USA, 98108
| |
Collapse
|
4
|
Chen J, Bendowski KT, Bizanti A, Zhang Y, Ma J, Hoover DB, Gozal D, Shivkumar K, Cheng ZJ. Distribution and morphology of calcitonin gene-related peptide (CGRP) innervation in flat mounts of whole rat atria and ventricles. Auton Neurosci 2024; 251:103127. [PMID: 38211380 PMCID: PMC11639590 DOI: 10.1016/j.autneu.2023.103127] [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: 09/18/2023] [Revised: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 01/13/2024]
Abstract
Calcitonin gene-related peptide (CGRP) is widely used as a marker for nociceptive afferent axons. However, the distribution of CGRP-IR axons has not been fully determined in the whole rat heart. Immunohistochemically labeled flat-mounts of the right and left atria and ventricles, and the interventricular septum (IVS) in rats for CGRP were assessed with a Zeiss imager to generate complete montages of the entire atria, ventricles, and septum, and a confocal microscope was used to acquire detailed images of selected regions. We found that 1) CGRP-IR axons extensively innervated all regions of the atrial walls including the sinoatrial node region, auricles, atrioventricular node region, superior/inferior vena cava, left pre-caval vein, and pulmonary veins. 2) CGRP-IR axons formed varicose terminals around individual neurons in some cardiac ganglia but passed through other ganglia without making appositions with cardiac neurons. 3) Varicose CGRP-IR axons innervated the walls of blood vessels. 4) CGRP-IR axons extensively innervated the right/left ventricular walls and IVS. Our data shows the rather ubiquitous distribution of CGRP-IR axons in the whole rat heart at single-cell/axon/varicosity resolution for the first time. This study lays the foundation for future studies to quantify the differences in CGRP-IR axon innervation between sexes, disease models, and species.
Collapse
Affiliation(s)
- Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| | - Kohlton T Bendowski
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| | - Ariege Bizanti
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Yuanyuan Zhang
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Jichao Ma
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA
| | - Donald B Hoover
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - David Gozal
- Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, USA
| | - Kalyanam Shivkumar
- Department of Medicine, Cardiac Arrhythmia Center and Neurocardiology Research Program of Excellence, University of California, Los Angeles, CA 90095, USA
| | - Zixi Jack Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816, USA.
| |
Collapse
|
5
|
Marchini T, Magnani N, Garces M, Kelly J, Paz M, Caceres L, Calabro V, Lasagni Vitar R, Caltana L, Contin M, Reynoso S, Lago N, Vico T, Vanasco V, Wolf D, Tripodi V, Gonzalez Maglio D, Alvarez S, Buchholz B, Berra A, Gelpi R, Evelson P. Chronic exposure to polluted urban air aggravates myocardial infarction by impaired cardiac mitochondrial function and dynamics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 295:118677. [PMID: 34906594 DOI: 10.1016/j.envpol.2021.118677] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 11/05/2021] [Accepted: 12/10/2021] [Indexed: 06/14/2023]
Abstract
Air pollution exposure positively correlates with increased cardiovascular morbidity and mortality rates, mainly due to myocardial infarction (MI). Herein, we aimed to study the metabolic mechanisms underlying this association, focusing on the evaluation of cardiac mitochondrial function and dynamics, together with its impact over MI progression. An initial time course study was performed in BALB/c mice breathing filtered air (FA) or urban air (UA) in whole-body exposure chambers located in Buenos Aires City downtown for up to 16 weeks (n = 8 per group and time point). After 12 weeks, lung inflammatory cell recruitment was evident in UA-exposed mice. Interestingly, impaired redox metabolism, characterized by decreased lung SOD activity and increased GSSG levels and NOX activity, precede local inflammation in this group. At this selected time point, additional mice were exposed to FA or UA (n = 12 per group) and alveolar macrophage PM uptake and nitric oxide (NO) production was observed in UA-exposed mice, together with increased pro-inflammatory cytokine levels (TNF-α and IL-6) in BAL and plasma. Consequently, impaired heart tissue oxygen metabolism and altered mitochondrial ultrastructure and function were observed in UA-exposed mice after 12 weeks, characterized by decreased active state respiration and ATP production rates, and enhanced mitochondrial H2O2 production. Moreover, disturbed cardiac mitochondrial dynamics was detected in this group. This scenario led to a significant increase in the area of infarcted tissue following myocardial ischemia reperfusion injury in vivo, from 43 ± 3% of the area at risk in mice breathing FA to 66 ± 4% in UA-exposed mice (n = 6 per group, p < 0.01), together with a sustained increase in LVEDP during myocardial reperfusion. Taken together, our data unravel cardiac mitochondrial mechanisms that contribute to the understanding of the adverse health effects of urban air pollution exposure, and ultimately highlight the importance of considering environmental factors in the development of cardiovascular diseases.
Collapse
Affiliation(s)
- Timoteo Marchini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Mariana Garces
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Jazmin Kelly
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Mariela Paz
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, C1113AAD, Argentina
| | - Lourdes Caceres
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Valeria Calabro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Romina Lasagni Vitar
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Laura Caltana
- CONICET - Universidad de Buenos Aires, Instituto de Biología Celular y Neurociencias (IBCN), Buenos Aires, C1121ABG, Argentina
| | - Mario Contin
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, C1113AAD, Argentina
| | - Sofia Reynoso
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Nestor Lago
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Tamara Vico
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Virginia Vanasco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Dennis Wolf
- University Heart Center Freiburg-Bad Krozingen, Cardiology and Angiology I, Faculty of Medicine, University of Freiburg, Freiburg, 79106, Germany
| | - Valeria Tripodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, C1113AAD, Argentina
| | - Daniel Gonzalez Maglio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, C1113AAD, Argentina
| | - Silvia Alvarez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina
| | - Bruno Buchholz
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Alejandro Berra
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Ricardo Gelpi
- CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina; Universidad de Buenos Aires, Facultad de Medicina, Departamento de Patología, Buenos Aires, C1113AAD, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Buenos Aires, C1113AAD, Argentina; CONICET - Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, C1113AAD, Argentina.
| |
Collapse
|
6
|
Naryzhnaya NV, Maslov LN, Derkachev IA, Ma H, Zhang Y, Prasad NR, Singh N, Fu F, Pei JM, Sarybaev A, Sydykov A. The effect of adaptation to hypoxia on cardiac tolerance to ischemia/reperfusion. J Biomed Res 2022:1-25. [PMID: 37183617 PMCID: PMC10387748 DOI: 10.7555/jbr.36.20220125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The acute myocardial infarction (AMI) and sudden cardiac death (SCD), both associated with acute cardiac ischemia, are one of the leading causes of adult death in economically developed countries. The development of new approaches for the treatment and prevention of AMI and SCD remains the highest priority for medicine. A study on the cardiovascular effects of chronic hypoxia (CH) may contribute to the development of these methods. Chronic hypoxia exerts both positive and adverse effects. The positive effects are the infarct-reducing, vasoprotective, and antiarrhythmic effects, which can lead to the improvement of cardiac contractility in reperfusion. The adverse effects are pulmonary hypertension and right ventricular hypertrophy. This review presents a comprehensive overview of how CH enhances cardiac tolerance to ischemia/reperfusion. It is an in-depth analysis of the published data on the underlying mechanisms, which can lead to future development of the cardioprotective effect of CH. A better understanding of the CH-activated protective signaling pathways may contribute to new therapeutic approaches in an increase of cardiac tolerance to ischemia/reperfusion.
Collapse
|
7
|
Kay MW, Jain V, Panjrath G, Mendelowitz D. Targeting Parasympathetic Activity to Improve Autonomic Tone and Clinical Outcomes. Physiology (Bethesda) 2022; 37:39-45. [PMID: 34486396 PMCID: PMC8742722 DOI: 10.1152/physiol.00023.2021] [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] [Indexed: 01/03/2023] Open
Abstract
In this review we will briefly summarize the evidence that autonomic imbalance, more specifically reduced parasympathetic activity to the heart, generates and/or maintains many cardiorespiratory diseases and will discuss mechanisms and sites, from myocytes to the brain, that are potential translational targets for restoring parasympathetic activity and improving cardiorespiratory health.
Collapse
Affiliation(s)
- Matthew W. Kay
- 1Department of Biomedical Engineering, George Washington University, Washington, District of Columbia
| | - Vivek Jain
- 2Division of Pulmonary Medicine, Department of Medicine, George Washington University, Washington, District of Columbia
| | - Gurusher Panjrath
- 3Division of Cardiology, Department of Medicine, George Washington University, Washington, District of Columbia
| | - David Mendelowitz
- 4Department of Pharmacology and Physiology, George Washington University, Washington, District of Columbia
| |
Collapse
|
8
|
Agaltsov MV, Drapkina OM. Obstructive sleep apnea and cardiovascular comorbidity: common pathophysiological mechanisms to cardiovascular disease. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2021. [DOI: 10.20996/1819-6446-2021-08-05] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Obstructive sleep apnea (OSA) is associated with many cardiovascular and metabolic diseases. Sleep apnea causes intermittent hypoxemia, chest pressure fluctuations and a reaction from the cerebral cortex in the form of a short awakening during sleep (EEG-activation). The consequences of pathological pathways are studied in experimental models involving cell cultures, animals, and healthy volunteers. At present, the negative impact of intermittent hypoxemia on a variety of pathophysiological disorders of the heart and blood vessels (vascular tone fluctuations, thickening of the intimamedia complex in the vascular wall, direct damaging effect on the myocardium) has a great evidence base. Two other pathological components of OSA (pressure fluctuations and EEG-activation) can also affect cardiovascular system, mainly affecting the increase in blood pressure and changing cardiac hemodynamics. Although these reactions are considered separately in the review, with the development of sleep apnea they occur sequentially and are closely interrelated. As a result, these pathological pathways trigger further pathophysiological mechanisms acting on the heart and blood vessels. It is known that these include excessive sympathetic activation, inflammation, oxidative stress and metabolic dysregulation. In many respects being links of one process, these mechanisms can trigger damage to the vascular wall, contributing to the formation of atherosclerotic lesions. The accumulated data with varying degrees of reliability confirm the participation of OSA through these processes in the formation of cardiovascular disorders. There are factors limiting direct evidence of this interaction (sleep deprivation, causing similar changes, as well as the inability to share the contribution of other risk factors for cardiovascular diseases, in particular arterial hypertension, obesity, which are often associated with OSA). It is necessary to continue the study of processes that implement the pathological effect of OSA on the cardiovascular system.
Collapse
Affiliation(s)
- M. V. Agaltsov
- National Medical Research Center for Therapy and Preventive Medicine
| | - O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
9
|
Espinoza L, Fedorchak S, Boychuk CR. Interplay Between Systemic Metabolic Cues and Autonomic Output: Connecting Cardiometabolic Function and Parasympathetic Circuits. Front Physiol 2021; 12:624595. [PMID: 33776789 PMCID: PMC7991741 DOI: 10.3389/fphys.2021.624595] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/12/2021] [Indexed: 12/21/2022] Open
Abstract
There is consensus that the heart is innervated by both the parasympathetic and sympathetic nervous system. However, the role of the parasympathetic nervous system in controlling cardiac function has received significantly less attention than the sympathetic nervous system. New neuromodulatory strategies have renewed interest in the potential of parasympathetic (or vagal) motor output to treat cardiovascular disease and poor cardiac function. This renewed interest emphasizes a critical need to better understand how vagal motor output is generated and regulated. With clear clinical links between cardiovascular and metabolic diseases, addressing this gap in knowledge is undeniably critical to our understanding of the interaction between metabolic cues and vagal motor output, notwithstanding the classical role of the parasympathetic nervous system in regulating gastrointestinal function and energy homeostasis. For this reason, this review focuses on the central, vagal circuits involved in sensing metabolic state(s) and enacting vagal motor output to influence cardiac function. It will review our current understanding of brainstem vagal circuits and their unique position to integrate metabolic signaling into cardiac activity. This will include an overview of not only how metabolic cues alter vagal brainstem circuits, but also how vagal motor output might influence overall systemic concentrations of metabolic cues known to act on the cardiac tissue. Overall, this review proposes that the vagal brainstem circuits provide an integrative network capable of regulating and responding to metabolic cues to control cardiac function.
