1
|
Wang X, Ribeiro C, Nilsson A, Escobar JB, Alber BR, Bethea JR, Polotsky VY, Kay MW, Schunke K, Mendelowitz D. Oxytocin Receptor Expression and Activation in Parasympathetic Brainstem Cardiac Vagal Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644204. [PMID: 40166340 PMCID: PMC11957148 DOI: 10.1101/2025.03.19.644204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Autonomic imbalance, particularly reduced activity from brainstem parasympathetic cardiac vagal neurons (CVNs) is a major characteristic of many cardiorespiratory diseases. Therapeutic approaches to selectively increase CVN activity have been limited by lack of identified selective translational targets. Recent work has shown that there is an important excitatory synaptic pathway from oxytocin (OXT) neurons in the paraventricular nucleus of the hypothalamus (PVN) to brainstem CVNs, and that OXT could provide a key selective excitation of CVNs. In clinical studies, intranasal OXT increases parasympathetic cardiac activity, autonomic balance, and reduces obstructive event durations and oxygen desaturations in obstructive sleep apnea patients. However, the mechanisms by which activation of hypothalamic OXT neurons, or intranasal OXT, increases brainstem parasympathetic cardiac activity is poorly understood. CVNs are located in two cholinergic brainstem nuclei: the nucleus ambiguus (NA) and dorsal motor nucleus of the vagus (DMNX). In this study we characterize the co-localization of OXT receptors in CVNs (OXTR), as well as non-CVN cholinergic neurons, located in the NA and DMNX nuclei. Selective chemogenetic excitation of OXTR+ CVNs was performed by expressing DREADDs with a combination of Cre and flp dependent viruses. We found that OXT receptors are highly expressed in CVNs in the DMNX and OXT increases DMNX CVN activity, but the receptors and responses are absent in CVNs in the NA. Selective chemogenetic activation of OXTR+ CVNs in the DMNX evoked a rapid and sustained bradycardia.
Collapse
|
2
|
Alanazi MM, Albaker AB, Alzaagi LA, Alsabhan JF, Alasmari F, Almutairi MM, Alharbi MS, Alasmari AF, Alqahtani F, Alsanea S. Oxytocin Protects PC12 Cells Against β-Amyloid-Induced Cell Injury. Pharmaceuticals (Basel) 2025; 18:390. [PMID: 40143166 PMCID: PMC11944556 DOI: 10.3390/ph18030390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Neurodegenerative diseases, particularly Alzheimer's disease (AD), are characterized by progressive cognitive decline and non-cognitive symptoms that significantly affect health and quality of life. Beta-amyloid (Aβ) protein accumulation is a key factor in AD pathology, leading to neuronal damage. Oxytocin (OXT), a neuropeptide with neuroprotective potential, has garnered interest owing to its ability to mitigate neurotoxicity. We hypothesized that oxytocin could protect PC12 cells from Aβ-induced cytotoxicity through antioxidant effects and modulation of apoptotic pathways (i.e., mitochondrial and MAPK pathways). In this study, we aim to assess oxytocin's protective effects on cell viability, oxidative stress, mitochondrial function, and apoptotic signaling. Methods: PC12 cells were treated with Aβ25-35 and pre-treated with varying oxytocin concentrations to assess cell viability, reactive oxygen species (ROS) generation, and mitochondrial membrane potential. Western blotting was performed to analyze the effects on mitochondrial apoptosis and MAPK pathways. Results: Oxytocin treatment significantly improved cell viability in a dose-dependent manner and reduced Aβ-induced oxidative stress and mitochondrial dysfunction. Oxytocin-treated groups exhibited decreased ROS levels, increased mitochondrial membrane potential, and modulation of apoptosis-related proteins. Oxytocin upregulated phosphorylated ERK1/2 and Bcl-2 while downregulating BAX and caspase-3, reducing the BAX/Bcl-2 ratio. Conclusions: Oxytocin effectively protects PC12 cells from Aβ-induced neurotoxicity, highlighting its potential as a therapeutic agent for AD. Further research is needed to clarify oxytocin's mechanisms and clinical implications in AD treatment.
Collapse
Affiliation(s)
- Mohammed Mufadhe Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Awatif B. Albaker
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Lamia A. Alzaagi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Jawza F. Alsabhan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M. Almutairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Metab S. Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sary Alsanea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
3
|
Habecker BA, Bers DM, Birren SJ, Chang R, Herring N, Kay MW, Li D, Mendelowitz D, Mongillo M, Montgomery JM, Ripplinger CM, Tampakakis E, Winbo A, Zaglia T, Zeltner N, Paterson DJ. Molecular and cellular neurocardiology in heart disease. J Physiol 2025; 603:1689-1728. [PMID: 38778747 PMCID: PMC11582088 DOI: 10.1113/jp284739] [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: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
This paper updates and builds on a previous White Paper in this journal that some of us contributed to concerning the molecular and cellular basis of cardiac neurobiology of heart disease. Here we focus on recent findings that underpin cardiac autonomic development, novel intracellular pathways and neuroplasticity. Throughout we highlight unanswered questions and areas of controversy. Whilst some neurochemical pathways are already demonstrating prognostic viability in patients with heart failure, we also discuss the opportunity to better understand sympathetic impairment by using patient specific stem cells that provides pathophysiological contextualization to study 'disease in a dish'. Novel imaging techniques and spatial transcriptomics are also facilitating a road map for target discovery of molecular pathways that may form a therapeutic opportunity to treat cardiac dysautonomia.
Collapse
Affiliation(s)
- Beth A Habecker
- Department of Chemical Physiology & Biochemistry, Department of Medicine Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | - Susan J Birren
- Department of Biology, Volen Center for Complex Systems, Brandeis University, Waltham, MA, USA
| | - Rui Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Johanna M Montgomery
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | | | - Annika Winbo
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Nadja Zeltner
- Departments of Biochemistry and Molecular Biology, Cell Biology, and Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Mazza M, Veneziani G, Lisci FM, Morini S, Traversi G, Sfratta G, Brisi C, Anesini MB, Bardi F, Benini E, Calderoni C, Chisari L, Crupi A, De Chiara E, Lo Giudice L, Onori L, Sessa I, Balocchi M, Pola R, Gaetani E, Simeoni B, Franceschi F, Sani G, Covino M, Lai C, Romagnoli E, Marano G. Mental Illness Strikes at the Heart: Impact of Psychiatric Diseases on Ventricular Ejection Fraction in Patients with Acute Coronary Syndromes. Life (Basel) 2025; 15:340. [PMID: 40141685 PMCID: PMC11944072 DOI: 10.3390/life15030340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/09/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Mental illnesses can have a significant impact on individuals experiencing acute coronary syndromes (ACS). Mental illnesses are associated with an increased cardiovascular risk profile and early onset of cardiovascular disease. A critical aspect of this interplay is the effect of psychiatric conditions on left ventricular ejection fraction (LVEF), a key parameter in evaluating cardiac function and predicting long-term outcomes in ACS patients. The present single-center, retrospective study investigated the associations between psychiatric conditions and cardiac function, with a focus on LVEF in ACS patients. The inclusion criteria were Italian nationality and 30 years or older. One hundred and sixty-four patients without (Mage = 68.8 ± 10.6, 62 females) and 161 patients with a psychiatric diagnosis (Mage = 68.4 ± 13.7, 63 females) were enrolled. The data collected included sociodemographic variables, psychiatric diagnoses, LVEF, ACS type (STEMI/NSTEMI), smoking status, previous interventions, and pharmacological treatments. Statistical analyses included chi-square, t-tests, ANOVAs, and ANCOVA to assess differences across groups. Findings revealed lower LVEF in patients with a psychiatric diagnosis compared to patients without a psychiatric diagnosis (p = 0.004, d = 0.36). Patients without a psychiatric diagnosis were associated with NSTEMI (p = 0.047, φ = 0.11), hypertension (p = 0.003, φ = -0.16), and dyslipidemia (p = 0.022, φ = -0.13). In contrast, patients with a psychiatric diagnosis were associated with STEMI (p = 0.047, φ = 0.11), neurological dysfunction (p = 0.014, φ = 0.14), and chronic obstructive pulmonary disease (p = 0.010, φ = 0.14). Among psychiatric diagnoses, anxiety disorders were associated with lower LVEF compared to substance abuse disorders (p = 0.012, d = -0.81). The findings underscore the complex relationship between mental illness and cardiac function, emphasising the need to integrate psychiatric evaluations into cardiology care to optimise the management of both mental and cardiovascular health. This study has several limitations, including its design, which prevents causal conclusions, and the use of convenience sampling, which limits the generalizability of the findings.
Collapse
Affiliation(s)
- Marianna Mazza
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Giorgio Veneziani
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University, Via degli Apuli 1, 00185 Rome, Italy
| | - Francesco Maria Lisci
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Sofia Morini
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy (E.R.)
| | - Gianandrea Traversi
- Unit of Medical Genetics, Department of Laboratory Medicine, Ospedale Isola Tiberina-Gemelli Isola, Via di Ponte Quattro Capi 39, 00186 Rome, Italy
| | - Greta Sfratta
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Caterina Brisi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Maria Benedetta Anesini
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Francesca Bardi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Elisabetta Benini
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Claudia Calderoni
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Luca Chisari
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Arianna Crupi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Emanuela De Chiara
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Luca Lo Giudice
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Luca Onori
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Ilenia Sessa
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Marta Balocchi
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Roberto Pola
- Section of Internal Medicine and Thromboembolic Diseases, Department of Internal Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Eleonora Gaetani
- Department of Translational Medicine and Surgery, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
- Unit of Internal Medicine, Cristo Re Hospital, Via delle Calasanziane 25, 00167 Rome, Italy
| | - Benedetta Simeoni
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (B.S.); (M.C.)
| | - Francesco Franceschi
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (B.S.); (M.C.)
| | - Gabriele Sani
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Marcello Covino
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy; (B.S.); (M.C.)
| | - Carlo Lai
- Department of Dynamic and Clinical Psychology, and Health Studies, Sapienza University, Via degli Apuli 1, 00185 Rome, Italy
| | - Enrico Romagnoli
- Department of Cardiovascular Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy (E.R.)
| | - Giuseppe Marano
- Unit of Psychiatry, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; (M.M.); (F.M.L.); (C.B.); (M.B.A.); (E.D.C.)
- Department of Neurosciences, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
5
|
Salazar-Ardiles C, Cornejo C, Paz C, Vasquez-Muñoz M, Arce-Alvarez A, Rodriguez-Fernandez M, Millet GP, Izquierdo M, Andrade DC. Effect of chronic exogenous oxytocin administration on exercise performance and cardiovagal control in hypobaric hypoxia in rats. Biol Res 2024; 57:88. [PMID: 39578887 PMCID: PMC11585223 DOI: 10.1186/s40659-024-00573-3] [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: 06/06/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Outstanding exercise performance has been associated with an exacerbated vagal outflow. Nevertheless, during high-altitude hypobaric-hypoxia (HH), there is a baroreflex-dependent parasympathetic withdrawal and exercise performance deterioration. Notably, vagal control is pivotal in exercise performance, and exogenous oxytocin (OXY) administration has been shown to enhance parasympathetic drive; however, no evidence shows their role in exercise performance during HH. Then, this study aimed to examine the effect of prolonged exogenous oxytocin (OXY) administration on exercise performance during hypobaric hypoxia (HH) in rats. RESULTS A vehicle group (n = 6) and an OXY group (n = 6) performed incremental exercise and baroreflex tests during both normobaric normoxia (NN) and HH (PO2: 100 mmHg, simulated 3,500 m) prior (pre-) and after (post-) 14 days of administration. The results showed that at pre-, there were no significant differences in exercise performance between the two groups, while at post-, the OXY group exhibited similar performance between NN and HH, while the Vehicle group maintained a significant decline in performance at HH compared to NN. At post-, the Vehicle group also demonstrated a reset in the baroreflex and a worse bradycardic response in HH, which was reversed in the OXY group, while the hypoxic ventilatory response was similar in both groups. CONCLUSION The findings suggest prolonged OXY administration prevents impaired exercise performance and vagal control during short-term HH.
Collapse
Affiliation(s)
- Camila Salazar-Ardiles
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
- Navarrabiomed, Hospital Universitario de Navarra (UHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Navarra, Spain
| | - Carlos Cornejo
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Cristobal Paz
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile
| | - Manuel Vasquez-Muñoz
- Dirección de Docencia de Especialidades Médicas, Dirección de Postgrado, Facultad de Medicina y Ciencias de la Salud, Universidad Mayor, Santiago, Chile
| | - Alexis Arce-Alvarez
- Escuela de Kinesiología, Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastian, Santiago, Chile
| | - Maria Rodriguez-Fernandez
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine, and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gregoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, CH-1015, Switzerland
| | - Mikel Izquierdo
- Navarrabiomed, Hospital Universitario de Navarra (UHN), Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Navarra, Spain
| | - David C Andrade
- Exercise Applied Physiology Laboratory, Centro de Investigación en Fisiología y Medicina de Altura (FIMEDALT), Departamento Biomedico, Facultad de Ciencias de la Salud, Universidad de Antofagasta, Antofagasta, Chile.
| |
Collapse
|
6
|
Hornung E, Robbins S, Srivastava A, Achanta S, Chen J, Cheng ZJ, Schwaber J, Vadigepalli R. Neuromodulatory co-expression in cardiac vagal motor neurons of the dorsal motor nucleus of the vagus. iScience 2024; 27:110549. [PMID: 39171288 PMCID: PMC11338141 DOI: 10.1016/j.isci.2024.110549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/31/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
Vagal innervation is well known to be crucial to the maintenance of cardiac health, and to protect and recover the heart from injury. Only recently has this role been shown to depend on the activity of the underappreciated dorsal motor nucleus of the vagus (DMV). By combining neural tracing, transcriptomics, and anatomical mapping in male and female Sprague-Dawley rats, we characterize cardiac-specific neuronal phenotypes in the DMV. We find that the DMV cardiac-projecting neurons differentially express pituitary adenylate cyclase-activating polypeptide (PACAP), cocaine- and amphetamine-regulated transcript (CART), and synucleins, as well as evidence that they participate in neuromodulatory co-expression involving catecholamines. The significance of these findings is enhanced by previous knowledge of the role of PACAP at the heart and of the other neuromodulators in peripheral vagal targets.
Collapse
Affiliation(s)
- Eden Hornung
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shaina Robbins
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Ankita Srivastava
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sirisha Achanta
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, BMS Building 20, Room 230, 4110 Libra Drive, Orlando, FL 32816, USA
| | - Zixi Jack Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, BMS Building 20, Room 230, 4110 Libra Drive, Orlando, FL 32816, USA
| | - James Schwaber
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
7
|
Cheung EC, Nilsson A, Venter I, Kowalik G, Ribeiro C, Rodriguez J, Kuraoka K, Russo R, Escobar JB, Alber BR, Mendelowitz D, Kay MW, Schunke KJ. Sex differences in cardiac transcriptomic response to neonatal sleep apnea. Physiol Rep 2024; 12:e16110. [PMID: 38981849 PMCID: PMC11233197 DOI: 10.14814/phy2.16110] [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: 03/25/2024] [Revised: 05/03/2024] [Accepted: 05/31/2024] [Indexed: 07/11/2024] Open
Abstract
Pediatric obstructive sleep apnea poses a significant health risk, with potential long-term consequences on cardiovascular health. This study explores the dichotomous nature of neonatal cardiac response to chronic intermittent hypoxia (CIH) between males and females, aiming to fill a critical knowledge gap in the understanding of sex-specific cardiovascular consequences of sleep apnea in early life. Neonates were exposed to CIH until p28 and underwent comprehensive in vivo physiological assessments, including whole-body plethysmography, treadmill stress-tests, and echocardiography. Results indicated that male CIH rats weighed 13.7% less than age-matched control males (p = 0.0365), while females exhibited a mild yet significant increased respiratory drive during sleep (93.94 ± 0.84 vs. 95.31 ± 0.81;p = 0.02). Transcriptomic analysis of left ventricular tissue revealed a substantial sex-based difference in the cardiac response to CIH, with males demonstrating a more pronounced alteration in gene expression compared to females (5986 vs. 3174 genes). The dysregulated miRNAs in males target metabolic genes, potentially predisposing the heart to altered metabolism and substrate utilization. Furthermore, CIH in males was associated with thinner left ventricular walls and dysregulation of genes involved in the cardiac action potential, possibly predisposing males to CIH-related arrhythmia. These findings emphasize the importance of considering sex-specific responses in understanding the cardiovascular implications of pediatric sleep apnea.
Collapse
Affiliation(s)
- Emily C. Cheung
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Anna Nilsson
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Ian Venter
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Grant Kowalik
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Caitlin Ribeiro
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Jeannette Rodriguez
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Kiralee Kuraoka
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| | - Rebekah Russo
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Joan B. Escobar
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Bridget R. Alber
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - David Mendelowitz
- Department of Pharmacology and PhysiologyThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Matthew W. Kay
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
| | - Kathryn J. Schunke
- Department of Biomedical EngineeringThe George Washington UniversityWashingtonDistrict of ColumbiaUSA
- Department of Anatomy, Biochemistry & PhysiologyUniversity of HawaiiHonoluluHawaiiUSA
| |
Collapse
|
8
|
de Barcellos Filho PG, Dantzler HA, Hasser EM, Kline DD. Oxytocin and corticotropin-releasing hormone exaggerate nucleus tractus solitarii neuronal and synaptic activity following chronic intermittent hypoxia. J Physiol 2024; 602:3375-3400. [PMID: 38698722 PMCID: PMC11251298 DOI: 10.1113/jp286069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 03/28/2024] [Indexed: 05/05/2024] Open
Abstract
Chronic intermittent hypoxia (CIH) in rodents mimics the hypoxia-induced elevation of blood pressure seen in individuals experiencing episodic breathing. The brainstem nucleus tractus solitarii (nTS) is the first site of visceral sensory afferent integration, and thus is critical for cardiorespiratory homeostasis and its adaptation during a variety of stressors. In addition, the paraventricular nucleus of the hypothalamus (PVN), in part through its nTS projections that contain oxytocin (OT) and/or corticotropin-releasing hormone (CRH), contributes to cardiorespiratory regulation. Within the nTS, these PVN-derived neuropeptides alter nTS activity and the cardiorespiratory response to hypoxia. Nevertheless, their contribution to nTS activity after CIH is not fully understood. We hypothesized that OT and CRH would increase nTS activity to a greater extent following CIH, and co-activation of OT+CRH receptors would further magnify nTS activity. Our data show that compared to their normoxic controls, 10 days' CIH exaggerated nTS discharge, excitatory synaptic currents and Ca2+ influx in response to CRH, which were further enhanced by the addition of OT. CIH increased the tonic functional contribution of CRH receptors, which occurred with elevation of mRNA and protein. Together, our data demonstrate that intermittent hypoxia exaggerates the expression and function of neuropeptides on nTS activity. KEY POINTS: Episodic breathing and chronic intermittent hypoxia (CIH) are associated with autonomic dysregulation, including elevated sympathetic nervous system activity. Altered nucleus tractus solitarii (nTS) activity contributes to this response. Neurons originating in the paraventricular nucleus (PVN), including those containing oxytocin (OT) and corticotropin-releasing hormone (CRH), project to the nTS, and modulate the cardiorespiratory system. Their role in CIH is unknown. In this study, we focused on OT and CRH individually and together on nTS activity from rats exposed to either CIH or normoxia control. We show that after CIH, CRH alone and with OT increased to a greater extent overall nTS discharge, neuronal calcium influx, synaptic transmission to second-order nTS neurons, and OT and CRH receptor expression. These results provide insights into the underlying circuits and mechanisms contributing to autonomic dysfunction during periods of episodic breathing.
Collapse
Affiliation(s)
- Procopio Gama de Barcellos Filho
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - Heather A. Dantzler
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - Eileen M. Hasser
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| | - David D. Kline
- Department of Biomedical Sciences, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
- Department of Dalton Cardiovascular Research Center, University of Missouri, 1500 Research Park Dr., Columbia, MO 65211, USA
| |
Collapse
|
9
|
Santiago HP, Leite LHR, Lima PMA, Fóscolo DRC, Natali AJ, Prímola-Gomes TN, Szawka RE, Coimbra CC. Effects of physical training on hypothalamic neuronal activation and expressions of vasopressin and oxytocin in SHR after running until fatigue. Pflugers Arch 2024; 476:365-377. [PMID: 38308122 DOI: 10.1007/s00424-024-02916-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/04/2024]
Abstract
To assess the influence of physical training on neuronal activation and hypothalamic expression of vasopressin and oxytocin in spontaneously hypertensive rats (SHR), untrained and trained normotensive rats and SHR were submitted to running until fatigue while internal body and tail temperatures were recorded. Hypothalamic c-Fos expression was evaluated in thermoregulatory centers such as the median preoptic nucleus (MnPO), medial preoptic nucleus (mPOA), paraventricular nucleus of the hypothalamus (PVN), and supraoptic nucleus (SON). The PVN and the SON were also investigated for vasopressin and oxytocin expressions. Although exercise training improved the workload performed by the animals, it was reduced in SHR and followed by increased internal body temperature due to tail vasodilation deficit. Physical training enhanced c-Fos expression in the MnPO, mPOA, and PVN of both strains, and these responses were attenuated in SHR. Vasopressin immunoreactivity in the PVN was also increased by physical training to a lesser extent in SHR. The already-reduced oxytocin expression in the PVN of SHR was increased in response to physical training. Within the SON, neuronal activation and the expressions of vasopressin and oxytocin were reduced by hypertension and unaffected by physical training. The data indicate that physical training counterbalances in part the negative effect of hypertension on hypothalamic neuronal activation elicited by exercise, as well as on the expression of vasopressin and oxytocin. These hypertension features seem to negatively influence the workload performed by SHR due to the hyperthermia derived from the inability of physical training to improve heat dissipation through skin vasodilation.
Collapse
Affiliation(s)
- Henrique P Santiago
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Laura H R Leite
- Departamento de Biofísica e Fisiologia, Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora, Minas Gerais, Brazil
| | - Paulo M A Lima
- Núcleo de Pesquisa da Faculdade de Medicina da Universidade de Rio Verde, Universidade de Rio Verde, Campus Goiânia, Goiânia, Brazil
| | - Daniela R C Fóscolo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Antônio José Natali
- Departamento de Educação Física, Universidade Federal de Viçosa, Viçosa, Minas Gerais, Brazil
| | | | - Raphael E Szawka
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cândido C Coimbra
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
10
|
Hare MT, Carter ME, Swoap SJ. Activation of oxytocinergic neurons enhances torpor in mice. J Comp Physiol B 2024; 194:95-104. [PMID: 38170253 DOI: 10.1007/s00360-023-01528-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Mus musculus enters a torpid state in response to caloric restriction in sub-thermoneutral ambient temperatures. This torpid state is characterized by an adaptive and controlled decrease in metabolic rate, heart rate, body temperature, and activity. Previous research has identified the paraventricular nucleus (PVN) within the hypothalamus, a region containing oxytocin neurons, as a location that is active during torpor onset. We hypothesized that oxytocin neurons within the PVN are part of this neural circuit and that activation of oxytocin neurons would deepen and lengthen torpor bouts. We report that activation of oxytocin neurons alone is not sufficient to induce a torpor-like state in the fed mouse, with no significant difference in body temperature or heart rate upon activation of oxytocin neurons. However, we found that activation of oxytocin neurons prior to the onset of daily torpor both deepens and lengthens the subsequent bout, with a 1.7 ± 0.4 °C lower body temperature and a 135 ± 32 min increase in length. We therefore conclude that oxytocin neurons are involved in the neural circuitry controlling daily torpor in the mouse.