Collapse
Affiliation(s)
- Liliana Espinoza
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Stephanie Fedorchak
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| | - Carie R Boychuk
- Department of Cellular and Integrative Physiology, Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, United States
| |
Collapse
|
10
|
Chen J, Gu H, Wurster RD, Cheng ZJ. The protective role of SOD1 overexpression in central mediation of bradycardia following chronic intermittent hypoxia in mice. Am J Physiol Regul Integr Comp Physiol 2021; 320:R317-R330. [PMID: 33296277 PMCID: PMC7988771 DOI: 10.1152/ajpregu.00147.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/27/2020] [Accepted: 12/03/2020] [Indexed: 01/07/2023]
Abstract
Obstructive sleep apnea (OSA) is a highly prevalent sleep disorder that is associated with many cardiovascular complications. Similar to OSA, chronic intermittent hypoxia (CIH) (a model for OSA) leads to oxidative stress and impairs baroreflex control of the heart rate (HR) in rodents. The baroreflex arc includes the aortic depressor nerve (ADN), vagal efferent, and central neurons. In this study, we used mice as a model to examine the effects of CIH on baroreflex sensitivity, aortic baroreceptor afferents, and central and vagal efferent components of the baroreflex circuitry. Furthermore, we tested whether human Cu/Zn Superoxide Dismutase (SOD1) overexpression in transgenic mice offers protection against CIH-induced deficit of the baroreflex arc. Wild-type C57BL/6J and SOD1 mice were exposed to room air (RA) or CIH and were then anesthetized, ventilated, and catheterized for measurement of mean arterial pressure (MAP) and HR. Compared with wild-type RA control, CIH impaired baroreflex sensitivity but increased maximum baroreceptor gain and bradycardic response to vagal efferent stimulation. Additionally, CIH reduced the bradycardic response to ADN stimulation, indicating a diminished central regulation of bradycardia. Interestingly, SOD1 overexpression prevented CIH-induced attenuation of HR responses to ADN stimulation and preserved HR responses to vagal efferent stimulation in transgenic mice. We suggest that CIH decreased central mediation of the baroreflex and SOD1 overexpression may prevent the CIH-induced central deficit.
Collapse
Affiliation(s)
- Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - He Gu
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| | - Robert D Wurster
- Department of Cellular and Molecular Physiology, Stritch School of Medicine, Loyola University, Maywood, Illinois
| | - Zixi Jack Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida
| |
Collapse
|
11
|
Rodrigues KL, Souza JR, Bazilio DS, de Oliveira M, Moraes MPS, Moraes DJA, Machado BH. Changes in the autonomic and respiratory patterns in mice submitted to short-term sustained hypoxia. Exp Physiol 2021; 106:759-770. [PMID: 33501717 DOI: 10.1113/ep089323] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 01/20/2021] [Indexed: 12/22/2022]
Abstract
NEW FINDINGS What is the central question of this study? Do mice submitted to sustained hypoxia present autonomic and respiratory changes similarly to rats? What is the main finding and its importance? Arterial pressure in the normal range, reduced baseline heart rate and tachypnoea were observed in behaving sustained hypoxia mice. Recordings in the in situ preparation of mice submitted to sustained hypoxia show an increase in cervical vagus nerve activity and a simultaneous reduction in thoracic sympathetic nerve activity correlated with changes in the respiratory cycle. Therefore, mice are an important model for studies on the modulation of sympathetic activity to the cardiovascular system and the vagus innervation of the upper airways due to changes in the respiratory network induced by sustained hypoxia. ABSTRACT Short-term sustained hypoxia (SH) in rats induces sympathetic overactivity and hypertension due to changes in sympathetic-respiratory coupling. However, there are no consistent data about the effect of SH on mice due to the different protocols of hypoxia and difficulties associated with the handling of these rodents under different experimental conditions. In situ recordings of autonomic and respiratory nerves in SH mice have not been performed yet. Herein, we evaluated the effects of SH ( F i O 2 = 0.1 for 24 h) on baseline mean arterial pressure (MAP), heart rate (HR), respiratory frequency (fR ) and responses to chemoreflex activation in behaving SH mice. A characterization of changes in cervical vagus (cVN), thoracic sympathetic (tSN), phrenic (PN) and abdominal (AbN) nerves in SH mice using the in situ working heart-brainstem preparation was also performed. SH mice presented normal MAP, significant reduction in baseline HR, increase in baseline fR , as well as increase in the magnitude of bradycardic response to chemoreflex activation. In in situ preparations, SH mice presented a reduction in PN discharge frequency, and increases in the time of expiration and incidence of late-expiratory bursts in AbN activity. Nerve recordings also indicated a significant increase in cVN activity and a significant reduction in tSN activity during expiration in SH mice. These findings make SH mice an important experimental model for better understanding how changes in the respiratory network may impact on the modulation of vagal control to the upper airways, as well as in the sympathetic activity to the cardiovascular system.
Collapse
Affiliation(s)
- Karla L Rodrigues
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Juliana R Souza
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Darlan S Bazilio
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Mauro de Oliveira
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Melina P S Moraes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Davi J A Moraes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Benedito H Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| |
Collapse
|
12
|
Ucak S, Dissanayake HU, Sutherland K, de Chazal P, Cistulli PA. Heart rate variability and obstructive sleep apnea: Current perspectives and novel technologies. J Sleep Res 2021; 30:e13274. [PMID: 33462936 DOI: 10.1111/jsr.13274] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022]
Abstract
Obstructive sleep apnea (OSA) is a highly prevalent condition, resulting in recurrent hypoxic events, sleep arousal, and daytime sleepiness. Patients with OSA are at an increased risk of cardiovascular morbidity and mortality. The mechanisms underlying the development of cardiovascular disease in OSA are multifactorial and cause a cascade of events. The primary contributing factor is sympathetic overactivity. Heart rate variability (HRV) can be used to evaluate shifts in the autonomic nervous system, during sleep and in response to treatment in patients with OSA. Newer technologies are aimed at improving HRV analysis to accelerate processing time, improve the diagnosis of OSA, and detection of cardiovascular risk. The present review will present contemporary understandings and uses for HRV, specifically in the realms of physiology, technology, and clinical management.
Collapse
Affiliation(s)
- Seren Ucak
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Hasthi U Dissanayake
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Kate Sutherland
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Philip de Chazal
- Faculty of Engineering, School of Biomedical Engineering, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Peter A Cistulli
- Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia.,Department of Respiratory and Sleep Medicine, Royal North Shore Hospital, Sydney, NSW, Australia
| |
Collapse
|
13
|
AlMarabeh S, O'Neill J, Cavers J, Lucking EF, O'Halloran KD, Abdulla MH. Chronic intermittent hypoxia impairs diuretic and natriuretic responses to volume expansion in rats with preserved low-pressure baroreflex control of the kidney. Am J Physiol Renal Physiol 2021; 320:F1-F16. [PMID: 33166181 DOI: 10.1152/ajprenal.00377.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 12/17/2022] Open
Abstract
We examined the effects of exposure to chronic intermittent hypoxia (CIH) on baroreflex control of renal sympathetic nerve activity (RSNA) and renal excretory responses to volume expansion (VE) before and after intrarenal transient receptor potential vanilloid 1 (TRPV1) blockade by capsaizepine (CPZ). Male Wistar rats were exposed to 96 cycles of hypoxia per day for 14 days (CIH) or normoxia. Urine flow and absolute Na+ excretion during VE were less in CIH-exposed rats, but the progressive decrease in RSNA during VE was preserved. Assessment of the high-pressure baroreflex revealed an increase in the operating and response range of RSNA and decreased slope in CIH-exposed rats with substantial hypertension [+19 mmHg basal mean arterial pressure (MAP)] but not in a second cohort with modest hypertension (+12 mmHg). Intrarenal CPZ caused diuresis, natriuresis, and a reduction in MAP in sham-exposed (sham) and CIH-exposed rats. After intrarenal CPZ, diuretic and natriuretic responses to VE in CIH-exposed rats were equivalent to those of sham rats. TRPV1 expression in the renal pelvic wall was similar in both experimental groups. Exposure to CIH did not elicit glomerular hypertrophy, renal inflammation, or oxidative stress. We conclude that exposure to CIH 1) does not impair the low-pressure baroreflex control of RSNA; 2) has modest effects on the high-pressure baroreflex control of RSNA, most likely indirectly due to hypertension; 3) can elicit hypertension in the absence of kidney injury; and 4) impairs diuretic and natriuretic responses to fluid overload. Our results suggest that exposure to CIH causes renal dysfunction, which may be relevant to obstructive sleep apnea.
Collapse
Affiliation(s)
- Sara AlMarabeh
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Julie O'Neill
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Jeremy Cavers
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Mohammed H Abdulla
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
14
|
Alzahrani AA, Cao LL, Aldossary HS, Nathanael D, Fu J, Ray CJ, Brain KL, Kumar P, Coney AM, Holmes AP. β-Adrenoceptor blockade prevents carotid body hyperactivity and elevated vascular sympathetic nerve density induced by chronic intermittent hypoxia. Pflugers Arch 2021; 473:37-51. [PMID: 33210151 PMCID: PMC7782391 DOI: 10.1007/s00424-020-02492-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022]
Abstract
Carotid body (CB) hyperactivity promotes hypertension in response to chronic intermittent hypoxia (CIH). The plasma concentration of adrenaline is reported to be elevated in CIH and our previous work suggests that adrenaline directly activates the CB. However, a role for chronic adrenergic stimulation in mediating CB hyperactivity is currently unknown. This study evaluated whether beta-blocker treatment with propranolol (Prop) prevented the development of CB hyperactivity, vascular sympathetic nerve growth and hypertension caused by CIH. Adult male Wistar rats were assigned into 1 of 4 groups: Control (N), N + Prop, CIH and CIH + Prop. The CIH paradigm consisted of 8 cycles h-1, 8 h day-1, for 3 weeks. Propranolol was administered via drinking water to achieve a dose of 40 mg kg-1 day-1. Immunohistochemistry revealed the presence of both β1 and β2-adrenoceptor subtypes on the CB type I cell. CIH caused a 2-3-fold elevation in basal CB single-fibre chemoafferent activity and this was prevented by chronic propranolol treatment. Chemoafferent responses to hypoxia and mitochondrial inhibitors were attenuated by propranolol, an effect that was greater in CIH animals. Propranolol decreased respiratory frequency in normoxia and hypoxia in N and CIH. Propranolol also abolished the CIH mediated increase in vascular sympathetic nerve density. Arterial blood pressure was reduced in propranolol groups during hypoxia. Propranolol exaggerated the fall in blood pressure in most (6/7) CIH animals during hypoxia, suggestive of reduced sympathetic tone. These findings therefore identify new roles for β-adrenergic stimulation in evoking CB hyperactivity, sympathetic vascular hyperinnervation and altered blood pressure control in response to CIH.