Collapse
Affiliation(s)
- Maia T Hare
- Department of Biology, Williams College, Williamstown, MA, 01267, USA
- Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA
| | - Matthew E Carter
- Department of Biology, Williams College, Williamstown, MA, 01267, USA
| | - Steven J Swoap
- Department of Biology, Williams College, Williamstown, MA, 01267, USA.
| |
Collapse
|
11
|
Schunke KJ, Rodriguez J, Dyavanapalli J, Schloen J, Wang X, Escobar J, Kowalik G, Cheung EC, Ribeiro C, Russo R, Alber BR, Dergacheva O, Chen SW, Murillo-Berlioz AE, Lee KB, Trachiotis G, Entcheva E, Brantner CA, Mendelowitz D, Kay MW. Outcomes of hypothalamic oxytocin neuron-driven cardioprotection after acute myocardial infarction. Basic Res Cardiol 2023; 118:43. [PMID: 37801130 PMCID: PMC10558415 DOI: 10.1007/s00395-023-01013-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/07/2023]
Abstract
Altered autonomic balance is a hallmark of numerous cardiovascular diseases, including myocardial infarction (MI). Although device-based vagal stimulation is cardioprotective during chronic disease, a non-invasive approach to selectively stimulate the cardiac parasympathetic system immediately after an infarction does not exist and is desperately needed. Cardiac vagal neurons (CVNs) in the brainstem receive powerful excitation from a population of neurons in the paraventricular nucleus (PVN) of the hypothalamus that co-release oxytocin (OXT) and glutamate to excite CVNs. We tested if chemogenetic activation of PVN-OXT neurons following MI would be cardioprotective. The PVN of neonatal rats was transfected with vectors to selectively express DREADDs within OXT neurons. At 6 weeks of age, an MI was induced and DREADDs were activated with clozapine-N-oxide. Seven days following MI, patch-clamp electrophysiology confirmed the augmented excitatory neurotransmission from PVN-OXT neurons to downstream nuclei critical for parasympathetic activity with treatment (43.7 ± 10 vs 86.9 ± 9 pA; MI vs. treatment), resulting in stark improvements in survival (85% vs. 95%; MI vs. treatment), inflammation, fibrosis assessed by trichrome blue staining, mitochondrial function assessed by Seahorse assays, and reduced incidence of arrhythmias (50% vs. 10% cumulative incidence of ventricular fibrillation; MI vs. treatment). Myocardial transcriptomic analysis provided molecular insight into potential cardioprotective mechanisms, which revealed the preservation of beneficial signaling pathways, including muscarinic receptor activation, in treated animals. These comprehensive results demonstrate that the PVN-OXT network could be a promising therapeutic target to quickly activate beneficial parasympathetic-mediated cellular pathways within the heart during the early stages of infarction.
Collapse
Affiliation(s)
- Kathryn J Schunke
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA.
- Department of Anatomy, Biochemistry and Physiology, University of Hawaii, 651 Ilalo St, Honolulu, HI, BSB 211 96813, USA.
| | - Jeannette Rodriguez
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - John Schloen
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Xin Wang
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Joan Escobar
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Grant Kowalik
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Emily C Cheung
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Caitlin Ribeiro
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Rebekah Russo
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Bridget R Alber
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA
| | - Sheena W Chen
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Alejandro E Murillo-Berlioz
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Kyongjune B Lee
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Gregory Trachiotis
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
- Division of Cardiothoracic Surgery and Cardiothoracic Research, Veterans Affairs Medical Center, 50 Irving St. NW, Washington, DC, 20422, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA
| | - Christine A Brantner
- The GWU Nanofabrication and Imaging Center, 800 22nd Street NW, Washington, DC, 20052, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Suite 640 Ross Hall, 2300 Eye St. NW, Washington, DC, 20052, USA.
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Suite 5000 Science and Engineering Hall, 800 22nd Street NW, Washington, DC, 20052, USA.
| |
Collapse
|
12
|
Ruyle BC, Lima-Silveira L, Martinez D, Cummings KJ, Heesch CM, Kline DD, Hasser EM. Paraventricular nucleus projections to the nucleus tractus solitarii are essential for full expression of hypoxia-induced peripheral chemoreflex responses. J Physiol 2023; 601:4309-4336. [PMID: 37632733 DOI: 10.1113/jp284907] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/13/2023] [Indexed: 08/28/2023] Open
Abstract
The hypothalamic paraventricular nucleus (PVN) is essential to peripheral chemoreflex neurocircuitry, but the specific efferent pathways utilized are not well defined. The PVN sends dense projections to the nucleus tractus solitarii (nTS), which exhibits neuronal activation following a hypoxic challenge. We hypothesized that nTS-projecting PVN (PVN-nTS) neurons contribute to hypoxia-induced nTS neuronal activation and cardiorespiratory responses. To selectively target PVN-nTS neurons, rats underwent bilateral nTS nanoinjection of retrogradely transported adeno-associated virus (AAV) driving Cre recombinase expression. We then nanoinjected into PVN AAVs driving Cre-dependent expression of Gq or Gi designer receptors exclusively activated by designer drugs (DREADDs) to test the degree that selective activation or inhibition, respectively, of the PVN-nTS pathway affects the hypoxic ventilatory response (HVR) of conscious rats. We used immunohistochemistry for Fos and extracellular recordings to examine how DREADD activation influences PVN-nTS neuronal activation by hypoxia. Pathway activation enhanced the HVR at moderate hypoxic intensities and increased PVN and nTS Fos immunoreactivity in normoxia and hypoxia. In contrast, PVN-nTS inhibition reduced both the HVR and PVN and nTS neuronal activation following hypoxia. To further confirm selective pathway effects on central cardiorespiratory output, rats underwent hypoxia before and after bilateral nTS nanoinjections of C21 to activate or inhibit PVN-nTS terminals. PVN terminal activation within the nTS enhanced tachycardic, sympathetic and phrenic (PhrNA) nerve activity responses to hypoxia whereas inhibition attenuated hypoxia-induced increases in nTS neuronal action potential discharge and PhrNA. The results demonstrate the PVN-nTS pathway enhances nTS neuronal activation and is necessary for full cardiorespiratory responses to hypoxia. KEY POINTS: The hypothalamic paraventricular nucleus (PVN) contributes to peripheral chemoreflex cardiorespiratory responses, but specific PVN efferent pathways are not known. The nucleus tractus solitarii (nTS) is the first integration site of the peripheral chemoreflex, and the nTS receives dense projections from the PVN. Selective GqDREADD activation of the PVN-nTS pathway was shown to enhance ventilatory responses to hypoxia and activation (Fos immunoreactivity (IR)) of nTS neurons in conscious rats, augmenting the sympathetic and phrenic nerve activity (SSNA and PhrNA) responses to hypoxia in anaesthetized rats. Selective GiDREADD inhibition of PVN-nTS neurons attenuates ventilatory responses, nTS neuronal Fos-IR, action potential discharge and PhrNA responses to hypoxia. These results demonstrate that a projection from the PVN to the nTS is critical for full chemoreflex responses to hypoxia.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
| | - Ludmila Lima-Silveira
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Diana Martinez
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Kevin J Cummings
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Cheryl M Heesch
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - David D Kline
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Eileen M Hasser
- Department of Biomedical Sciences, University of Missouri, Columbia, MO, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
13
|
Xi H, Li X, Zhou Y, Sun Y. The Regulatory Effect of the Paraventricular Nucleus on Hypertension. Neuroendocrinology 2023; 114:1-13. [PMID: 37598678 DOI: 10.1159/000533691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 08/17/2023] [Indexed: 08/22/2023]
Abstract
Hypertension is among the most harmful factors of cardiovascular and cerebrovascular diseases and poses an urgent problem for the development of human society. In addition to previous studies on its pathogenesis focusing on the peripheral sympathetic nervous system, investigating the central causes of high blood pressure involving the neuroendocrine and neuroinflammatory mechanisms of the hypothalamic paraventricular nucleus (PVN) is paramount. This nucleus is considered to regulate the output of neurohormones and sympathetic nerve activity. In this article, we focussed on the neuroendocrine mechanism, primarily exploring the specific contributions and interactions of various neurons and neuroendocrine hormones, including GABAergic and glutamatergic neurons, nitric oxide, arginine vasopressin, oxytocin, and the renin-angiotensin system. Additionally, the neuroinflammatory mechanism in the PVN was discussed, encompassing microglia, reactive oxygen species, inflammatory factors, and pathways, as well as immune connections between the brain and extracerebral organs. Notably, the two central mechanisms involved in the PVN not only exist independently but also communicate with each other, jointly maintaining the hypertensive state of the body. Furthermore, we introduce well-known molecules and signal transduction pathways within the PVN that can play a regulatory role in the two mechanisms to provide a basis and inspire ideas for further research.
Collapse
Affiliation(s)
- Hanyu Xi
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Xingru Li
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yun Zhou
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
- Department of Nephrology, Shanxi Provincial Integrated Traditional Chinese Medicine and Western Medicine Hospital, Taiyuan, China
| | - Yaojun Sun
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, China
- School of Basic Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
Ramirez JM, Carroll MS, Burgraff N, Rand CM, Weese-Mayer DE. A narrative review of the mechanisms and consequences of intermittent hypoxia and the role of advanced analytic techniques in pediatric autonomic disorders. Clin Auton Res 2023; 33:287-300. [PMID: 37326924 DOI: 10.1007/s10286-023-00958-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/25/2023] [Indexed: 06/17/2023]
Abstract
Disorders of autonomic functions are typically characterized by disturbances in multiple organ systems. These disturbances are often comorbidities of common and rare diseases, such as epilepsy, sleep apnea, Rett syndrome, congenital heart disease or mitochondrial diseases. Characteristic of many autonomic disorders is the association with intermittent hypoxia and oxidative stress, which can cause or exaggerate a variety of other autonomic dysfunctions, making the treatment and management of these syndromes very complex. In this review we discuss the cellular mechanisms by which intermittent hypoxia can trigger a cascade of molecular, cellular and network events that result in the dysregulation of multiple organ systems. We also describe the importance of computational approaches, artificial intelligence and the analysis of big data to better characterize and recognize the interconnectedness of the various autonomic and non-autonomic symptoms. These techniques can lead to a better understanding of the progression of autonomic disorders, ultimately resulting in better care and management.
Collapse
Affiliation(s)
- Jan-Marino Ramirez
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA, 98101, USA.
- Departments of Neurological Surgery and Pediatrics, University of Washington School of Medicine, 1900 Ninth Avenue, Seattle, WA, 98101, USA.
| | - Michael S Carroll
- Data Analytics and Reporting, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Nicholas Burgraff
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA, 98101, USA
| | - Casey M Rand
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Debra E Weese-Mayer
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Division of Autonomic Medicine, Stanley Manne Children's Research Institute at Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| |
Collapse
|
15
|
Dergacheva O, Polotsky VY, Mendelowitz D. Oxytocin mediated excitation of hypoglossal motoneurons: implications for treating obstructive sleep apnea. Sleep 2023; 46:zsad009. [PMID: 36846973 PMCID: PMC10091096 DOI: 10.1093/sleep/zsad009] [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: 08/24/2022] [Revised: 11/21/2022] [Indexed: 03/01/2023] Open
Abstract
Clinical studies have shown that oxytocin administered intranasally (IN) decreased the incidence and duration of obstructive events in patients with obstructive sleep apnea (OSA). Although the mechanisms by which oxytocin promotes these beneficial effects are unknown, one possible target of oxytocin could be the excitation of tongue-projecting hypoglossal motoneurons in the medulla, that exert central control of upper airway patency. This study tested the hypothesis that IN oxytocin enhances tongue muscle activity via the excitation of hypoglossal motoneurons projecting to tongue protrudor muscles (PMNs). To test this hypothesis we performed in vivo and in vitro electrophysiological studies in C57BL6/J mice as well as fluorescent imaging studies in transgenic mice in which neurons that express oxytocin receptors co-express fluorescent protein. IN oxytocin significantly increased the amplitude of inspiratory-related tongue muscle activity. This effect was abolished by severing the medial branch of hypoglossal nerve that innervates PMNs of the tongue. Oxytocin receptor-positive neurons were more prevalent in the population of PMNs than in retractor-projecting hypoglossal motoneurons (RMNs). Oxytocin administration increased action potential firing in PMNs, but had no significant effect on firing activity in RMNs. In conclusion, IN oxytocin stimulates respiratory-relating tongue muscle activity likely acting on central hypoglossal motoneurons that provide tongue protrusion and upper airway opening. This mechanism may play a role in oxytocin-induced reductions in upper airway obstructions in patients with OSA.
Collapse
Affiliation(s)
- Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| | - Vsevolod Y Polotsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC 20037, USA
| |
Collapse
|
16
|
Rodriguez J, Escobar JB, Cheung EC, Kowalik G, Russo R, Dyavanapalli J, Alber BR, Harral G, Gill A, Melkie M, Jain V, Schunke KJ, Mendelowitz D, Kay MW. Hypothalamic Oxytocin Neuron Activation Attenuates Intermittent Hypoxia-Induced Hypertension and Cardiac Dysfunction in an Animal Model of Sleep Apnea. Hypertension 2023; 80:882-894. [PMID: 36794581 PMCID: PMC10027399 DOI: 10.1161/hypertensionaha.122.20149] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Obstructive sleep apnea is a prevalent and poorly treated cardiovascular disease that leads to hypertension and autonomic imbalance. Recent studies that restore cardiac parasympathetic tone using selective activation of hypothalamic oxytocin neurons have shown beneficial cardiovascular outcomes in animal models of cardiovascular disease. This study aimed to determine if chemogenetic activation of hypothalamic oxytocin neurons in animals with existing obstructive sleep apnea-induced hypertension would reverse or blunt the progression of autonomic and cardiovascular dysfunction. METHODS Two groups of rats were exposed to chronic intermittent hypoxia (CIH), a model of obstructive sleep apnea, for 4 weeks to induce hypertension. During an additional 4 weeks of exposure to CIH, 1 group was treated with selective activation of hypothalamic oxytocin neurons while the other group was untreated. RESULTS Hypertensive animals exposed to CIH and treated with daily hypothalamic oxytocin neuron activation had lower blood pressure, faster heart rate recovery times after exercise, and improved indices of cardiac function compared with untreated hypertensive animals. Microarray analysis suggested that, compared with treated animals, untreated animals had gene expression profiles associated with cellular stress response activation, hypoxia-inducible factor stabilization, and myocardial extracellular matrix remodeling and fibrosis. CONCLUSIONS In animals already presenting with CIH-induced hypertension, chronic activation of hypothalamic oxytocin neurons blunted the progression of hypertension and conferred cardioprotection after an additional 4 weeks of CIH exposure. These results have significant clinical translation for the treatment of cardiovascular disease in patients with obstructive sleep apnea.
Collapse
Affiliation(s)
- Jeannette Rodriguez
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| | - Joan B Escobar
- Department of Pharmacology and Physiology (J.B.E., E.C.C., J.D., D.M.), The George Washington University, Washington, DC
| | - Emily C Cheung
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
- Department of Pharmacology and Physiology (J.B.E., E.C.C., J.D., D.M.), The George Washington University, Washington, DC
| | - Grant Kowalik
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| | - Rebekah Russo
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| | - Jhansi Dyavanapalli
- Department of Pharmacology and Physiology (J.B.E., E.C.C., J.D., D.M.), The George Washington University, Washington, DC
| | - Bridget R Alber
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| | - Grey Harral
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| | - Aman Gill
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| | - Makeda Melkie
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| | - Vivek Jain
- Department of Medicine (V.J.), The George Washington University, Washington, DC
| | - Kathryn J Schunke
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
- Department of Anatomy, Biochemistry & Physiology, University of Hawaii, Honolulu, HI (K.J.S.)
| | - David Mendelowitz
- Department of Pharmacology and Physiology (J.B.E., E.C.C., J.D., D.M.), The George Washington University, Washington, DC
| | - Matthew W Kay
- Department of Biomedical Engineering (J.R., E.C.C., G.K., R.R., B.R.A., G.H., A.G., M.M., K.J.S., M.W.K.), The George Washington University, Washington, DC
| |
Collapse
|
17
|
Burtscher J, Niedermeier M, Hüfner K, van den Burg E, Kopp M, Stoop R, Burtscher M, Gatterer H, Millet GP. The interplay of hypoxic and mental stress: Implications for anxiety and depressive disorders. Neurosci Biobehav Rev 2022; 138:104718. [PMID: 35661753 DOI: 10.1016/j.neubiorev.2022.104718] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Adequate oxygen supply is essential for the human brain to meet its high energy demands. Therefore, elaborate molecular and systemic mechanism are in place to enable adaptation to low oxygen availability. Anxiety and depressive disorders are characterized by alterations in brain oxygen metabolism and of its components, such as mitochondria or hypoxia inducible factor (HIF)-pathways. Conversely, sensitivity and tolerance to hypoxia may depend on parameters of mental stress and the severity of anxiety and depressive disorders. Here we discuss relevant mechanisms of adaptations to hypoxia, as well as their involvement in mental stress and the etiopathogenesis of anxiety and depressive disorders. We suggest that mechanisms of adaptations to hypoxia (including metabolic responses, inflammation, and the activation of chemosensitive brain regions) modulate and are modulated by stress-related pathways and associated psychiatric diseases. While severe chronic hypoxia or dysfunctional hypoxia adaptations can contribute to the pathogenesis of anxiety and depressive disorders, harnessing controlled responses to hypoxia to increase cellular and psychological resilience emerges as a novel treatment strategy for these diseases.
Collapse
Affiliation(s)
- Johannes Burtscher
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| | - Martin Niedermeier
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Katharina Hüfner
- Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, University Clinic for Psychiatry II, Innsbruck Medical University, Innsbruck, Austria
| | - Erwin van den Burg
- Department of Psychiatry, Center of Psychiatric Neuroscience (CNP), University Hospital of Lausanne (CHUV), Prilly, Lausanne, Switzerland
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Ron Stoop
- Department of Psychiatry, Center of Psychiatric Neuroscience (CNP), University Hospital of Lausanne (CHUV), Prilly, Lausanne, Switzerland
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Hannes Gatterer
- Institute of Mountain Emergency Medicine, Eurac Research, Bolzano, Italy
| | - Grégoire P Millet
- Institute of Sport Sciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Goh KK, Chen CYA, Wu TH, Chen CH, Lu ML. Crosstalk between Schizophrenia and Metabolic Syndrome: The Role of Oxytocinergic Dysfunction. Int J Mol Sci 2022; 23:ijms23137092. [PMID: 35806096 PMCID: PMC9266532 DOI: 10.3390/ijms23137092] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
The high prevalence of metabolic syndrome in persons with schizophrenia has spurred investigational efforts to study the mechanism beneath its pathophysiology. Early psychosis dysfunction is present across multiple organ systems. On this account, schizophrenia may be a multisystem disorder in which one organ system is predominantly affected and where other organ systems are also concurrently involved. Growing evidence of the overlapping neurobiological profiles of metabolic risk factors and psychiatric symptoms, such as an association with cognitive dysfunction, altered autonomic nervous system regulation, desynchrony in the resting-state default mode network, and shared genetic liability, suggest that metabolic syndrome and schizophrenia are connected via common pathways that are central to schizophrenia pathogenesis, which may be underpinned by oxytocin system dysfunction. Oxytocin, a hormone that involves in the mechanisms of food intake and metabolic homeostasis, may partly explain this piece of the puzzle in the mechanism underlying this association. Given its prosocial and anorexigenic properties, oxytocin has been administered intranasally to investigate its therapeutic potential in schizophrenia and obesity. Although the pathophysiology and mechanisms of oxytocinergic dysfunction in metabolic syndrome and schizophrenia are both complex and it is still too early to draw a conclusion upon, oxytocinergic dysfunction may yield a new mechanistic insight into schizophrenia pathogenesis and treatment.
Collapse
Affiliation(s)
- Kah Kheng Goh
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Cynthia Yi-An Chen
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
| | - Tzu-Hua Wu
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Clinical Pharmacy, School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Chun-Hsin Chen
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Mong-Liang Lu
- Department of Psychiatry, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan; (K.K.G.); (C.Y.-A.C.); (C.-H.C.)
- Psychiatric Research Center, Wan-Fang Hospital, Taipei Medical University, Taipei 116, Taiwan;
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Correspondence:
| |
Collapse
|
19
|
Wang SC, Zhang F, Zhu H, Yang H, Liu Y, Wang P, Parpura V, Wang YF. Potential of Endogenous Oxytocin in Endocrine Treatment and Prevention of COVID-19. Front Endocrinol (Lausanne) 2022; 13:799521. [PMID: 35592777 PMCID: PMC9110836 DOI: 10.3389/fendo.2022.799521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/14/2022] [Indexed: 01/09/2023] Open
Abstract
Coronavirus disease 2019 or COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a significant threat to the health of human beings. While wearing mask, maintaining social distance and performing self-quarantine can reduce virus spreading passively, vaccination actively enhances immune defense against COVID-19. However, mutations of SARS-CoV-2 and presence of asymptomatic carriers frustrate the effort of completely conquering COVID-19. A strategy that can reduce the susceptibility and thus prevent COVID-19 while blocking viral invasion and pathogenesis independent of viral antigen stability is highly desirable. In the pathogenesis of COVID-19, endocrine disorders have been implicated. Correspondingly, many hormones have been identified to possess therapeutic potential of treating COVID-19, such as estrogen, melatonin, corticosteroids, thyroid hormone and oxytocin. Among them, oxytocin has the potential of both treatment and prevention of COVID-19. This is based on oxytocin promotion of immune-metabolic homeostasis, suppression of inflammation and pre-existing comorbidities, acceleration of damage repair, and reduction of individuals' susceptibility to pathogen infection. Oxytocin may specifically inactivate SARS-COV-2 spike protein and block viral entry into cells via angiotensin-converting enzyme 2 by suppressing serine protease and increasing interferon levels and number of T-lymphocytes. In addition, oxytocin can promote parasympathetic outflow and the secretion of body fluids that could dilute and even inactivate SARS-CoV-2 on the surface of cornea, oral cavity and gastrointestinal tract. What we need to do now is clinical trials. Such trials should fully balance the advantages and disadvantages of oxytocin application, consider the time- and dose-dependency of oxytocin effects, optimize the dosage form and administration approach, combine oxytocin with inhibitors of SARS-CoV-2 replication, apply specific passive immunization, and timely utilize efficient vaccines. Meanwhile, blocking COVID-19 transmission chain and developing other efficient anti-SARS-CoV-2 drugs are also important. In addition, relative to the complex issues with drug applications over a long term, oxytocin can be mobilized through many physiological stimuli, and thus used as a general prevention measure. In this review, we explore the potential of oxytocin for treatment and prevention of COVID-19 and perhaps other similar pathogens.