Collapse
Affiliation(s)
- Abdulaziz A Alzahrani
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Respiratory Care Department, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Lily L Cao
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Hayyaf S Aldossary
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- College of Medicine, Basic Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Demitris Nathanael
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Jiarong Fu
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Clare J Ray
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Keith L Brain
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Prem Kumar
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Andrew M Coney
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Andrew P Holmes
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
- Institute of Cardiovascular Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
15
|
Dyavanapalli J. Novel approaches to restore parasympathetic activity to the heart in cardiorespiratory diseases. Am J Physiol Heart Circ Physiol 2020; 319:H1153-H1161. [PMID: 33035444 DOI: 10.1152/ajpheart.00398.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neural control of the heart is regulated by sympathetic and parasympathetic divisions of the autonomic nervous system, both opposing each other to maintain cardiac homeostasis via regulating heart rate, conduction velocity, force of contraction, and coronary blood flow. Sympathetic hyperactivity and diminished parasympathetic activity are the characteristic features of many cardiovascular disease states including hypertension, myocardial ischemia, and arrhythmias that result in heart failure. Restoring parasympathetic activity to the heart has recently been identified as the promising approach to treat such conditions. However, approaches that used vagal nerve stimulation have been shown to be unsuccessful in heart failure. This review focuses on novel chemogenetic approaches used to identify the cardioprotective nature of activating neural points along the vagal pathway (both central and peripheral) while being selectively therapeutic in heart failure and obstructive sleep apnea.
Collapse
Affiliation(s)
- Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, George Washington University, Washington, District of Columbia
| |
Collapse
|
16
|
Exaggerated potassium current reduction by oxytocin in visceral sensory neurons following chronic intermittent hypoxia. Auton Neurosci 2020; 229:102735. [PMID: 33032244 DOI: 10.1016/j.autneu.2020.102735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 11/22/2022]
Abstract
Oxytocin (OT) from the hypothalamus is increased in several cardiorespiratory nuclei and systemically in response to a variety of stimuli and stressors, including hypoxia. Within the nucleus tractus solitarii (nTS), the first integration site for cardiorespiratory reflexes, OT enhances synaptic transmission, action potential (AP) discharge, and cardiac baroreflex gain. The hypoxic stressor obstructive sleep apnea, and its CIH animal model, elevates blood pressure and alters heart rate variability. The nTS receives sensory input from baroafferent neurons that originate in the nodose ganglia. Nodose neurons express the OT receptor (OTR) whose activation elevates intracellular calcium. However, the influence of OT on other ion channels, especially potassium channels important for neuronal activity during CIH, is less known. This study sought to determine the mechanism (s) by which OT modulates sensory afferent-nTS mediated reflexes normally and after CIH. Nodose ganglia neurons from male Sprague-Dawley rats were examined after 10d CIH (6% O2 every 3 min) or their normoxic (21% O2) control. OTR mRNA and protein were identified in Norm and CIH ganglia and was similar between groups. To examine OTR function, APs and potassium currents (IK) were recorded in dissociated neurons. Compared to Norm, after CIH OT depolarized neurons and reduced current-induced AP discharge. After CIH OT also produced a greater reduction in IK that where tetraethylammonium-sensitive. These data demonstrate after CIH OT alters ionic currents in nodose ganglia cells to likely influence cardiorespiratory reflexes and overall function.
Collapse
|
17
|
Gauda EB, Conde S, Bassi M, Zoccal DB, Almeida Colombari DS, Colombari E, Despotovic N. Leptin: Master Regulator of Biological Functions that Affects Breathing. Compr Physiol 2020; 10:1047-1083. [PMID: 32941688 DOI: 10.1002/cphy.c190031] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Obesity is a global epidemic in developed countries accounting for many of the metabolic and cardiorespiratory morbidities that occur in adults. These morbidities include type 2 diabetes, sleep-disordered breathing (SDB), obstructive sleep apnea, chronic intermittent hypoxia, and hypertension. Leptin, produced by adipocytes, is a master regulator of metabolism and of many other biological functions including central and peripheral circuits that control breathing. By binding to receptors on cells and neurons in the brainstem, hypothalamus, and carotid body, leptin links energy and metabolism to breathing. In this comprehensive article, we review the central and peripheral locations of leptin's actions that affect cardiorespiratory responses during health and disease, with a particular focus on obesity, SDB, and its effects during early development. Obesity-induced hyperleptinemia is associated with centrally mediated hypoventilation with decrease CO2 sensitivity. On the other hand, hyperleptinemia augments peripheral chemoreflexes to hypoxia and induces sympathoexcitation. Thus, "leptin resistance" in obesity is relative. We delineate the circuits responsible for these divergent effects, including signaling pathways. We review the unique effects of leptin during development on organogenesis, feeding behavior, and cardiorespiratory responses, and how undernutrition and overnutrition during critical periods of development can lead to cardiorespiratory comorbidities in adulthood. We conclude with suggestions for future directions to improve our understanding of leptin dysregulation and associated clinical diseases and possible therapeutic targets. Lastly, we briefly discuss the yin and the yang, specifically the contribution of relative adiponectin deficiency in adults with hyperleptinemia to the development of metabolic and cardiovascular disease. © 2020 American Physiological Society. Compr Physiol 10:1047-1083, 2020.
Collapse
Affiliation(s)
- Estelle B Gauda
- Division of Neonatology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Silvia Conde
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Lisboa, Portugal
| | - Mirian Bassi
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Debora Simoes Almeida Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Nikola Despotovic
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Deng B, Zhu W, Duan Y, Hu Y, Chen X, Song S, Yi Z, Song Y. Exendin‑4 promotes osteogenic differentiation of adipose‑derived stem cells and facilitates bone repair. Mol Med Rep 2019; 20:4933-4942. [PMID: 31661134 PMCID: PMC6854547 DOI: 10.3892/mmr.2019.10764] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammation-related bone defects pose a heavy burden on patients and orthopedic surgeons. Although stem-cell-based bone repair has developed rapidly, it is of great significance to characterize bio-active molecules that facilitate bone regeneration. It is reported that a glucagon-like peptide 1 receptor agonist, exendin-4, promoted bone regeneration mediated by the transplantation of adipose-derived stem cells in a metaphyseal defect mouse model of femur injury. However, the underlying mechanism is unclear. Bone imaging, immunohistochemistry real-time PCR and western blot analysis were used in the present study, and the results revealed that exendin-4 increased the transcription of the osteogenic differentiation-related genes and induced osteogenic differentiation in situ. Furthermore, the present data obtained from sorted adipose-derived stem cells revealed that exendin-4 promoted osteogenic differentiation and inhibited adipogenic differentiation in vitro. These findings indicated that exendin-4 facilitates osteogenic differentiation of transplanted adipose-derived stem cells for bone repair and illuminated clinical prospects of both adipose-derived stem cells and exendin-4 in stem-cell-based bone defect repair.
Collapse
Affiliation(s)
- Banglian Deng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wenzhong Zhu
- Department of Stomatology, Shaanxi Province Geriatric Hospital, Xi'an, Shaanxi 710005, P.R. China
| | - Yansheng Duan
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yuqian Hu
- Department of Stomatology, The Faculty of Medicine, Eastern University of Liaoning, Shenyang, Liaoning 110000, P.R. China
| | - Xuefeng Chen
- Xuefeng Dental Care Huaian, Huaian, Jiangsu 223000, P.R. China
| | - Shuang Song
- Health Science Center, Peking University, Beijing 100000, P.R. China
| | - Zian Yi
- Department of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yingliang Song
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Key Laboratory of Stomatology, Department of Oral Implantation, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
19
|
Mashaqi S, Gozal D. The impact of obstructive sleep apnea and PAP therapy on all-cause and cardiovascular mortality based on age and gender - a literature review. Respir Investig 2019; 58:7-20. [PMID: 31631059 DOI: 10.1016/j.resinv.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/28/2019] [Accepted: 08/20/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is a common sleep disorder which negatively impacts different body systems, especially the cardiovascular system. The correlation between sleep related breathing disorders and cardiovascular diseases has been well studied. However, the impact of OSA on cardiovascular related mortality and the role of positive airway pressure therapy in decreasing mortality is unclear. We reviewed studies investigating the impact of OSA on all-cause and cardiovascular related mortality in both genders, and in different age groups. METHODS A literature search (PubMed) using two phrases "obstructive sleep apnea and co-morbidities in males and females" and "obstructive sleep apnea and co-morbidities by age" yielded a total of 214 articles. Nineteen articles met the inclusion criteria. RESULTS The studies reviewed showed conflicting results. Some showed that OSA increases all cause and cardiovascular related mortality predominantly in the middle-aged group (40-65) followed by a plateau or a reduction in mortality. Other studies showed a positive linear correlation between OSA and mortality up to the age of 80. The same controversy was noted for gender; some studies did not observe an increase in mortality in females with OSA, while others observed a trend for an increase in mortality in females. CONCLUSION There is a debate in the literature regarding the impact of OSA on all-cause and cardiovascular mortality in both genders and in different age groups. However, the variation in results might be related to different study designs and significant epidemiological prevalence of OSA in males and females.
Collapse
Affiliation(s)
- Saif Mashaqi
- Department of Sleep Medicine, University of North Dakota School of Medicine and Health Sciences, Fargo, ND, USA.
| | - David Gozal
- Department of Child Health and the Child Health Research Institute, University of Missouri School of Medicine, Columbia, MO, USA
| |
Collapse
|
20
|
Zubcevic J, Richards EM, Yang T, Kim S, Sumners C, Pepine CJ, Raizada MK. Impaired Autonomic Nervous System-Microbiome Circuit in Hypertension. Circ Res 2019; 125:104-116. [PMID: 31219753 PMCID: PMC6588177 DOI: 10.1161/circresaha.119.313965] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypertension affects an estimated 103 million Americans, yet gaps in knowledge continue to limit its successful management. Rapidly emerging evidence is linking gut dysbiosis to many disorders and diseases including hypertension. The evolution of the -omics techniques has allowed determination of the abundance and potential function of gut bacterial species by next-generation bacterial sequencing, whereas metabolomics techniques report shifts in bacterial metabolites in the systemic circulation of hypertensive patients and rodent models of hypertension. The gut microbiome and host have evolved to exist in balance and cooperation, and there is extensive crosstalk between the 2 to maintain this balance, including during regulation of blood pressure. However, an understanding of the mechanisms of dysfunctional host-microbiome interactions in hypertension is still lacking. Here, we synthesize some of our recent data with published reports and present concepts and a rationale for our emerging hypothesis of a dysfunctional gut-brain axis in hypertension. Hopefully, this new information will improve the understanding of hypertension and help to address some of these knowledge gaps.
Collapse
Affiliation(s)
- Jasenka Zubcevic
- Department of Physiological Sciences, College of Veterinary Medicine; University of Florida, Gainesville FL32610
| | - Elaine M. Richards
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Tao Yang
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Seungbum Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Colin Sumners
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville FL32610
| | - Mohan K Raizada
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville FL32610
| |
Collapse
|
21
|
AlMarabeh S, Abdulla MH, O'Halloran KD. Is Aberrant Reno-Renal Reflex Control of Blood Pressure a Contributor to Chronic Intermittent Hypoxia-Induced Hypertension? Front Physiol 2019; 10:465. [PMID: 31105584 PMCID: PMC6491928 DOI: 10.3389/fphys.2019.00465] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 04/04/2019] [Indexed: 12/16/2022] Open
Abstract
Renal sensory nerves are important in the regulation of body fluid and electrolyte homeostasis, and blood pressure. Activation of renal mechanoreceptor afferents triggers a negative feedback reno-renal reflex that leads to the inhibition of sympathetic nervous outflow. Conversely, activation of renal chemoreceptor afferents elicits reflex sympathoexcitation. Dysregulation of reno-renal reflexes by suppression of the inhibitory reflex and/or activation of the excitatory reflex impairs blood pressure control, predisposing to hypertension. Obstructive sleep apnoea syndrome (OSAS) is causally related to hypertension. Renal denervation in patients with OSAS or in experimental models of chronic intermittent hypoxia (CIH), a cardinal feature of OSAS due to recurrent apnoeas (pauses in breathing), results in a decrease in circulating norepinephrine levels and attenuation of hypertension. The mechanism of the beneficial effect of renal denervation on blood pressure control in models of CIH and OSAS is not fully understood, since renal denervation interrupts renal afferent signaling to the brain and sympathetic efferent signals to the kidneys. Herein, we consider the currently proposed mechanisms involved in the development of hypertension in CIH disease models with a focus on oxidative and inflammatory mediators in the kidneys and their potential influence on renal afferent control of blood pressure, with wider consideration of the evidence available from a variety of hypertension models. We draw focus to the potential contribution of aberrant renal afferent signaling in the development, maintenance and progression of high blood pressure, which may have relevance to CIH-induced hypertension.
Collapse
Affiliation(s)
- Sara AlMarabeh
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Mohammed H Abdulla
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine and Health, University College Cork, Cork, Ireland
| |
Collapse
|
22
|
do Carmo JM, da Silva AA, Moak SP, da Silva FS, Spradley FT, Hall JE. Role of melanocortin 4 receptor in hypertension induced by chronic intermittent hypoxia. Acta Physiol (Oxf) 2019; 225:e13222. [PMID: 30466186 DOI: 10.1111/apha.13222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 11/13/2018] [Accepted: 11/15/2018] [Indexed: 12/23/2022]
Abstract
AIM We previously demonstrated that central nervous system (CNS) melanocortin 4 receptors (MC4R) play a key role in regulating blood pressure (BP) in some conditions associated with increased SNS activity, including obesity. In this study, we examined whether activation of CNS MC4R contributes to chronic intermittent hypoxia (CIH)-induced hypertension and ventilatory responses to hypercapnia. METHODS Rats were instrumented with an intracerebroventricular (ICV) cannula in the lateral cerebral ventricle for continuous infusion of MC4R antagonist (SHU-9119) and telemetry probes for measuring mean arterial pressure (MAP) and heart rate (HR). Untreated and SHU-9119-treated rats as well as obese and lean MC4R-deficient rats were exposed to CIH for 7-18 consecutive days. RESULTS Chronic intermittent hypoxia reduced cumulative food intake by 18 ± 5 g while MAP and HR increased by 10 ± 3 mm Hg and 9 ± 5 bpm in untreated rats. SHU-9119 increased food intake (from 15 ± 1 to 46 ± 3 g) and prevented CIH-induced reduction in food intake. CIH-induced hypertension was not attenuated by MC4R antagonism (average increase of 10 ± 1 vs 9 ± 1 mm Hg for untreated and SHU-9119 treated rats). In obese MC4R-deficient rats, CIH for 7 days raised BP by 11 ± 4 mm Hg. However, when MC4R-deficient rats were food restricted to prevent obesity, CIH-induced hypertension was attenuated by 32%. We also found that MC4R deficiency was associated with impaired ventilatory responses to hypercapnia independently of obesity. CONCLUSION These results show that obesity and the CNS melanocortin system interact in complex ways to elevate BP during CIH and that MC4R may be important in the ventilatory responses to hypercapnia.
Collapse
Affiliation(s)
- Jussara M. do Carmo
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular‐Renal Research Center University of Mississippi Medical Center Jackson Mississippi
| | - Alexandre A. da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular‐Renal Research Center University of Mississippi Medical Center Jackson Mississippi
- Barão de Mauá University Center Ribeirão Preto Brazil
- Universidade Estadual de Minas Gerais Passos Brazil
| | - Sydney P. Moak
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular‐Renal Research Center University of Mississippi Medical Center Jackson Mississippi
| | - Fernanda S. da Silva
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular‐Renal Research Center University of Mississippi Medical Center Jackson Mississippi
- Barão de Mauá University Center Ribeirão Preto Brazil
| | - Frank T. Spradley
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular‐Renal Research Center University of Mississippi Medical Center Jackson Mississippi
- Department of Surgery University of Mississippi Medical Center Jackson Mississippi
| | - John E. Hall
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, Cardiovascular‐Renal Research Center University of Mississippi Medical Center Jackson Mississippi
| |
Collapse
|
23
|
Liu W, Zhang W, Wang T, Wu J, Zhong X, Gao K, Liu Y, He X, Zhou Y, Wang H, Zeng H. Obstructive sleep apnea syndrome promotes the progression of aortic dissection via a ROS- HIF-1α-MMPs associated pathway. Int J Biol Sci 2019; 15:2774-2782. [PMID: 31853217 PMCID: PMC6909961 DOI: 10.7150/ijbs.34888] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/20/2019] [Indexed: 01/25/2023] Open
Abstract
Aims: Obstructive sleep apnea syndrome (OSAS) has been increasingly recognized as an independent risk factor for aortic dissection (AD) and it is strongly associated with the extent of intermittent hypoxia and re-oxygenation (IH). This study aimed to clarify role of ROS- HIF-1α-MMPs pathway in the pathogenesis of AD and whether the HIF-1α inhibitor attenuates AD formation. Methods and results: 8-week-old male ApoE-/- mice were given β-aminopropionitrile at a concentration of 0.1 % for 3 weeks and infused via osmotic mini pumps with either saline or 2,500 ng/min/kg angiotensin II (Ang II) for 2 weeks. To mimic the OSAS, one group was exposed to IH, which consisted of alternating cycles of 20.9% O2/8% O2 FiO2 (30 episodes per hour) with 20 s at the nadir FiO2 during the 12-h light phase, 2 weeks before Ang II infusion. After Ang II infusion, we assessed remodeling in the aorta by echocardiography, histological and immunohistochemical analysis. IH treatment resulted in significant enlargement of the luminal area, destruction of the media, marked thickening of the adventitia, higher incidence of AD formation and lower survival rate in compared with the Ang II only group. Moreover, IH exposure markedly increased the aortic ROS production and subsequent HIF-1α expression, which in turn promoted the expressions of VEGF, MMP2 and MMP9 and finally leading to the progression of AD. Besides, in vitro study confirmed that IH induced HIF-1α expression plays an important role in the induction of MMPs and that is regulated by the PI3K/AKT/FRAP pathway. Intriguingly, a selective HIF-1α inhibitor KC7F2 could significantly ameliorate IH exposure induced aforementioned deleterious effects in vitro and in vivo.Conclusion: OSAS induced IH can promote the occurrence and progression of AD via a ROS- HIF-1α-MMPs associated pathway. The selective HIF-1α inhibitor KC7F2 could be a novel therapeutic agent for AD patient with OSAS.
Collapse
Affiliation(s)
- Wanjun Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Wenjun Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Tao Wang
- Department of Cardiology, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, 261000, PR China
| | - Jinhua Wu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Xiaodan Zhong
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Kun Gao
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Yujian Liu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Xingwei He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
| | - Yiwu Zhou
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Hongjie Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
- ✉ Corresponding author: Hongjie Wang, , Tel. +86-27-8369-3794, Fax: +86-27-8366-3186; Hesong Zeng, , Tel. +86-27-8369-2850, Fax: +86-27-8366-3186
| | - Hesong Zeng
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, 430030, PR China
- ✉ Corresponding author: Hongjie Wang, , Tel. +86-27-8369-3794, Fax: +86-27-8366-3186; Hesong Zeng, , Tel. +86-27-8369-2850, Fax: +86-27-8366-3186
| |
Collapse
|
24
|
Abstract
Obstructive sleep apnoea (OSA) is recognized as a major public health burden conveying a significant risk of cardiovascular diseases (CVD) and mortality. Continuous positive airway pressure (CPAP) is the treatment of choice for the majority of patients with OSA but the benefit of CPAP on CVD is uncertain. Thus, a greater understanding of the mechanisms by which OSA leads to CVD might identify novel therapeutic approaches. Intermittent hypoxia (IH), a hallmark feature of OSA, plays a key role in the pathogenesis and experimental studies using animal and cell culture studies suggest that IH mediates CVD through activation of multiple mechanistic pathways such as sympathetic excitation, inflammation, oxidative stress or metabolic dysregulation. Recurrent arousals, intrathoracic pressure swings and concomitant obesity likely play important additive roles in this process. In this review, the available evidence of the pathophysiological mechanisms of CVD in OSA is explored with a specific emphasis on IH, recurrent arousals and intrathoracic pressure swings as the main pathophysiological triggers.
Collapse
Affiliation(s)
- Silke Ryan
- Pulmonary and Sleep Disorders Unit, St. Vincent's University Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
25
|
Xu WX, Yu JL, Feng Y, Yan QX, Li XY, Li Y, Liu Z, Wang D, Sun X, Li KX, Wang LQ, Qiao GF, Li BY. Spontaneous activities in baroreflex afferent pathway contribute dominant role in parasympathetic neurocontrol of blood pressure regulation. CNS Neurosci Ther 2018; 24:1219-1230. [PMID: 30044043 DOI: 10.1111/cns.13039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022] Open
Abstract
AIM To study the dominant role of parasympathetic inputs at cellular level of baroreflex afferent pathway and underlying mechanism in neurocontrol of blood pressure regulation. METHODS Whole-cell patch-clamp and animal study were conducted. RESULTS For the first time, we demonstrated the spontaneous activities from resting membrane potential in myelinated A- and Ah-type baroreceptor neurons (BRNs, the 1st-order), but not in unmyelinated C-types, using vagus-nodose slice of adult female rats. These data were further supported by the notion that the spontaneous synaptic currents could only be seen in the pharmacologically and electrophysiologically defined myelinated A- and Ah-type baroreceptive neurons (the 2nd-order) of NTS using brainstem slice of adult female rats. The greater frequency and the larger amplitude of the spontaneous excitatory postsynaptic currents (EPSCs) compared with the inhibitory postsynaptic currents (IPSCs) were only observed in Ah-types. The ratio of EPSCs:IPSCs was estimated at 3:1 and higher. These results confirmed that the afferent-specific spontaneous activities were generated from baroreflex afferent pathway in female-specific subpopulation of myelinated Ah-type BRNs in nodose and baroreceptive neurons in NTS, which provided a novel insight into the dominant role of sex-specific baroreflex-evoked parasympathetic drives in retaining a stable and lower blood pressure status in healthy subjects, particularly in females. CONCLUSION The data from current investigations establish a new concept for the role of Ah-type baroreceptor/baroreceptive neurons in controlling blood pressure stability and provide a new pathway for pharmacological intervention for hypertension and cardiovascular diseases.