Collapse
Affiliation(s)
- Stephani C. Wang
- Division of Cardiology, Department of Medicine, University of California-Irvine, Irvine, CA, United States
| | - Fengmin Zhang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Hui Zhu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Haipeng Yang
- Neonatal Division of the Department of Pediatrics, Harbin Medical University The Fourth Affiliated Hospital, Harbin, China
| | - Yang Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Ping Wang
- Department of Genetics, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
20
|
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
|
21
|
Szczepanska-Sadowska E, Wsol A, Cudnoch-Jedrzejewska A, Żera T. Complementary Role of Oxytocin and Vasopressin in Cardiovascular Regulation. Int J Mol Sci 2021; 22:11465. [PMID: 34768894 PMCID: PMC8584236 DOI: 10.3390/ijms222111465] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/17/2022] Open
Abstract
The neurons secreting oxytocin (OXY) and vasopressin (AVP) are located mainly in the supraoptic, paraventricular, and suprachiasmatic nucleus of the brain. Oxytocinergic and vasopressinergic projections reach several regions of the brain and the spinal cord. Both peptides are released from axons, soma, and dendrites and modulate the excitability of other neuroregulatory pathways. The synthesis and action of OXY and AVP in the peripheral organs (eye, heart, gastrointestinal system) is being investigated. The secretion of OXY and AVP is influenced by changes in body fluid osmolality, blood volume, blood pressure, hypoxia, and stress. Vasopressin interacts with three subtypes of receptors: V1aR, V1bR, and V2R whereas oxytocin activates its own OXTR and V1aR receptors. AVP and OXY receptors are present in several regions of the brain (cortex, hypothalamus, pons, medulla, and cerebellum) and in the peripheral organs (heart, lungs, carotid bodies, kidneys, adrenal glands, pancreas, gastrointestinal tract, ovaries, uterus, thymus). Hypertension, myocardial infarction, and coexisting factors, such as pain and stress, have a significant impact on the secretion of oxytocin and vasopressin and on the expression of their receptors. The inappropriate regulation of oxytocin and vasopressin secretion during ischemia, hypoxia/hypercapnia, inflammation, pain, and stress may play a significant role in the pathogenesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Laboratory of Centre for Preclinical Research, Chair and Department of Experimental and Clinical Physiology, Medical University of Warsaw, 02-091 Warsaw, Poland; (A.W.); (A.C.-J.); (T.Ż.)
| | | | | | | |
Collapse
|
22
|
Epigenetic Alterations in Pediatric Sleep Apnea. Int J Mol Sci 2021; 22:ijms22179523. [PMID: 34502428 PMCID: PMC8430725 DOI: 10.3390/ijms22179523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 08/30/2021] [Indexed: 12/03/2022] Open
Abstract
Pediatric obstructive sleep apnea has significant negative effects on health and behavior in childhood including depression, failure to thrive, neurocognitive impairment, and behavioral issues. It is strongly associated with an increased risk for chronic adult disease such as obesity and diabetes, accelerated atherosclerosis, and endothelial dysfunction. Accumulating evidence suggests that adult-onset non-communicable diseases may originate from early life through a process by which an insult applied at a critical developmental window causes long-term effects on the structure or function of an organism. In recent years, there has been increased interest in the role of epigenetic mechanisms in the pathogenesis of adult disease susceptibility. Epigenetic mechanisms that influence adaptive variability include histone modifications, non-coding RNAs, and DNA methylation. This review will highlight what is currently known about the phenotypic associations of epigenetic modifications in pediatric obstructive sleep apnea and will emphasize the importance of epigenetic changes as both modulators of chronic disease and potential therapeutic targets.
Collapse
|
23
|
Wang X, Escobar JB, Mendelowitz D. Sex Differences in the Hypothalamic Oxytocin Pathway to Locus Coeruleus and Augmented Attention with Chemogenetic Activation of Hypothalamic Oxytocin Neurons. Int J Mol Sci 2021; 22:ijms22168510. [PMID: 34445224 PMCID: PMC8395169 DOI: 10.3390/ijms22168510] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/16/2022] Open
Abstract
The tightly localized noradrenergic neurons (NA) in the locus coeruleus (LC) are well recognized as essential for focused arousal and novelty-oriented responses, while many children with autism spectrum disorder (ASD) exhibit diminished attention, engagement and orienting to exogenous stimuli. This has led to the hypothesis that atypical LC activity may be involved in ASD. Oxytocin (OXT) neurons and receptors are known to play an important role in social behavior, pair bonding and cognitive processes and are under investigation as a potential treatment for ASD. However, little is known about the neurotransmission from hypothalamic paraventricular (PVN) OXT neurons to LC NA neurons. In this study, we test, in male and female rats, whether PVN OXT neurons excite LC neurons, whether oxytocin is released and involved in this neurotransmission, and whether activation of PVN OXT neurons alters novel object recognition. Using "oxytocin sniffer cells" (CHO cells that express the human oxytocin receptor and a Ca indicator) we show that there is release of OXT from hypothalamic PVN OXT fibers in the LC. Optogenetic excitation of PVN OXT fibers excites LC NA neurons by co-release of OXT and glutamate, and this neurotransmission is greater in males than females. In male, but not in female animals, chemogenetic activation of PVN OXT neurons increases attention to novel objects.
Collapse
|
24
|
Cavalcante GL, Brognara F, Oliveira LVDC, Lataro RM, Durand MDT, Oliveira AP, Nóbrega ACL, Salgado HC, Sabino JPJ. Benefits of pharmacological and electrical cholinergic stimulation in hypertension and heart failure. Acta Physiol (Oxf) 2021; 232:e13663. [PMID: 33884761 DOI: 10.1111/apha.13663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 03/12/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022]
Abstract
Systemic arterial hypertension and heart failure are cardiovascular diseases that affect millions of individuals worldwide. They are characterized by a change in the autonomic nervous system balance, highlighted by an increase in sympathetic activity associated with a decrease in parasympathetic activity. Most therapeutic approaches seek to treat these diseases by medications that attenuate sympathetic activity. However, there is a growing number of studies demonstrating that the improvement of parasympathetic function, by means of pharmacological or electrical stimulation, can be an effective tool for the treatment of these cardiovascular diseases. Therefore, this review aims to describe the advances reported by experimental and clinical studies that addressed the potential of cholinergic stimulation to prevent autonomic and cardiovascular imbalance in hypertension and heart failure. Overall, the published data reviewed demonstrate that the use of central or peripheral acetylcholinesterase inhibitors is efficient to improve the autonomic imbalance and hemodynamic changes observed in heart failure and hypertension. Of note, the baroreflex and the vagus nerve activation have been shown to be safe and effective approaches to be used as an alternative treatment for these cardiovascular diseases. In conclusion, pharmacological and electrical stimulation of the parasympathetic nervous system has the potential to be used as a therapeutic tool for the treatment of hypertension and heart failure, deserving to be more explored in the clinical setting.
Collapse
Affiliation(s)
- Gisele L. Cavalcante
- Graduate Program in Pharmaceutical Sciences Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
- Department of Pharmacology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Fernanda Brognara
- Department of Physiology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Lucas Vaz de C. Oliveira
- Graduate Program in Pharmaceutical Sciences Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
| | - Renata M. Lataro
- Department of Physiological Sciences Center of Biological Sciences Federal University of Santa Catarina Florianópolis SP Brazil
| | | | - Aldeidia P. Oliveira
- Graduate Program in Pharmacology Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
| | | | - Helio C. Salgado
- Department of Physiology Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - João Paulo J. Sabino
- Graduate Program in Pharmaceutical Sciences Department of Biophysics and Physiology Federal University of Piaui Teresina PI Brazil
| |
Collapse
|
25
|
Iovino M, Messana T, Tortora A, Giusti C, Lisco G, Giagulli VA, Guastamacchia E, De Pergola G, Triggiani V. Oxytocin Signaling Pathway: From Cell Biology to Clinical Implications. Endocr Metab Immune Disord Drug Targets 2021; 21:91-110. [PMID: 32433011 DOI: 10.2174/1871530320666200520093730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 04/04/2020] [Accepted: 04/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In addition to the well-known role played in lactation and parturition, Oxytocin (OT) and OT receptor (OTR) are involved in many other aspects such as the control of maternal and social behavior, the regulation of the growth of the neocortex, the maintenance of blood supply to the cortex, the stimulation of limbic olfactory area to mother-infant recognition bond, and the modulation of the autonomic nervous system via the vagal pathway. Moreover, OT and OTR show antiinflammatory, anti-oxidant, anti-pain, anti-diabetic, anti-dyslipidemic and anti-atherogenic effects. OBJECTIVE The aim of this narrative review is to summarize the main data coming from the literature dealing with the role of OT and OTR in physiology and pathologic conditions focusing on the most relevant aspects. METHODS Appropriate keywords and MeSH terms were identified and searched in Pubmed. Finally, references of original articles and reviews were examined. RESULTS We report the most significant and updated data on the role played by OT and OTR in physiology and different clinical contexts. CONCLUSION Emerging evidence indicates the involvement of OT system in several pathophysiological mechanisms influencing brain anatomy, cognition, language, sense of safety and trust and maternal behavior, with the possible use of exogenous administered OT in the treatment of specific neuropsychiatric conditions. Furthermore, it modulates pancreatic β-cell responsiveness and lipid metabolism leading to possible therapeutic use in diabetic and dyslipidemic patients and for limiting and even reversing atherosclerotic lesions.
Collapse
Affiliation(s)
- Michele Iovino
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Tullio Messana
- Infantile Neuropsychiatry, IRCCS - Institute of Neurological Sciences, Bologna, Italy
| | - Anna Tortora
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Consuelo Giusti
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giuseppe Lisco
- Hospital Unit of Endocrinology, Perrino Hospital, Brindisi, Italy
| | - Vito Angelo Giagulli
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Edoardo Guastamacchia
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Giovanni De Pergola
- Clinical Nutrition Unit, Medical Oncology, Department of Internal Medicine and Clinical Oncology, University of Bari, School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| | - Vincenzo Triggiani
- Interdisciplinary Department of Medicine-Section of Internal Medicine, Geriatrics, Endocrinology and Rare Diseases. University of Bari "Aldo Moro", School of Medicine, Policlinico, Piazza Giulio Cesare 11, 70124, Bari, Italy
| |
Collapse
|
26
|
Wang SC, Wang YF. Cardiovascular protective properties of oxytocin against COVID-19. Life Sci 2021; 270:119130. [PMID: 33513400 PMCID: PMC7837104 DOI: 10.1016/j.lfs.2021.119130] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2 infection or COVID-19 has become a worldwide pandemic; however, effective treatment for COVID-19 remains to be established. Along with acute respiratory distress syndrome (ARDS), new and old cardiovascular injuries are important causes of significant morbidity and mortality in COVID-19. Exploring new approaches managing cardiovascular complications is essential in controlling the disease progression and preventing long-term complications. Oxytocin (OXT), an immune-regulating neuropeptide, has recently emerged as a strong candidate for treatment and prevention of COVID-19 pandemic. OXT carries special functions in immunologic defense, homeostasis and surveillance. It suppresses neutrophil infiltration and inflammatory cytokine release, activates T-lymphocytes, and antagonizes negative effects of angiotensin II and other key pathological events of COVID-19. Additionally, OXT can promote γ-interferon expression to inhibit cathepsin L and increases superoxide dismutase expression to reduce heparin and heparan sulphate fragmentation. Through these mechanisms, OXT can block viral invasion, suppress cytokine storm, reverse lymphocytopenia, and prevent progression to ARDS and multiple organ failures. Importantly, besides prevention of metabolic disorders associated with atherosclerosis and diabetes mellitus, OXT can protect the heart and vasculature through suppressing hypertension and brain-heart syndrome, and promoting regeneration of injured cardiomyocytes. Unlike other therapeutic agents, exogenous OXT can be used safely without the side-effects seen in remdesivir and corticosteroid. Importantly, OXT can be mobilized endogenously to prevent pathogenesis of COVID-19. This article summarizes our current understandings of cardiovascular pathogenesis caused by COVID-19, explores the protective potentials of OXT against COVID-19-associated cardiovascular diseases, and discusses challenges in applying OXT in treatment and prevention of COVID-19. Chemical compounds Angiotensin-converting enzyme 2 (ACE2); atrial natriuretic peptide (ANP); cathepsin L; heparan sulphate proteoglycans (HSPGs); interferon; interleukin; oxytocin; superoxide dismutase; transmembrane serine protease isoform 2 (TMPRSS2).
Collapse
Affiliation(s)
- Stephani C Wang
- Division of Cardiology, Department of Medicine, University of California-Irvine, Irvine, CA, USA.
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China.
| |
Collapse
|
27
|
Moreno A, Kowalik G, Mendelowitz D, Kay MW. Optogenetic Control of Cardiac Autonomic Neurons in Transgenic Mice. Methods Mol Biol 2021; 2191:309-321. [PMID: 32865752 DOI: 10.1007/978-1-0716-0830-2_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Optogenetic technology has enabled unparalleled insights into cellular and organ physiology by providing exquisite temporal and spatial control of biological pathways. Here, an optogenetic approach is presented for selective activation of the intrinsic cardiac nervous system in excised perfused mouse hearts. The breeding of transgenic mice that have selective expression of channelrhodopsin in either catecholaminergic or cholinergic neurons is described. An approach for perfusing hearts excised from those animals, recording the ECG to measure heart rate changes, and an illumination technique using a custom micro-LED light source to activate channelrhodopsin is explained. We have used these methods in ongoing studies of the kinetics of autonomic control of cardiac electrophysiology and contractility, demonstrating the proven utility of optogenetic technology to enable unparalleled spatiotemporal anatomic-functional probing of the intrinsic cardiac nervous system.
Collapse
Affiliation(s)
- Angel Moreno
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Grant Kowalik
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
| |
Collapse
|
28
|
Brain oxytocin: how puzzle stones from animal studies translate into psychiatry. Mol Psychiatry 2021; 26:265-279. [PMID: 32514104 PMCID: PMC7278240 DOI: 10.1038/s41380-020-0802-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/14/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
The neuropeptide oxytocin has attracted great attention of the general public, basic neuroscience researchers, psychologists, and psychiatrists due to its profound pro-social, anxiolytic, and "anti-stress" behavioral and physiological effects, and its potential application for treatment of mental diseases associated with altered socio-emotional competence. During the last decade, substantial progress has been achieved in understanding the complex neurobiology of the oxytocin system, including oxytocinergic pathways, local release patterns, and oxytocin receptor distribution in the brain, as well as intraneuronal oxytocin receptor signaling. However, the picture of oxytocin actions remains far from being complete, and the central question remains: "How does a single neuropeptide exert such pleotropic actions?" Although this phenomenon, typical for many of about 100 identified neuropeptides, may emerge from the anatomical divergence of oxytocin neurons, their multiple central projections, distinct oxytocin-sensitive cell types in different brain regions, and multiple intraneuronal signaling pathways determining the specific cellular response, further basic studies are required. In conjunction, numerous reports on positive effects of intranasal application of oxytocin on human brain networks controlling socio-emotional behavior in health and disease require harmonic tandems of basic researchers and clinicians. During the COVID-19 crisis in 2020, oxytocin research seems central as question of social isolation-induced inactivation of the oxytocin system, and buffering effects of either activation of the endogenous system or intranasal application of synthetic oxytocin need to be thoroughly investigated.
Collapse
|
29
|
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
|
30
|
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: 4] [Impact Index Per Article: 0.8] [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
|
31
|
Zasadny FM, Dyavanapalli J, Dowling NM, Mendelowitz D, Kay MW. Cholinergic stimulation improves electrophysiological rate adaptation during pressure overload-induced heart failure in rats. Am J Physiol Heart Circ Physiol 2020; 319:H1358-H1368. [PMID: 33006920 PMCID: PMC7792708 DOI: 10.1152/ajpheart.00293.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Left ventricular (LV) electrical maladaptation to increased heart rate in failing myocardium contributes to morbidity and mortality. Recently, cardiac cholinergic neuron activation reduced loss of contractile function resulting from chronic trans-aortic constriction (TAC) in rats. We hypothesized that chronic activation of cardiac cholinergic neurons would also reduce TAC-induced derangement of cardiac electrical activity. METHODS We investigated electrophysiological rate adaptation in TAC rat hearts with and without daily chemogenetic activation of hypothalamic oxytocin neurons for downstream cardiac cholinergic neuron stimulation. Sprague Dawley rat hearts were excised, perfused, and optically mapped under dynamic pacing after 16 weeks of TAC with or without 12 weeks of daily chemogenetic treatment. Action potential duration (APD60) and conduction velocity (CV) maps were analyzed for regional rate adaptation to dynamic pacing. RESULTS At lower pacing rates, untreated TAC induced elevated LV epicardial APD60. Fitted APD60 steady state (APDss) was reduced in treated TAC hearts. At higher pacing rates, treatment heterogeneously reduced APD60 compared to untreated TAC hearts. Variance of conduction loss was reduced in treated hearts compared to untreated hearts during fast pacing. However, CV was markedly reduced in both treated and untreated TAC hearts throughout dynamic pacing. At 150msec pacing cycle length, APD60 v. diastolic interval (DI) dispersion was reduced in treated hearts compared to untreated hearts. CONCLUSIONS Chronic activation of cardiac cholinergic neurons improved electrophysiological adaptation to increases in pacing rate during development of TAC-induced heart failure. This provides insight into the electrophysiological benefits of cholinergic stimulation as a treatment for heart failure patients.
Collapse
Affiliation(s)
| | | | | | - David Mendelowitz
- Pharmacology and Physiology, George Washington University, United States
| | - Matthew W Kay
- Biomedical Engineering, George Washington University, United States
| |
Collapse
|
32
|
McKay EC, Counts SE. Oxytocin Receptor Signaling in Vascular Function and Stroke. Front Neurosci 2020; 14:574499. [PMID: 33071746 PMCID: PMC7544744 DOI: 10.3389/fnins.2020.574499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/31/2020] [Indexed: 12/13/2022] Open
Abstract
The oxytocin receptor (OXTR) is a G protein-coupled receptor with a diverse repertoire of intracellular signaling pathways, which are activated in response to binding oxytocin (OXT) and a similar nonapeptide, vasopressin. This review summarizes the cell and molecular biology of the OXTR and its downstream signaling cascades, particularly focusing on the vasoactive functions of OXTR signaling in humans and animal models, as well as the clinical applications of OXTR targeting cerebrovascular accidents.
Collapse
Affiliation(s)
- Erin C McKay
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Scott E Counts
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Department of Family Medicine, Michigan State University, Grand Rapids, MI, United States.,Hauenstein Neurosciences Center, Mercy Health Saint Mary's Hospital, Grand Rapids, MI, United States.,Michigan Alzheimer's Disease Research Center, Ann Arbor, MI, United States
| |
Collapse
|
33
|
Augoulea A, Armeni E, Paschou SA, Georgiopoulos G, Stamatelopoulos K, Lambrinoudaki I. Breastfeeding is associated with lower subclinical atherosclerosis in postmenopausal women. Gynecol Endocrinol 2020; 36:796-799. [PMID: 32584151 DOI: 10.1080/09513590.2020.1782374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Objective: To evaluate the association between a personal history of lactation and indices of subclinical atherosclerosis in postmenopausal women.Methods: We evaluated the association between a history of breastfeeding and indices of subclinical atherosclerosis (pulse wave velocity, PWV; intima-media thickness [IMT]; atherosclerotic plaque presence) in 197 parous postmenopausal women with history of breastfeeding.Results: Women who reported breastfeeding ≥6 months when compared with women who reported breastfeeding for 1-5 months exhibited significantly lower values of common carotid artery IMT (Model R2=15.7%, b-coefficient = -0.170, 95% CI: -0.208-0.001, p-value = .019) and lower odds of subclinical atherosclerosis (Model X2=28.127, OR = 0.491, 95% CI 0.318-0.999, p-value = .049), adjusting for traditional cardiovascular risk factors.Conclusions: Postmenopausal women with a history of breastfeeding for at least 6 months have a lower prevalence of subclinical atherosclerosis, independently of traditional cardiovascular risk factors. A longer duration of breastfeeding may have a beneficial effect on subclinical atherosclerosis later in life.
Collapse
Affiliation(s)
- Areti Augoulea
- 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, University of Athens, Athens, Greece
| | - Eleni Armeni
- 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, University of Athens, Athens, Greece
| | - Stavroula A Paschou
- 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, University of Athens, Athens, Greece
| | - Georgios Georgiopoulos
- Vascular Laboratory, Department of Clinical Therapeutics, Alexandra Hospital, University of Athens, Athens, Greece
| | - Kimon Stamatelopoulos
- Vascular Laboratory, Department of Clinical Therapeutics, Alexandra Hospital, University of Athens, Athens, Greece
| | - Irene Lambrinoudaki
- 2nd Department of Obstetrics and Gynecology, Aretaieio Hospital, University of Athens, Athens, Greece
| |
Collapse
|
34
|
Jain V, Kimbro S, Kowalik G, Milojevic I, Maritza Dowling N, Hunley AL, Hauser K, Andrade DC, Del Rio R, Kay MW, Mendelowitz D. Intranasal oxytocin increases respiratory rate and reduces obstructive event duration and oxygen desaturation in obstructive sleep apnea patients: a randomized double blinded placebo controlled study. Sleep Med 2020; 74:242-247. [PMID: 32862007 DOI: 10.1016/j.sleep.2020.05.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Activation of the oxytocin network has shown benefits in animal models of Obstructive Sleep Apnea (OSA) as well as other cardiorespiratory diseases. We sought to determine if nocturnal intranasal oxytocin administration could have beneficial effects in reducing the duration and/or frequency of obstructive events in obstructive sleep apnea subjects. METHODS Two sequential standard "in-lab" polysomnogram (PSG) sleep studies were performed in patients diagnosed with OSA that were randomly assigned to initially receive either placebo or oxytocin (40 i.u.) administered intranasally in this double blinded randomized placebo controlled study. Changes in cardiorespiratory events during sleep, including apnea and hypopnea durations and frequency, risk of event-associated bradycardias, arterial oxygen desaturation and respiratory rate were assessed in 2 h epochs following sleep onset. Oxytocin significantly decreased the duration of obstructive events, as well as the oxygen desaturations and incidence of bradycardia that were associated with these events. Notably, oxytocin increased respiratory rate during non-obstructive periods. There were no significant changes in sleep architecture and no adverse effects were reported. CONCLUSIONS Oxytocin administration can benefit OSA subjects by reducing the duration and adverse consequences of obstructive events. Oxytocin could also be beneficial in situations involving respiratory depression as oxytocin increased respiratory rate. Additional studies are needed to further understand the mechanisms by which oxytocin promotes these changes in cardiorespiratory function. The long-term efficacy and optimal dose of intranasal oxytocin treatment should also be determined in OSA subjects. ClinicalTrials.gov NCT03148899.
Collapse
Affiliation(s)
- Vivek Jain
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Shawn Kimbro
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Grant Kowalik
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Ivana Milojevic
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - N Maritza Dowling
- Department of Acute & Chronic Care, School of Nursing, Department of Epidemiology & Biostatistics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Anne Lloyd Hunley
- Department of Medicine, The George Washington University, Washington, DC, USA
| | - Kelsey Hauser
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA
| | - David C Andrade
- Centro de Fisiología Del Ejercicio, Universidad Mayor, Santiago, Chile; Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile; Center for Aging and Regeneration (CARE-UC), Pontificia Universidad Católica de Chile, Santiago, Chile; Centro de Excelencia de Biomedicina en Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Matthew W Kay
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University, Washington, DC, USA.
| |
Collapse
|
35
|
Dyavanapalli J, Rodriguez J, Rocha Dos Santos C, Escobar JB, Dwyer MK, Schloen J, Lee KM, Wolaver W, Wang X, Dergacheva O, Michelini LC, Schunke KJ, Spurney CF, Kay MW, Mendelowitz D. Activation of Oxytocin Neurons Improves Cardiac Function in a Pressure-Overload Model of Heart Failure. ACTA ACUST UNITED AC 2020; 5:484-497. [PMID: 32478209 PMCID: PMC7251188 DOI: 10.1016/j.jacbts.2020.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 01/26/2023]
Abstract
Hypothalamic OXT neurons were chronically activated using a chemogenetic approach in an animal model of HF. Synaptic release of OXT onto parasympathetic autonomic targets was reduced in animals with HF but restored with daily treatment consisting of activation of OXT neurons. Long-term daily OXT neuron activation increased parasympathetic activity to the heart and reduced mortality, cardiac inflammation, and fibrosis and improved critical longitudinal in vivo indices of cardiac function. The benefits in cardiac function and autonomic balance in HF closely tracked the study-designed differences in initiation of OXT neuron activation in different groups.