Collapse
Affiliation(s)
- Wen-Xiao Xu
- Department of Orthopedic Surgery, the 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jin-Ling Yu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan Feng
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Qiu-Xin Yan
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xin-Yu Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ying Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhuo Liu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.,Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Di Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.,Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Xun Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ke-Xin Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lu-Qi Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.,Department of Biomedical Engineering, Indiana University Purdue University Indianapolis School of Engineering and Technology, Indianapolis, Indiana
| | - Guo-Fen Qiao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Bai-Yan Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
26
|
Bober SL, Ciriello J, Jones DL. Atrial arrhythmias and autonomic dysfunction in rats exposed to chronic intermittent hypoxia. Am J Physiol Heart Circ Physiol 2018; 314:H1160-H1168. [DOI: 10.1152/ajpheart.00173.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Obstructive sleep apnea, which involves chronic intermittent hypoxia (CIH), is a major risk factor for developing atrial fibrillation (AF). Whether or not CIH alone alters cardiac mechanisms to support AF is unknown. This study investigated the effects of CIH on atrial electrophysiology and arrhythmia vulnerability and evaluated the role of autonomics in CIH promotion of AF. Adult male Sprague-Dawley rats were exposed to 8 h/day of CIH or normoxia for 7 days. After exposure, rats were anesthetized for intracardiac electrophysiological experiments. Atrial effective refractory periods (AERPs) and AF inducibility were determined using programmed electrical stimulation and burst pacing in the absence and presence of autonomic receptor agonists and antagonists. Western blot analysis measured atrial protein expression of muscarinic M2, M3, and β1-adrenergic receptors. Compared with normoxia-exposed control rats, CIH-exposed rats had enhanced AF vulnerability using both programmed electrical stimulation and burst pacing, accompanied by greater AERP responses to carbachol and propranolol, lesser responses to isoproterenol, and higher atrial M2 receptor protein levels. Enhanced atrial vulnerability was accentuated by carbachol and abolished by atropine, indicating that the AF-promoting effects of CIH depended principally on parasympathetic activation. Enhancement of atrial vulnerability and AERP shortening with cholinergic agonists in CIH-exposed rats is consistent with sensitivity to parasympathetic activation. Higher responses to adrenergic receptor blockade in CIH-exposed rats is consistent with sympathetic potentiation. These findings implicate CIH as an important mediator of enhanced AF susceptibility in obstructive sleep apnea and provide novel insights into the underlying mechanisms. NEW & NOTEWORTHY Our study demonstrates, for the first time, that chronic intermittent hypoxia alone enhances vulnerability to atrial arrhythmia induction, which depends principally on parasympathetic activation. Enhanced atrial vulnerability was accompanied by heightened electrophysiological responses of the atrial myocardium to carbachol and isoproterenol, dampened responses to propranolol, and increased atrial M2 receptor protein levels.
Collapse
Affiliation(s)
- Sara L. Bober
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - John Ciriello
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Douglas L. Jones
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
27
|
Machado BH, Zoccal DB, Moraes DJA. Neurogenic hypertension and the secrets of respiration. Am J Physiol Regul Integr Comp Physiol 2017; 312:R864-R872. [PMID: 28438764 PMCID: PMC6148211 DOI: 10.1152/ajpregu.00505.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 11/22/2022]
Abstract
Despite recent advances in the knowledge of the neural control of cardiovascular function, the cause of sympathetic overactivity in neurogenic hypertension remains unknown. Studies from our laboratory point out that rats submitted to chronic intermittent hypoxia (CIH), an experimental model of neurogenic hypertension, present changes in the central respiratory network that impact the pattern of sympathetic discharge and the levels of arterial pressure. In addition to the fine coordination of respiratory muscle contraction and relaxation, which is essential for O2 and CO2 pulmonary exchanges, neurons of the respiratory network are connected precisely to the neurons controlling the sympathetic activity in the brain stem. This respiratory-sympathetic neuronal interaction provides adjustments in the sympathetic outflow to the heart and vasculature during each respiratory phase according to the metabolic demands. Herein, we report that CIH-induced sympathetic over activity and mild hypertension are associated with increased frequency discharge of ventral medullary presympathetic neurons. We also describe that their increased frequency discharge is dependent on synaptic inputs, mostly from neurons of the brain stem respiratory network, rather than changes in their intrinsic electrophysiological properties. In perspective, we are taking into consideration the possibility that changes in the central respiratory rhythm/pattern generator contribute to increased sympathetic outflow and the development of neurogenic hypertension. Our experimental evidence provides support for the hypothesis that changes in the coupling of respiratory and sympathetic networks might be one of the unrevealed secrets of neurogenic hypertension in rats.
Collapse
Affiliation(s)
- Benedito H Machado
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; and
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry of Araraquara, São Paulo State University, Araraquara, São Paulo, Brazil
| | - Davi J A Moraes
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; and
| |
Collapse
|
28
|
Cheng Z(J. Vagal cardiac efferent innervation in F344 rats: Effects of chronic intermittent hypoxia. Auton Neurosci 2017; 203:9-16. [DOI: 10.1016/j.autneu.2016.10.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 10/05/2016] [Accepted: 10/27/2016] [Indexed: 12/27/2022]
|
29
|
Gemel J, Su Z, Gileles-Hillel A, Khalyfa A, Gozal D, Beyer EC. Intermittent hypoxia causes NOX2-dependent remodeling of atrial connexins. BMC Cell Biol 2017; 18:7. [PMID: 28124622 PMCID: PMC5267331 DOI: 10.1186/s12860-016-0117-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Obstructive sleep apnea has been linked to the development of heart disease and arrhythmias, including atrial fibrillation. Since altered conduction through gap junction channels can contribute to the pathogenesis of such arrhythmias, we examined the abundance and distributions of the major cardiac gap junction proteins, connexin40 (Cx40) and connexin43 (Cx43) in mice treated with sleep fragmentation or intermittent hypoxia (IH) as animal models of the components of obstructive sleep apnea. RESULTS Wild type C57BL/6 mice or mice lacking NADPH 2 (NOX2) oxidase activity (gp91phox(-/Y)) were exposed to room air or to SF or IH for 6 weeks. Then, the mice were sacrificed, and atria and ventricles were immediately dissected. The abundances of Cx40 or Cx43 in atria and ventricles were unaffected by SF. In contrast, immunoblots showed that the abundance of atrial Cx40 and Cx43 and ventricular Cx43 were reduced in mice exposed to IH. qRT-PCR demonstrated significant reductions of atrial Cx40 and Cx43 mRNAs. Immunofluorescence microscopy revealed that the abundance and size of gap junctions containing Cx40 or Cx43 were reduced in atria by IH treatment of mice. However, no changes of connexin abundance or gap junction size/abundance were observed in IH-treated NOX2-null mice. CONCLUSIONS These results demonstrate that intermittent hypoxia (but not sleep fragmentation) causes reductions and remodeling of atrial Cx40 and Cx43. These alterations may contribute to the substrate for atrial fibrillation that develops in response to obstructive sleep apnea. Moreover, these connexin changes are likely generated in response to reactive oxygen species generated by NOX2.
Collapse
Affiliation(s)
- Joanna Gemel
- Department of Pediatrics, University of Chicago, 900 E. 57th St. KCBD 5152, Chicago, IL, 60637, USA
| | - Zihan Su
- Present address: Williams College, Williamstown, MA, USA
| | - Alex Gileles-Hillel
- Present address: Department of Pediatrics, Hadassah-Hebrew University Medical Center, Mt. Scopus, Jerusalem, Israel
| | - Abdelnaby Khalyfa
- Department of Pediatrics, University of Chicago, 900 E. 57th St. KCBD 5152, Chicago, IL, 60637, USA
| | - David Gozal
- Department of Pediatrics, University of Chicago, 900 E. 57th St. KCBD 5152, Chicago, IL, 60637, USA
| | - Eric C Beyer
- Department of Pediatrics, University of Chicago, 900 E. 57th St. KCBD 5152, Chicago, IL, 60637, USA.
| |
Collapse
|
30
|
Moraes DJ, Bonagamba LG, da Silva MP, Mecawi AS, Antunes-Rodrigues J, Machado BH. Respiratory Network Enhances the Sympathoinhibitory Component of Baroreflex of Rats Submitted to Chronic Intermittent Hypoxia. Hypertension 2016; 68:1021-30. [DOI: 10.1161/hypertensionaha.116.07731] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/09/2016] [Indexed: 11/16/2022]
Abstract
Chronic intermittent hypoxia (CIH) produces respiratory-related sympathetic overactivity and hypertension in rats. In this study, we tested the hypothesis that the enhanced central respiratory modulation of sympathetic activity after CIH also decreases the sympathoinhibitory component of baroreflex of rats, which may contribute to the development of hypertension. Wistar rats were exposed to CIH or normoxia (control group) for 10 days. Phrenic nerve, thoracic sympathetic nerve, and neurons in the rostral ventrolateral medulla and caudal ventrolateral medulla were recorded in in situ preparations of rats. Baroreflex regulation of thoracic sympathetic nerve, rostral ventrolateral medulla, and caudal ventrolateral medulla neurons activities were evaluated in different phases of respiration in response to either aortic depressor nerve stimulation or pressure stimuli. CIH rats presented higher respiratory-related thoracic sympathetic nerve and rostral ventrolateral medulla presympathetic neurons activities at the end of expiration in relation to control rats, which are indexes of respiratory-related sympathetic overactivity. Baroreflex-evoked thoracic sympathetic nerve inhibition during expiration, but not during inspiration, was enhanced in CIH when compared with control rats. In addition, CIH selectively enhanced the expiratory-related baroreceptor inputs, probably through caudal ventrolateral medulla neurons, to the respiratory-modulated bulbospinal rostral ventrolateral medulla presympathetic neurons. These findings support the concept that the onset of hypertension, mediated by sympathetic overactivity, after 10 days of CIH is not secondary to a reduction in sympathoinhibitory component of baroreflex. Instead, it was observed an increase in the gain of sympathoinhibitory component in in situ preparations of rats, suggesting that changes in the respiratory-related sympathetic network after CIH also play a key role in preventing greater increase in arterial pressure.
Collapse
Affiliation(s)
- Davi J.A. Moraes
- From the Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Leni G.H. Bonagamba
- From the Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Melina P. da Silva
- From the Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - André S. Mecawi
- From the Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - José Antunes-Rodrigues
- From the Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| | - Benedito H. Machado
- From the Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Brazil
| |
Collapse
|
31
|
Morgan BJ, Bates ML, Rio RD, Wang Z, Dopp JM. Oxidative stress augments chemoreflex sensitivity in rats exposed to chronic intermittent hypoxia. Respir Physiol Neurobiol 2016; 234:47-59. [PMID: 27595979 DOI: 10.1016/j.resp.2016.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/29/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022]
Abstract
Chronic exposure to intermittent hypoxia (CIH) elicits plasticity of the carotid sinus and phrenic nerves via reactive oxygen species (ROS). To determine whether CIH-induced alterations in ventilation, metabolism, and heart rate are also dependent on ROS, we measured responses to acute hypoxia in conscious rats after 14 and 21 d of either CIH or normoxia (NORM), with or without concomitant administration of allopurinol (xanthine oxidase inhibitor), combined allopurinol plus losartan (angiotensin II type 1 receptor antagonist), or apocynin (NADPH oxidase inhibitor). Carotid body nitrotyrosine production was measured by immunohistochemistry. CIH produced an increase in the ventilatory response to acute hypoxia that was virtually eliminated by all three pharmacologic interventions. CIH caused a robust increase in carotid body nitrotyrosine production that was greatly attenuated by allopurinol plus losartan and by apocynin but unaffected by allopurinol. CIH caused a decrease in metabolic rate and a reduction in hypoxic bradycardia. Both of these effects were prevented by allopurinol, allopurinol plus losartan, and apocynin.
Collapse
Affiliation(s)
- Barbara J Morgan
- John Rankin Laboratory of Pulmonary Medicine, School of Medicine and Public Health: University of Wisconsin, Madison, WI 53706, USA; Department of Orthopedics and Rehabilitation, School of Medicine and Public Health; University of Wisconsin, Madison, WI 53706, USA.
| | - Melissa L Bates
- Department of Health and Human Physiology, College of Liberal Arts and Sciences; University of Iowa, Iowa City, IA 52242, USA
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Universidad Autónoma de Chile, Santiago, Chile
| | - Zunyi Wang
- Department of Surgical Sciences, School of Veterinary Medicine; University of Wisconsin, Madison, WI 53706, USA
| | - John M Dopp
- Pharmacy Practice Division, School of Pharmacy; University of Wisconsin, Madison, WI 53705, USA
| |
Collapse
|
32
|
Choy L, Yeo JM, Tse V, Chan SP, Tse G. Cardiac disease and arrhythmogenesis: Mechanistic insights from mouse models. IJC HEART & VASCULATURE 2016; 12:1-10. [PMID: 27766308 PMCID: PMC5064289 DOI: 10.1016/j.ijcha.2016.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/02/2016] [Indexed: 12/19/2022]
Abstract
The mouse is the second mammalian species, after the human, in which substantial amount of the genomic information has been analyzed. With advances in transgenic technology, mutagenesis is now much easier to carry out in mice. Consequently, an increasing number of transgenic mouse systems have been generated for the study of cardiac arrhythmias in ion channelopathies and cardiomyopathies. Mouse hearts are also amenable to physical manipulation such as coronary artery ligation and transverse aortic constriction to induce heart failure, radiofrequency ablation of the AV node to model complete AV block and even implantation of a miniature pacemaker to induce cardiac dyssynchrony. Last but not least, pharmacological models, despite being simplistic, have enabled us to understand the physiological mechanisms of arrhythmias and evaluate the anti-arrhythmic properties of experimental agents, such as gap junction modulators, that may be exert therapeutic effects in other cardiac diseases. In this article, we examine these in turn, demonstrating that primary inherited arrhythmic syndromes are now recognized to be more complex than abnormality in a particular ion channel, involving alterations in gene expression and structural remodelling. Conversely, in cardiomyopathies and heart failure, mutations in ion channels and proteins have been identified as underlying causes, and electrophysiological remodelling are recognized pathological features. Transgenic techniques causing mutagenesis in mice are extremely powerful in dissecting the relative contributions of different genes play in producing disease phenotypes. Mouse models can serve as useful systems in which to explore how protein defects contribute to arrhythmias and direct future therapy.
Collapse
Affiliation(s)
- Lois Choy
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Jie Ming Yeo
- School of Medicine, Imperial College London, SW7 2AZ, UK
| | - Vivian Tse
- Department of Physiology, McGill University, Canada
| | - Shing Po Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Gary Tse
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
33
|
Dyavanapalli J, Dergacheva O, Wang X, Mendelowitz D. Parasympathetic Vagal Control of Cardiac Function. Curr Hypertens Rep 2016; 18:22. [PMID: 26849575 DOI: 10.1007/s11906-016-0630-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This brief review focuses on four new topics, with novel and clinically significant consequences, concerning the powerful influence of parasympathetic activity on cardiac function. In this short summary, we will highlight very recent and important work, published in the last 3-4 years, that (1) challenges the paradigm that parasympathetic activity to the heart is involved in the control of heart rate but plays little role in other cardiac functions, (2) characterizes important long-range synaptic pathways to parasympathetic cardiac vagal neurons that are involved in "higher" brain functions (such as arousal and emotional challenges), (3) asks whether implantable chronic vagal nerve stimulation is a promising clinical tool for treating cardiovascular diseases, and (4) describes newly identified neuropeptides and other modulators that can influence the generation and maintenance of parasympathetic activity to the heart.
Collapse
Affiliation(s)
- Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, The George Washington University, 2300 Eye St NW, Washington, DC, 20037, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, The George Washington University, 2300 Eye St NW, Washington, DC, 20037, USA
| | - Xin Wang
- Department of Pharmacology and Physiology, The George Washington University, 2300 Eye St NW, Washington, DC, 20037, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University, 2300 Eye St NW, Washington, DC, 20037, USA.
| |
Collapse
|
34
|
Del Rio R, Andrade DC, Lucero C, Arias P, Iturriaga R. Carotid Body Ablation Abrogates Hypertension and Autonomic Alterations Induced by Intermittent Hypoxia in Rats. Hypertension 2016; 68:436-45. [PMID: 27381902 DOI: 10.1161/hypertensionaha.116.07255] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/06/2016] [Indexed: 01/12/2023]
Abstract
Chronic intermittent hypoxia (CIH), the main feature of obstructive sleep apnea, enhances carotid body (CB) chemosensory responses to hypoxia and produces autonomic dysfunction, cardiac arrhythmias, and hypertension. We tested whether autonomic alterations, arrhythmogenesis, and the progression of hypertension induced by CIH depend on the enhanced CB chemosensory drive, by ablation of the CB chemoreceptors. Male Sprague-Dawley rats were exposed to control (Sham) conditions for 7 days and then to CIH (5% O2, 12/h 8 h/d) for a total of 28 days. At 21 days of CIH exposure, rats underwent bilateral CB ablation and then exposed to CIH for 7 additional days. Arterial blood pressure and ventilatory chemoreflex response to hypoxia were measured in conscious rats. In addition, cardiac autonomic imbalance, cardiac baroreflex gain, and arrhythmia score were assessed during the length of the experiments. In separate experimental series, we measured extracellular matrix remodeling content in cardiac atrial tissue and systemic oxidative stress. CIH induced hypertension, enhanced ventilatory response to hypoxia, induced autonomic imbalance toward sympathetic preponderance, reduced baroreflex gain, and increased arrhythmias and atrial fibrosis. CB ablation normalized blood pressure, reduced ventilatory response to hypoxia, and restored cardiac autonomic and baroreflex function. In addition, CB ablation reduced the number of arrhythmias, but not extracellular matrix remodeling or systemic oxidative stress, suggesting that reductions in arrhythmia incidence during CIH were related to normalization of cardiac autonomic balance. Present results show that autonomic alterations induced by CIH are critically dependent on the CB and support a main role for the CB in the CIH-induced hypertension.
Collapse
Affiliation(s)
- Rodrigo Del Rio
- From the Laboratory of Cardiorespiratory Control, Center of Biomedical Research, Universidad Autónoma de Chile, Santiago, Chile (R.D.R., C.L.); and Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile (D.C.A., P.A., R.I.)
| | - David C Andrade
- From the Laboratory of Cardiorespiratory Control, Center of Biomedical Research, Universidad Autónoma de Chile, Santiago, Chile (R.D.R., C.L.); and Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile (D.C.A., P.A., R.I.)
| | - Claudia Lucero
- From the Laboratory of Cardiorespiratory Control, Center of Biomedical Research, Universidad Autónoma de Chile, Santiago, Chile (R.D.R., C.L.); and Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile (D.C.A., P.A., R.I.)
| | - Paulina Arias
- From the Laboratory of Cardiorespiratory Control, Center of Biomedical Research, Universidad Autónoma de Chile, Santiago, Chile (R.D.R., C.L.); and Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile (D.C.A., P.A., R.I.)
| | - Rodrigo Iturriaga
- From the Laboratory of Cardiorespiratory Control, Center of Biomedical Research, Universidad Autónoma de Chile, Santiago, Chile (R.D.R., C.L.); and Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile (D.C.A., P.A., R.I.).
| |
Collapse
|
35
|
Iturriaga R, Del Rio R, Idiaquez J, Somers VK. Carotid body chemoreceptors, sympathetic neural activation, and cardiometabolic disease. Biol Res 2016; 49:13. [PMID: 26920146 PMCID: PMC4768417 DOI: 10.1186/s40659-016-0073-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/12/2016] [Indexed: 11/10/2022] Open
Abstract
The carotid body (CB) is the main peripheral chemoreceptor that senses the arterial PO2, PCO2 and pH. In response to hypoxemia, hypercapnia and acidosis, carotid chemosensory discharge elicits reflex respiratory, autonomic and cardiovascular adjustments. The classical construct considers the CB as the main peripheral oxygen sensor, triggering reflex physiological responses to acute hypoxemia and facilitating the ventilatory acclimation to chronic hypoxemia at high altitude. However, a growing body of experimental evidence supports the novel concept that an abnormally enhanced CB chemosensory input to the brainstem contributes to overactivation
of the sympathetic nervous system, and consequent pathology. Indeed, the CB has been implicated in several diseases associated with increases in central sympathetic outflow. These include hypertension, heart failure, sleep apnea, chronic obstructive pulmonary disease and metabolic syndrome. Indeed, ablation of the CB has been proposed for the treatment of severe and resistant hypertension in humans. In this review, we will analyze and discuss new evidence supporting an important role for the CB chemoreceptor in the progression of autonomic and cardiorespiratory alterations induced by heart failure, obstructive sleep apnea, chronic obstructive pulmonary disease and metabolic syndrome.
Collapse
Affiliation(s)
- Rodrigo Iturriaga
- Laboratorio de Neurobiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Centro de Investigación Biomédica, Universidad Autónoma de Chile, Santiago, Chile. .,Dirección de Investigación, Universidad Científica del Sur, Lima, Peru.
| | - Juan Idiaquez
- Catedra de Neurología, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile.
| | - Virend K Somers
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
36
|
Morgan BJ, Adrian R, Wang ZY, Bates ML, Dopp JM. Chronic intermittent hypoxia alters ventilatory and metabolic responses to acute hypoxia in rats. J Appl Physiol (1985) 2016; 120:1186-95. [PMID: 26917692 DOI: 10.1152/japplphysiol.00015.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/22/2016] [Indexed: 12/18/2022] Open
Abstract
We determined the effects of chronic exposure to intermittent hypoxia (CIH) on chemoreflex control of ventilation in conscious animals. Adult male Sprague-Dawley rats were exposed to CIH [nadir oxygen saturation (SpO2), 75%; 15 events/h; 10 h/day] or normoxia (NORM) for 21 days. We assessed the following responses to acute, graded hypoxia before and after exposures: ventilation (V̇e, via barometric plethysmography), V̇o2 and V̇co2 (analysis of expired air), heart rate (HR), and SpO2 (pulse oximetry via neck collar). We quantified hypoxia-induced chemoreceptor sensitivity by calculating the stimulus-response relationship between SpO2 and the ventilatory equivalent for V̇co2 (linear regression). An additional aim was to determine whether CIH causes proliferation of carotid body glomus cells (using bromodeoxyuridine). CIH exposure increased the slope of the V̇e/V̇co2/SpO2 relationship and caused hyperventilation in normoxia. Bromodeoxyuridine staining was comparable in CIH and NORM. Thus our CIH paradigm augmented hypoxic chemosensitivity without causing glomus cell proliferation.
Collapse
Affiliation(s)
- Barbara J Morgan
- John Rankin Laboratory of Pulmonary Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin; Department of Orthopedics and Rehabilitation, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin;
| | - Russell Adrian
- John Rankin Laboratory of Pulmonary Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin; Department of Orthopedics and Rehabilitation, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Zun-Yi Wang
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin
| | - Melissa L Bates
- Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, Iowa; and
| | - John M Dopp
- Pharmacy Practice Division, School of Pharmacy, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
37
|
The Effects of Experimental Sleep Apnea on Cardiac and Respiratory Functions in 6 and 18 Month Old Dystrophic (mdx) Mice. PLoS One 2016; 11:e0147640. [PMID: 26808526 PMCID: PMC4726600 DOI: 10.1371/journal.pone.0147640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/05/2016] [Indexed: 11/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal disease where over 90% of patients succumb to respiratory or cardiac failure. Sleep apnea and sleep disordered breathing (SDB) are noted in a plurality of DMD patients, and the resulting nocturnal episodic hypoxia (EH) cannot be ruled out as a contributing factor to cardiac and respiratory dysfunction. In this study, we investigated the impact of long-term episodic hypoxia, which mimics the cyclic hypoxia seen in sleep apnea, on cardiac and respiratory function in a murine model of DMD (mdx mice). Since the severity and prevalence of sleep apnea in DMD increases with age, we studied the impact of EH on young (6-month) and on older (18-month) mdx mice. Mice were either exposed for 12 weeks to EH (8 hours/day, 5 days/week) or to room air. We noted a significant increase in left ventricular (LV) dilatation (transthoracic echocardiography) on EH exposure in both age groups, but reduced LV contractility was seen only in 6-month old mice. With EH exposure, an increased fibrosis (hydroxyproline) was noted in both cardiac and diaphragm muscle in 18-month but not 6-month old mice. No significant change in relative diaphragm strength (in-vitro) was noted on EH exposure in 18-month old mice. In contrast, EH exposed 6-month old mice showed a significant increase in relative diaphragm strength. EH exposure did not result in any significant change in ventilatory parameters (barometric plethysmography) in awake 6-month old mdx mice. In contrast, 18-month old mdx mice showed considerable ventilatory dysfunction, consistent with reduced ventilatory reserve. Our findings highlight that sleep apnea impacts respiratory and cardiac function in muscular dystrophy, and that EH can have divergent effects on both systems. To our knowledge, this is the first comprehensive study to investigate the impact of EH on cardiac and respiratory function in mdx mice.