This work shows long-term restoration of the hypothalamic oxytocin (OXT) network preserves OXT release, reduces mortality, cardiac inflammation, fibrosis, and improves autonomic tone and cardiac function in a model of heart failure. Intranasal administration of OXT in patients mimics the short-term changes seen in animals by increasing parasympathetic—and decreasing sympathetic—cardiac activity. This work provides the essential translational foundation to determine if approaches that mimic paraventricular nucleus (PVN) OXT neuron activation, such as safe, noninvasive, and well-tolerated intranasal administration of OXT, can be beneficial in patients with heart failure.
Collapse
Key Words
- ANOVA, analysis of variance
- CHO, Chinese hamster ovary
- CNO, clozapine-N-oxide
- CVN, cardiac vagal neuron
- ChR2, channelrhodopsin
- DMNX, dorsal motor nucleus of the vagus
- DREADD, designer receptors exclusively activated by designer drug
- HF, heart failure
- IL, interleukin
- LV, left ventricle
- LVDP, left ventricle- developed pressure
- OXT, oxytocin
- PVN, paraventricular nucleus of the hypothalamus
- SD, standard deviation
- TAC, transascending aortic constriction
- heart failure
- oxytocin
- parasympathetic
Collapse
Affiliation(s)
- Jhansi Dyavanapalli
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Jeannette Rodriguez
- Department of Biomedical Engineering, George Washington University, Washington, DC
| | | | - Joan B Escobar
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Mary Kate Dwyer
- Department of Biomedical Engineering, George Washington University, Washington, DC
| | - John Schloen
- Department of Biomedical Engineering, George Washington University, Washington, DC
| | - Kyung-Min Lee
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Whitney Wolaver
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Xin Wang
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Olga Dergacheva
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| | - Lisete C Michelini
- Department of Physiology, Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo/SP, Brazil
| | - Kathryn J Schunke
- Department of Biomedical Engineering, George Washington University, Washington, DC
| | - Christopher F Spurney
- Children's National Heart Institute, Center for Genetic Medicine Research, Children's National Health System, Washington, DC
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Washington, DC
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC
| |
Collapse
|
36
|
Japundžić-Žigon N, Lozić M, Šarenac O, Murphy D. Vasopressin & Oxytocin in Control of the Cardiovascular System: An Updated Review. Curr Neuropharmacol 2020; 18:14-33. [PMID: 31544693 PMCID: PMC7327933 DOI: 10.2174/1570159x17666190717150501] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/03/2019] [Accepted: 07/06/2019] [Indexed: 01/19/2023] Open
Abstract
Since the discovery of vasopressin (VP) and oxytocin (OT) in 1953, considerable knowledge has been gathered about their roles in cardiovascular homeostasis. Unraveling VP vasoconstrictor properties and V1a receptors in blood vessels generated powerful hemostatic drugs and drugs effective in the treatment of certain forms of circulatory collapse (shock). Recognition of the key role of VP in water balance via renal V2 receptors gave birth to aquaretic drugs found to be useful in advanced stages of congestive heart failure. There are still unexplored actions of VP and OT on the cardiovascular system, both at the periphery and in the brain that may open new venues in treatment of cardiovascular diseases. After a brief overview on VP, OT and their peripheral action on the cardiovascular system, this review focuses on newly discovered hypothalamic mechanisms involved in neurogenic control of the circulation in stress and disease.
Collapse
Affiliation(s)
| | - Maja Lozić
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Olivera Šarenac
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - David Murphy
- School of Clinical Sciences, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
37
|
Buemann B, Uvnäs-Moberg K. Oxytocin may have a therapeutical potential against cardiovascular disease. Possible pharmaceutical and behavioral approaches. Med Hypotheses 2020; 138:109597. [PMID: 32032912 DOI: 10.1016/j.mehy.2020.109597] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/11/2020] [Accepted: 01/22/2020] [Indexed: 12/22/2022]
Abstract
Based on the ancient role of oxytocin and its homologues as amplifiers of reproduction we argue for an evolutionary coupling of oxytocin to signaling pathway which support restorative mechanisms of cells and tissue. In particular, the survival and function of different categories of stem cells and primordial cells are enhanced by mitogen-activated protein kinase (MAPK) pathways. Furthermore, oxytocin stimulates the AMP-activated protein kinase pathway (AMPK) in numerous of cell types which promotes the maintenance of different cell structures. This involves autophagic processes and, in particular, may support the renewal of mitochondria. Mitochondrial fitness may protect against oxidative and inflammatory stress - a well-documented effect of oxytocin. The combined specific trophic and protective effects oxytocin may delay several degenerative phenomena including sarcopenia, type-2 diabetes and atherosclerosis. These effects may be exerted both on a central level supporting the function and integrity of the hypothalamus and peripherally acting directly on blood vessels, pancreas, heart, skeletal muscles and adipose tissue etc. Furthermore, in the capacity of being both a hormone and neuromodulator, oxytocin interacts with numerous of regulatory mechanisms particularly the autonomic nervous system and HPA-axis which may reduce blood pressure and affect the immune function. The potential of the oxytocin system as a behavioral and molecular target for the prevention and treatment of cardiovascular disease is discussed. Focus is put on the affiliative and sexual significance and the different options and limitations associated with a pharmaceutical approach. MeSH: Aging, Atherosclerosis, Heart, Hypothalamus, Inflammation, Love, Orgasm, Oxytocin.
Collapse
Affiliation(s)
| | - Kerstin Uvnäs-Moberg
- Department of Animal Environment and Health, Swedish University of Agricultural Sciences, Skara, Sweden
| |
Collapse
|
38
|
Wsol A, Wojno O, Puchalska L, Wrzesien R, Szczepanska-Sadowska E, Cudnoch-Jedrzejewska A. Impaired hypotensive effects of centrally acting oxytocin in SHR and WKY rats exposed to chronic mild stress. Am J Physiol Regul Integr Comp Physiol 2020; 318:R160-R172. [DOI: 10.1152/ajpregu.00050.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The present study was designed to determine the role of centrally acting oxytocin (OT) in the regulation of blood pressure during chronic mild stress (CMS) in spontaneously hypertensive (SHR; n = 36) and normotensive Wistar-Kyoto (WKY; n = 38) rats. The rats were implanted with osmotic minipumps for intracerebroventricular infusions of 0.9% NaCl, OT, and oxytocin receptor antagonist (OTANT) and divided into two groups: SHR and WKY 1) exposed to 4-wk CMS and 2) not exposed to stress (controls). After 4 wk, hemodynamic parameters were recorded at rest and after an application of acute stressor [air-jet stress (AJS)]. Resting mean arterial blood pressure (MAP) was significantly lower in CMS-exposed SHR and WKY infused with OT than in the corresponding groups receiving saline. Exposure to CMS exaggerated the AJS-dependent pressor response in WKY receiving saline but not in the corresponding group of SHR. OT infusion reduced the AJS-dependent pressor response in both CMS-exposed and not exposed SHR and in CMS-exposed WKY. Intracerebroventricular infusion of OTANT potentiated the AJS-dependent pressor response in both stressed and not stressed WKY rats but not in SHR. The results show that centrally delivered OT decreases resting MAP during CMS in both SHR and WKY rats and that in SHR it reduces pressor responses to AJS under control and CMS conditions, whereas in WKY this effect is significant only after CMS exposure. The study indicates that endogenous centrally acting OT may play an essential role in buffering pressor responses to AJS in CMS-exposed and not exposed WKY rats and that this function is significantly impaired in SHR.
Collapse
Affiliation(s)
- A. Wsol
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - O. Wojno
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - L. Puchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - R. Wrzesien
- Department of Animal Breeding, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - E. Szczepanska-Sadowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - A. Cudnoch-Jedrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
39
|
Ruyle BC, Martinez D, Heesch CM, Kline DD, Hasser EM. The PVN enhances cardiorespiratory responses to acute hypoxia via input to the nTS. Am J Physiol Regul Integr Comp Physiol 2019; 317:R818-R833. [PMID: 31509428 PMCID: PMC6962628 DOI: 10.1152/ajpregu.00135.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/26/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Chemoreflex neurocircuitry includes the paraventricular nucleus (PVN), but the role of PVN efferent projections to specific cardiorespiratory nuclei is unclear. We hypothesized that the PVN contributes to cardiorespiratory responses to hypoxia via projections to the nucleus tractus solitarii (nTS). Rats received bilateral PVN microinjections of adeno-associated virus expressing inhibitory designer receptor exclusively activated by designer drug (GiDREADD) or green fluorescent protein (GFP) control. Efficacy of GiDREADD inhibition by the designer receptor exclusively activated by designer drug (DREADD) agonist Compound 21 (C21) was verified in PVN slices; C21 reduced evoked action potential discharge by reducing excitability to injected current in GiDREADD-expressing PVN neurons. We evaluated hypoxic ventilatory responses (plethysmography) and PVN and nTS neuronal activation (cFos immunoreactivity) to 2 h hypoxia (10% O2) in conscious GFP and GiDREADD rats after intraperitoneal C21 injection. Generalized PVN inhibition via systemic C21 blunted hypoxic ventilatory responses and reduced PVN and also nTS neuronal activation during hypoxia. To determine if the PVN-nTS pathway contributes to these effects, we evaluated cardiorespiratory responses to hypoxia during selective PVN terminal inhibition in the nTS. Anesthetized GFP and GiDREADD rats exposed to brief hypoxia (10% O2, 45 s) exhibited depressor and tachycardic responses and increased sympathetic and phrenic nerve activity. C21 was then microinjected into the nTS, followed after 60 min by another hypoxic episode. In GiDREADD but not GFP rats, PVN terminal inhibition by nTS C21 strongly attenuated the phrenic amplitude response to hypoxia. Interestingly, C21 augmented tachycardic and sympathetic responses without altering the coupling of splanchnic sympathetic nerve activity to phrenic nerve activity during hypoxia. Data demonstrate that the PVN, including projections to the nTS, is critical in shaping sympathetic and respiratory responses to hypoxia.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Diana Martinez
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Cheryl M Heesch
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - David D Kline
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Eileen M Hasser
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
40
|
Mukerjee S, Gao H, Xu J, Sato R, Zsombok A, Lazartigues E. ACE2 and ADAM17 Interaction Regulates the Activity of Presympathetic Neurons. Hypertension 2019; 74:1181-1191. [PMID: 31564162 DOI: 10.1161/hypertensionaha.119.13133] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Brain renin angiotensin system within the paraventricular nucleus plays a critical role in balancing excitatory and inhibitory inputs to modulate sympathetic output and blood pressure regulation. We previously identified ACE2 and ADAM17 as a compensatory enzyme and a sheddase, respectively, involved in brain renin angiotensin system regulation. Here, we investigated the opposing contribution of ACE2 and ADAM17 to hypothalamic presympathetic activity and ultimately neurogenic hypertension. New mouse models were generated where ACE2 and ADAM17 were selectively knocked down from all neurons (AC-N) or Sim1 neurons (SAT), respectively. Neuronal ACE2 deletion revealed a reduction of inhibitory inputs to AC-N presympathetic neurons relevant to blood pressure regulation. Primary neuron cultures confirmed ACE2 expression on GABAergic neurons synapsing onto excitatory neurons within the hypothalamus but not on glutamatergic neurons. ADAM17 expression was shown to colocalize with angiotensin-II type 1 receptors on Sim1 neurons, and the pressor relevance of this neuronal population was demonstrated by photoactivation. Selective knockdown of ADAM17 was associated with a reduction of FosB gene expression, increased vagal tone, and prevented the acute pressor response to centrally administered angiotensin-II. Chronically, SAT mice exhibited a blunted blood pressure elevation and preserved ACE2 activity during development of salt-sensitive hypertension. Bicuculline injection in those models confirmed the supporting role of ACE2 on GABAergic tone to the paraventricular nucleus. Together, our study demonstrates the contrasting impact of ACE2 and ADAM17 on neuronal excitability of presympathetic neurons within the paraventricular nucleus and the consequences of this mutual regulation in the context of neurogenic hypertension.