Collapse
|
38
|
SOD1 Overexpression Preserves Baroreflex Control of Heart Rate with an Increase of Aortic Depressor Nerve Function. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:3686829. [PMID: 26823951 PMCID: PMC4707341 DOI: 10.1155/2016/3686829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 10/01/2015] [Indexed: 02/07/2023]
Abstract
Overproduction of reactive oxygen species (ROS), such as the superoxide radical (O2∙−), is associated with diseases which compromise cardiac autonomic function. Overexpression of SOD1 may offer protection against ROS damage to the cardiac autonomic nervous system, but reductions of O2∙− may interfere with normal cellular functions. We have selected the C57B6SJL-Tg (SOD1)2 Gur/J mouse as a model to determine whether SOD1 overexpression alters cardiac autonomic function, as measured by baroreflex sensitivity (BRS) and aortic depressor nerve (ADN) recordings, as well as evaluation of baseline heart rate (HR) and mean arterial pressure (MAP). Under isoflurane anesthesia, C57 wild-type and SOD1 mice were catheterized with an arterial pressure transducer and measurements of HR and MAP were taken. After establishing a baseline, hypotension and hypertension were induced by injection of sodium nitroprusside (SNP) and phenylephrine (PE), respectively, and ΔHR versus ΔMAP were recorded as a measure of baroreflex sensitivity (BRS). SNP and PE treatment were administered sequentially after a recovery period to measure arterial baroreceptor activation by recording aortic depressor nerve activity. Our findings show that overexpression of SOD1 in C57B6SJL-Tg (SOD1)2 Gur/J mouse preserved the normal HR, MAP, and BRS but enhanced aortic depressor nerve function.
Collapse
|
39
|
Efficacy of carvedilol in reversing hypertension induced by chronic intermittent hypoxia in rats. Eur J Pharmacol 2015; 765:58-67. [DOI: 10.1016/j.ejphar.2015.08.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/02/2015] [Accepted: 08/16/2015] [Indexed: 10/23/2022]
|
40
|
Del Rio R, Moya EA, Iturriaga R. Carotid body potentiation during chronic intermittent hypoxia: implication for hypertension. Front Physiol 2014; 5:434. [PMID: 25429271 PMCID: PMC4228839 DOI: 10.3389/fphys.2014.00434] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/24/2014] [Indexed: 11/30/2022] Open
Abstract
Autonomic dysfunction is involved in the development of hypertension in humans with obstructive sleep apnea, and animals exposed to chronic intermittent hypoxia (CIH). It has been proposed that a crucial step in the development of the hypertension is the potentiation of the carotid body (CB) chemosensory responses to hypoxia, but the temporal progression of the CB chemosensory, autonomic and hypertensive changes induced by CIH are not known. We tested the hypothesis that CB potentiation precedes the autonomic imbalance and the hypertension in rats exposed to CIH. Thus, we studied the changes in CB chemosensory and ventilatory responsiveness to hypoxia, the spontaneous baroreflex sensitivity (BRS), heart rate variability (HRV) and arterial blood pressure in pentobarbital anesthetized rats exposed to CIH for 7, 14, and 21 days. After 7 days of CIH, CB chemosensory and ventilatory responses to hypoxia were enhanced, while BRS was significantly reduced by 2-fold in CIH-rats compared to sham-rats. These alterations persisted until 21 days of CIH. After 14 days, CIH shifted the HRV power spectra suggesting a predominance of sympathetic over parasympathetic tone. In contrast, hypertension was found after 21 days of CIH. Concomitant changes between the gain of spectral HRV, BRS, and ventilatory hypoxic chemoreflex showed that the CIH-induced BRS attenuation preceded the HRV changes. CIH induced a simultaneous decrease of the BRS gain along with an increase of the hypoxic ventilatory gain. Present results show that CIH-induced persistent hypertension was preceded by early changes in CB chemosensory control of cardiorespiratory and autonomic function.
Collapse
Affiliation(s)
- Rodrigo Del Rio
- Laboratorio de Neurobiología, Departamento Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile ; Laboratory of Cardiorespiratory Control, Center of Biomedical Research, Universidad Autónoma de Chile Santiago, Chile
| | - Esteban A Moya
- Laboratorio de Neurobiología, Departamento Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Rodrigo Iturriaga
- Laboratorio de Neurobiología, Departamento Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| |
Collapse
|
41
|
Dergacheva O. Chronic intermittent hypoxia alters neurotransmission from lateral paragigantocellular nucleus to parasympathetic cardiac neurons in the brain stem. J Neurophysiol 2014; 113:380-9. [PMID: 25318765 DOI: 10.1152/jn.00302.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Patients with sleep-related disorders, including obstructive sleep apnea (OSA), have an increased risk of cardiovascular diseases. OSA events are more severe in rapid eye movement (REM) sleep. REM sleep further increases the risk of adverse cardiovascular events by diminishing cardioprotective parasympathetic activity. The mechanisms underlying REM sleep-related reduction in parasympathetic activity likely include activation of inhibitory input to cardiac vagal neurons (CVNs) in the brain stem originating from the lateral paragigantocellular nucleus (LPGi), a nucleus that plays a role in REM sleep control. This study tests the hypothesis that chronic intermittent hypoxia and hypercapnia (CIHH), an animal model of OSA, inhibits CVNs because of exaggeration of the GABAergic pathway from the LPGi to CVNs. GABAergic neurotransmission to CVNs evoked by electrical stimulation of the LPGi was examined with whole cell patch-clamp recordings in an in vitro brain slice preparation in rats exposed to CIHH and control rats. GABAergic synaptic events were enhanced after 4-wk CIHH in both male and female rats, to a greater degree in males. Acute hypoxia and hypercapnia (H/H) reversibly diminished the LPGi-evoked GABAergic neurotransmission to CVNs. However, GABAergic synaptic events were enhanced after acute H/H in CIHH male animals. Orexin-A elicited a reversible inhibition of LPGi-evoked GABAergic currents in control animals but evoked no significant changes in CIHH male rats. In conclusion, exaggerated inhibitory neurotransmission from the LPGi to CVNs in CIHH animals would reduce cardioprotective parasympathetic activity and enhance the risk of adverse cardiovascular events.
Collapse
Affiliation(s)
- Olga Dergacheva
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
| |
Collapse
|
42
|
Lucking EF, O'Halloran KD, Jones JFX. Increased cardiac output contributes to the development of chronic intermittent hypoxia-induced hypertension. Exp Physiol 2014; 99:1312-24. [DOI: 10.1113/expphysiol.2014.080556] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Eric F. Lucking
- UCD School of Medicine and Medical Science; University College Dublin; Dublin 4 Ireland
| | - Ken D. O'Halloran
- Department of Physiology; School of Medicine; University College Cork; Cork Ireland
| | - James F. X. Jones
- UCD School of Medicine and Medical Science; University College Dublin; Dublin 4 Ireland
| |
Collapse
|
43
|
Diogo LN, Monteiro EC. The efficacy of antihypertensive drugs in chronic intermittent hypoxia conditions. Front Physiol 2014; 5:361. [PMID: 25295010 PMCID: PMC4170135 DOI: 10.3389/fphys.2014.00361] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 09/03/2014] [Indexed: 12/22/2022] Open
Abstract
Sleep apnea/hypopnea disorders include centrally originated diseases and obstructive sleep apnea (OSA). This last condition is renowned as a frequent secondary cause of hypertension (HT). The mechanisms involved in the pathogenesis of HT can be summarized in relation to two main pathways: sympathetic nervous system stimulation mediated mainly by activation of carotid body (CB) chemoreflexes and/or asphyxia, and, by no means the least important, the systemic effects of chronic intermittent hypoxia (CIH). The use of animal models has revealed that CIH is the critical stimulus underlying sympathetic activity and hypertension, and that this effect requires the presence of functional arterial chemoreceptors, which are hyperactive in CIH. These models of CIH mimic the HT observed in humans and allow the study of CIH independently without the mechanical obstruction component. The effect of continuous positive airway pressure (CPAP), the gold standard treatment for OSA patients, to reduce blood pressure seems to be modest and concomitant antihypertensive therapy is still required. We focus this review on the efficacy of pharmacological interventions to revert HT associated with CIH conditions in both animal models and humans. First, we explore the experimental animal models, developed to mimic HT related to CIH, which have been used to investigate the effect of antihypertensive drugs (AHDs). Second, we review what is known about drug efficacy to reverse HT induced by CIH in animals. Moreover, findings in humans with OSA are cited to demonstrate the lack of strong evidence for the establishment of a first-line antihypertensive regimen for these patients. Indeed, specific therapeutic guidelines for the pharmacological treatment of HT in these patients are still lacking. Finally, we discuss the future perspectives concerning the non-pharmacological and pharmacological management of this particular type of HT.
Collapse
Affiliation(s)
- Lucilia N Diogo
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa Lisboa, Portugal
| | - Emília C Monteiro
- Centro de Estudos de Doenças Crónicas, CEDOC, NOVA Medical School/Faculdade de Ciências Médicas, Universidade Nova de Lisboa Lisboa, Portugal
| |
Collapse
|
44
|
Shin MK, Yao Q, Jun JC, Bevans-Fonti S, Yoo DY, Han W, Mesarwi O, Richardson R, Fu YY, Pasricha PJ, Schwartz AR, Shirahata M, Polotsky VY. Carotid body denervation prevents fasting hyperglycemia during chronic intermittent hypoxia. J Appl Physiol (1985) 2014; 117:765-76. [PMID: 25103977 DOI: 10.1152/japplphysiol.01133.2013] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Obstructive sleep apnea causes chronic intermittent hypoxia (IH) and is associated with impaired glucose metabolism, but mechanisms are unknown. Carotid bodies orchestrate physiological responses to hypoxemia by activating the sympathetic nervous system. Therefore, we hypothesized that carotid body denervation would abolish glucose intolerance and insulin resistance induced by chronic IH. Male C57BL/6J mice underwent carotid sinus nerve dissection (CSND) or sham surgery and then were exposed to IH or intermittent air (IA) for 4 or 6 wk. Hypoxia was administered by decreasing a fraction of inspired oxygen from 20.9% to 6.5% once per minute, during the 12-h light phase (9 a.m.-9 p.m.). As expected, denervated mice exhibited blunted hypoxic ventilatory responses. In sham-operated mice, IH increased fasting blood glucose, baseline hepatic glucose output (HGO), and expression of a rate-liming hepatic enzyme of gluconeogenesis phosphoenolpyruvate carboxykinase (PEPCK), whereas the whole body glucose flux during hyperinsulinemic euglycemic clamp was not changed. IH did not affect glucose tolerance after adjustment for fasting hyperglycemia in the intraperitoneal glucose tolerance test. CSND prevented IH-induced fasting hyperglycemia and increases in baseline HGO and liver PEPCK expression. CSND trended to augment the insulin-stimulated glucose flux and enhanced liver Akt phosphorylation at both hypoxic and normoxic conditions. IH increased serum epinephrine levels and liver sympathetic innervation, and both increases were abolished by CSND. We conclude that chronic IH induces fasting hyperglycemia increasing baseline HGO via the CSN sympathetic output from carotid body chemoreceptors, but does not significantly impair whole body insulin sensitivity.
Collapse
Affiliation(s)
- Mi-Kyung Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qiaoling Yao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jonathan C Jun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shannon Bevans-Fonti
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Doo-Young Yoo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Woobum Han
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Omar Mesarwi
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ria Richardson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ya-Yuan Fu
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Pankaj J Pasricha
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Alan R Schwartz
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Machiko Shirahata
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland;
| |
Collapse
|
45
|
Pintér A, Horváth T, Tóth A, Kádár K, Kollai M. Impaired baroreflex function is related to reduced carotid artery elasticity in patients with tetralogy of Fallot. Auton Neurosci 2014; 183:94-9. [DOI: 10.1016/j.autneu.2014.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 12/02/2013] [Accepted: 02/21/2014] [Indexed: 11/30/2022]
|
46
|
Dergacheva O, Dyavanapalli J, Piñol RA, Mendelowitz D. Chronic intermittent hypoxia and hypercapnia inhibit the hypothalamic paraventricular nucleus neurotransmission to parasympathetic cardiac neurons in the brain stem. Hypertension 2014; 64:597-603. [PMID: 24958501 DOI: 10.1161/hypertensionaha.114.03603] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Obstructive sleep apnea is associated with chronic intermittent hypoxia/hypercapnia (CIHH) episodes during sleep that heighten sympathetic and diminish parasympathetic activity to the heart. Although one population of neurons in the paraventricular nucleus of the hypothalamus strongly influences sympathetic tone and has increased activity after CIHH, little is known about the role of this pathway to parasympathetic neurons and how this network is altered in CIHH. We hypothesized that CIHH inhibits the excitatory pathway from the paraventricular nucleus of the hypothalamus to parasympathetic cardiac vagal neurons in the brain stem. To test this hypothesis, channelrhodopsin was selectively expressed, using viral vectors, in neurons in the paraventricular nucleus of the hypothalamus and channelrhodopsin-expressing fibers were photoactivated to evoke postsynaptic currents in cardiac vagal neurons in brain stem slices. Excitatory postsynaptic currents were diminished in animals exposed to CIHH. The paired-pulse and prolonged facilitation of the postsynaptic current amplitudes and frequencies evoked by paired and bursts of photoactivation of channelrhodopsin fibers, respectively, occurred in unexposed rats but were blunted in CIHH animals. In response to an acute challenge of hypoxia/hypercapnia, the amplitude of postsynaptic events was unchanged during, but increased after hypoxia/hypercapnia in unexposed animals. In contrast, postsynaptic currents were inhibited during hypoxia/hypercapnia in rats exposed to CIHH. In conclusion, the excitatory pathway to cardiac vagal neurons is diminished in response to both acute and chronic exposures to hypoxia/hypercapnia. This could elicit a reduced cardioprotective parasympathetic activity and an enhanced risk of adverse cardiovascular events in episodes of apnea and chronic obstructive sleep apnea.
Collapse
Affiliation(s)
- Olga Dergacheva
- From the Department of Pharmacology and Physiology, Department of Anesthesiology and Critical Care Medicine, The George Washington University, Washington, DC
| | - Jhansi Dyavanapalli
- From the Department of Pharmacology and Physiology, Department of Anesthesiology and Critical Care Medicine, The George Washington University, Washington, DC
| | - Ramón A Piñol
- From the Department of Pharmacology and Physiology, Department of Anesthesiology and Critical Care Medicine, The George Washington University, Washington, DC
| | - David Mendelowitz
- From the Department of Pharmacology and Physiology, Department of Anesthesiology and Critical Care Medicine, The George Washington University, Washington, DC.
| |
Collapse
|
47
|
Dyavanapalli J, Jameson H, Dergacheva O, Jain V, Alhusayyen M, Mendelowitz D. Chronic intermittent hypoxia-hypercapnia blunts heart rate responses and alters neurotransmission to cardiac vagal neurons. J Physiol 2014; 592:2799-811. [PMID: 24835174 DOI: 10.1113/jphysiol.2014.273482] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Patients with obstructive sleep apnoea experience chronic intermittent hypoxia-hypercapnia (CIHH) during sleep that elicit sympathetic overactivity and diminished parasympathetic activity to the heart, leading to hypertension and depressed baroreflex sensitivity. The parasympathetic control of heart rate arises from pre-motor cardiac vagal neurons (CVNs) located in nucleus ambiguus (NA) and dorsal motor nucleus of the vagus (DMNX). The mechanisms underlying diminished vagal control of heart rate were investigated by studying the changes in blood pressure, heart rate, and neurotransmission to CVNs evoked by acute hypoxia-hypercapnia (H-H) and CIHH. In vivo telemetry recordings of blood pressure and heart rate were obtained in adult rats during 4 weeks of CIHH exposure. Retrogradely labelled CVNs were identified in an in vitro brainstem slice preparation obtained from adult rats exposed either to air or CIHH for 4 weeks. Postsynaptic inhibitory or excitatory currents were recorded using whole cell voltage clamp techniques. Rats exposed to CIHH had increases in blood pressure, leading to hypertension, and blunted heart rate responses to acute H-H. CIHH induced an increase in GABAergic and glycinergic neurotransmission to CVNs in NA and DMNX, respectively; and a reduction in glutamatergic neurotransmission to CVNs in both nuclei. CIHH blunted the bradycardia evoked by acute H-H and abolished the acute H-H evoked inhibition of GABAergic transmission while enhancing glycinergic neurotransmission to CVNs in NA. These changes with CIHH inhibit CVNs and vagal outflow to the heart, both in acute and chronic exposures to H-H, resulting in diminished levels of cardioprotective parasympathetic activity to the heart as seen in OSA patients.
Collapse
Affiliation(s)
- Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, 2300 Eye Street NW, The George Washington University, Washington, DC, 20037, USA
| | - Heather Jameson
- Department of Pharmacology and Physiology, 2300 Eye Street NW, The George Washington University, Washington, DC, 20037, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, 2300 Eye Street NW, The George Washington University, Washington, DC, 20037, USA
| | - Vivek Jain
- Department of Pharmacology and Physiology, 2300 Eye Street NW, The George Washington University, Washington, DC, 20037, USA Department of Medicine, Medical Faculty Associates, The George Washington University, Washington, DC, 20037, USA
| | - Mona Alhusayyen
- Department of Pharmacology and Physiology, 2300 Eye Street NW, The George Washington University, Washington, DC, 20037, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, 2300 Eye Street NW, The George Washington University, Washington, DC, 20037, USA
| |
Collapse
|
48
|
Mechanism of sympathetic activation and blood pressure elevation in humans and animals following acute intermittent hypoxia. PROGRESS IN BRAIN RESEARCH 2014; 209:131-46. [PMID: 24746046 DOI: 10.1016/b978-0-444-63274-6.00007-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Sleep apnea is associated with repeated episodes of hypoxemia, causing marked increase in sympathetic nerve activity and blood pressure. Considerable evidence suggests that intermittent hypoxia (IH) resulting from apnea is the primary stimulus for sympathetic overactivity in sleep apnea patients. Several IH protocols have been developed either in animals or in humans to investigate mechanisms underlying the altered autonomic regulation of the circulation. Most of these protocols involve several days (10-40 days) of IH exposure, that is, chronic intermittent hypoxia (CIH). Recent data suggest that a single session of IH exposure, that is, acute intermittent hypoxia (AIH), is already capable of increasing tonic sympathetic nerve output (sympathetic long-term facilitation, LTF) and altering chemo- and baroreflexes with or without elevation of blood pressure. This indicates that IH alters the autonomic neurocirculatory at a very early time point, although the mechanisms underlying this neuroplasticity have not been explored in detail. The purpose of this chapter is to briefly review the effects of AIH on sympathetic LTF and alteration of autonomic reflexes in comparison with the studies from CIH studies. We will also discuss the potential central and peripheral mechanism underlying sympathetic LTF.
Collapse
|
49
|
Dergacheva O, Weigand LA, Dyavanapalli J, Mares J, Wang X, Mendelowitz D. Function and modulation of premotor brainstem parasympathetic cardiac neurons that control heart rate by hypoxia-, sleep-, and sleep-related diseases including obstructive sleep apnea. PROGRESS IN BRAIN RESEARCH 2014; 212:39-58. [PMID: 25194192 DOI: 10.1016/b978-0-444-63488-7.00003-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Parasympathetic cardiac vagal neurons (CVNs) in the brainstem dominate the control of heart rate. Previous work has determined that these neurons are inherently silent, and their activity is largely determined by synaptic inputs to CVNs that include four major types of synapses that release glutamate, GABA, glycine, or serotonin. Whereas prior reviews have focused on glutamatergic, GABAergic and glycinergic pathways, and the receptors in CVNs activated by these neurotransmitters, this review focuses on the alterations in CVN activity with hypoxia-, sleep-, and sleep-related cardiovascular diseases including obstructive sleep apnea.
Collapse
Affiliation(s)
- Olga Dergacheva
- Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, DC, USA
| | - Letitia A Weigand
- Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, DC, USA
| | - Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, DC, USA
| | - Jacquelyn Mares
- Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, DC, USA
| | - Xin Wang
- Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, DC, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, School of Medicine, George Washington University, Washington, DC, USA.
| |
Collapse
|
50
|
Knight WD, Saxena A, Shell B, Nedungadi TP, Mifflin SW, Cunningham JT. Central losartan attenuates increases in arterial pressure and expression of FosB/ΔFosB along the autonomic axis associated with chronic intermittent hypoxia. Am J Physiol Regul Integr Comp Physiol 2013; 305:R1051-8. [PMID: 24026072 PMCID: PMC3840317 DOI: 10.1152/ajpregu.00541.2012] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 09/04/2013] [Indexed: 02/02/2023]
Abstract
Chronic intermittent hypoxia (CIH) increases mean arterial pressure (MAP) and FosB/ΔFosB staining in central autonomic nuclei. To test the role of the brain renin-angiotensin system (RAS) in CIH hypertension, rats were implanted with intracerebroventricular (icv) cannulae delivering losartan (1 μg/h) or vehicle (VEH) via miniosmotic pumps and telemetry devices for arterial pressure recording. A third group was given the same dose of losartan subcutaneously (sc). Two groups of losartan-treated rats served as normoxic controls. Rats were exposed to CIH or normoxia for 7 days and then euthanized for immunohistochemistry. Intracerebroventricular losartan attenuated CIH-induced increases in arterial pressure during CIH exposure (0800-1600 during the light phase) on days 1, 6, and 7 and each day during the normoxic dark phase. FosB/ΔFosB staining in the organum vasculosum of the lamina terminalis (OVLT), median preoptic nucleus (MnPO), paraventricular nucleus of the hypothalamus (PVN), the rostral ventrolateral medulla (RVLM), and the nucleus of the solitary tract (NTS) was decreased in icv losartan-treated rats. Subcutaneous losartan also reduced CIH hypertension during the last 2 days of CIH and produced bradycardia prior to the effect on blood pressure. Following sc losartan, FosB/ΔFosB staining was reduced only in the OVLT, MnPO, PVN, and NTS. These data indicate that the central and peripheral RAS contribute to CIH-induced hypertension and transcriptional activation of autonomic nuclei and that the contribution of the central RAS is greater during the normoxic dark phase of CIH hypertension.
Collapse
Affiliation(s)
- W David Knight
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, Virginia; and
| | | | | | | | | | | |
Collapse
|