Collapse
Affiliation(s)
- Snigdha Mukerjee
- From the Department of Pharmacology and Experimental Therapeutics (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,Cardiovascular Center of Excellence (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,Neuroscience Center of Excellence (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans
| | - Hong Gao
- Department of Physiology, School of Medicine (H.G., R.S., A.Z.), Tulane University, New Orleans.,Brain Institute (H.G., A.Z.), Tulane University, New Orleans
| | - Jiaxi Xu
- From the Department of Pharmacology and Experimental Therapeutics (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,Cardiovascular Center of Excellence (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,Neuroscience Center of Excellence (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,SouthEast Louisiana Veterans Health Care System, New Orleans (J.X., E.L.)
| | - Ryosuke Sato
- Department of Physiology, School of Medicine (H.G., R.S., A.Z.), Tulane University, New Orleans
| | - Andrea Zsombok
- Department of Physiology, School of Medicine (H.G., R.S., A.Z.), Tulane University, New Orleans.,Brain Institute (H.G., A.Z.), Tulane University, New Orleans
| | - Eric Lazartigues
- From the Department of Pharmacology and Experimental Therapeutics (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,Cardiovascular Center of Excellence (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,Neuroscience Center of Excellence (S.M., J.X., E.L.), Louisiana State University Health Sciences Center, New Orleans.,SouthEast Louisiana Veterans Health Care System, New Orleans (J.X., E.L.)
| |
Collapse
|
41
|
Reiss AB, Glass DS, Lam E, Glass AD, De Leon J, Kasselman LJ. Oxytocin: Potential to mitigate cardiovascular risk. Peptides 2019; 117:170089. [PMID: 31112739 DOI: 10.1016/j.peptides.2019.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/17/2019] [Accepted: 05/10/2019] [Indexed: 02/08/2023]
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide, despite multiple treatment options. In addition to elevated lipid levels, oxidative stress and inflammation are key factors driving atherogenesis and CVD. New strategies are required to mitigate risk and most urgently for statin-intolerant patients. The neuropeptide hormone oxytocin, synthesized in the brain hypothalamus, is worthy of consideration as a CVD ancillary treatment because it moderates factors directly linked to atherosclerotic CVD such as inflammation, weight gain, food intake and insulin resistance. Though initially studied for its contribution to parturition and lactation, oxytocin participates in social attachment and bonding, associative learning, memory and stress responses. Oxytocin has shown promise in animal models of atherosclerosis and in some human studies as well. A number of properties of oxytocin make it a candidate CVD treatment. Oxytocin not only lowers fat mass and cytokine levels, but also improves glucose tolerance, lowers blood pressure and relieves anxiety. Further, it has an important role in communication in the gut-brain axis that makes it a promising treatment for obesity and type 2 diabetes. Oxytocin acts through its receptor which is a class I G-protein-coupled receptor present in cells of the vascular system including the heart and arteries. While oxytocin is not used for heart disease at present, residual CVD risk remains in a substantial portion of patients despite multidrug regimens, leaving open the possibility of using the endogenous nonapeptide as an adjunct therapy. This review discusses the possible role for oxytocin in human CVD prevention and treatment.
Collapse
Affiliation(s)
- Allison B Reiss
- Department of Medicine and Research Institute, NYU Winthrop Hospital, Mineola NY 11501, USA.
| | - Daniel S Glass
- Department of Medicine and Research Institute, NYU Winthrop Hospital, Mineola NY 11501, USA
| | - Eric Lam
- Department of Medicine and Research Institute, NYU Winthrop Hospital, Mineola NY 11501, USA
| | - Amy D Glass
- Department of Medicine and Research Institute, NYU Winthrop Hospital, Mineola NY 11501, USA
| | - Joshua De Leon
- Department of Medicine and Research Institute, NYU Winthrop Hospital, Mineola NY 11501, USA
| | - Lora J Kasselman
- Department of Medicine and Research Institute, NYU Winthrop Hospital, Mineola NY 11501, USA
| |
Collapse
|
42
|
Farmer GE, Amune A, Bachelor ME, Duong P, Yuan JP, Cunningham JT. Sniffer cells for the detection of neural Angiotensin II in vitro. Sci Rep 2019; 9:8820. [PMID: 31217439 PMCID: PMC6584535 DOI: 10.1038/s41598-019-45262-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 06/04/2019] [Indexed: 12/29/2022] Open
Abstract
Neuropeptide release in the brain has traditionally been difficult to observe. Existing methods lack temporal and spatial resolution that is consistent with the function and size of neurons. We use cultured "sniffer cells" to improve the temporal and spatial resolution of observing neuropeptide release. Sniffer cells were created by stably transfecting Chinese Hamster Ovary (CHO) cells with plasmids encoding the rat angiotensin type 1a receptor and a genetically encoded Ca2+ sensor. Isolated, cultured sniffer cells showed dose-dependent increases in fluorescence in response to exogenously applied angiotensin II and III, but not other common neurotransmitters. Sniffer cells placed on the median preoptic nucleus (a presumptive site of angiotensin release) displayed spontaneous activity and evoked responses to either electrical or optogenetic stimulation of the subfornical organ. Stable sniffer cell lines could be a viable method for detecting neuropeptide release in vitro, while still being able to distinguish differences in neuropeptide concentration.
Collapse
Affiliation(s)
- George E Farmer
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, 76107, United States
| | - Anna Amune
- Texas A&M University, College Station, TX, United States
| | - Martha E Bachelor
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, 76107, United States
| | - Phong Duong
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, 76107, United States
| | - Joseph P Yuan
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, 76107, United States
| | - J Thomas Cunningham
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, 76107, United States.
| |
Collapse
|
43
|
Wang P, Wang SC, Yang H, Lv C, Jia S, Liu X, Wang X, Meng D, Qin D, Zhu H, Wang YF. Therapeutic Potential of Oxytocin in Atherosclerotic Cardiovascular Disease: Mechanisms and Signaling Pathways. Front Neurosci 2019; 13:454. [PMID: 31178679 PMCID: PMC6537480 DOI: 10.3389/fnins.2019.00454] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Coronary artery disease (CAD) is a major cardiovascular disease responsible for high morbidity and mortality worldwide. The major pathophysiological basis of CAD is atherosclerosis in association with varieties of immunometabolic disorders that can suppress oxytocin (OT) receptor (OTR) signaling in the cardiovascular system (CVS). By contrast, OT not only maintains cardiovascular integrity but also has the potential to suppress and even reverse atherosclerotic alterations and CAD. These protective effects of OT are associated with its protection of the heart and blood vessels from immunometabolic injuries and the resultant inflammation and apoptosis through both peripheral and central approaches. As a result, OT can decelerate the progression of atherosclerosis and facilitate the recovery of CVS from these injuries. At the cellular level, the protective effect of OT on CVS involves a broad array of OTR signaling events. These signals mainly belong to the reperfusion injury salvage kinase pathway that is composed of phosphatidylinositol 3-kinase-Akt-endothelial nitric oxide synthase cascades and extracellular signal-regulated protein kinase 1/2. Additionally, AMP-activated protein kinase, Ca2+/calmodulin-dependent protein kinase signaling and many others are also implicated in OTR signaling in the CVS protection. These signaling events interact coordinately at many levels to suppress the production of inflammatory cytokines and the activation of apoptotic pathways. A particular target of these signaling events is endoplasmic reticulum (ER) stress and mitochondrial oxidative stress that interact through mitochondria-associated ER membrane. In contrast to these protective effects and machineries, rare but serious cardiovascular disturbances were also reported in labor induction and animal studies including hypotension, reflexive tachycardia, coronary spasm or thrombosis and allergy. Here, we review our current understanding of the protective effect of OT against varieties of atherosclerotic etiologies as well as the approaches and underlying mechanisms of these effects. Moreover, potential cardiovascular disturbances following OT application are also discussed to avoid unwanted effects in clinical trials of OT usages.
Collapse
Affiliation(s)
- Ping Wang
- Department of Genetics, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Stephani C Wang
- Department of Medicine, Albany Medical Center, Albany, NY, United States
| | - Haipeng Yang
- Department of Pediatrics, The Forth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Chunmei Lv
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Shuwei Jia
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoyu Liu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Xiaoran Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Dexin Meng
- Department of Physiology, Jiamusi University, Jiamusi, China
| | - Danian Qin
- Department of Physiology, Shantou University of Medical College, Shantou, China
| | - Hui Zhu
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Yu-Feng Wang
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, China
| |
Collapse
|
44
|
Zachou G, Armeni E, Lambrinoudaki I. Lactation and maternal cardiovascular disease risk in later life. Maturitas 2019; 122:73-79. [PMID: 30797534 DOI: 10.1016/j.maturitas.2019.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/09/2019] [Accepted: 01/22/2019] [Indexed: 01/20/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. The identification of protective factors against cardiovascular disease is important with regard to public health policies. Lactation has multiple beneficial effects for both mother and child. This review summarizes the evidence on the association between lactation and maternal cardiovascular risk in later life. Lactation may help to reverse the metabolic and cardiovascular changes that take place during pregnancy. Overall, lactation seems to exert a protective effect against the development of hypertension, metabolic syndrome, and diabetes, whilst data on postpartum weight and lipidemic profile are less conclusive. Both subclinical and clinical cardiovascular disease are negatively associated with a history of lactation. Increased energy expenditure and a favorable hormonal and adipokine profile during lactation may explain these associations.
Collapse
Affiliation(s)
- Georgia Zachou
- 2nd Department of Obstetrics and Gynecology, Medical School, University of Athens, Aretaieio Hospital, 76 Vas. Sofias Str., GR 11528, Athens, Greece
| | - Eleni Armeni
- 2nd Department of Obstetrics and Gynecology, Medical School, University of Athens, Aretaieio Hospital, 76 Vas. Sofias Str., GR 11528, Athens, Greece
| | - Irene Lambrinoudaki
- 2nd Department of Obstetrics and Gynecology, Medical School, University of Athens, Aretaieio Hospital, 76 Vas. Sofias Str., GR 11528, Athens, Greece.
| |
Collapse
|
45
|
Next-Generation Tools to Study Autonomic Regulation In Vivo. Neurosci Bull 2018; 35:113-123. [PMID: 30560436 DOI: 10.1007/s12264-018-0319-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/29/2018] [Indexed: 12/31/2022] Open
Abstract
The recent development of tools to decipher the intricacies of neural networks has improved our understanding of brain function. Optogenetics allows one to assess the direct outcome of activating a genetically-distinct population of neurons. Neurons are tagged with light-sensitive channels followed by photo-activation with an appropriate wavelength of light to functionally activate or silence them, resulting in quantifiable changes in the periphery. Capturing and manipulating activated neuron ensembles, is a recently-designed technique to permanently label activated neurons responsible for a physiological function and manipulate them. On the other hand, neurons can be transfected with genetically-encoded Ca2+ indicators to capture the interplay between them that modulates autonomic end-points or somatic behavior. These techniques work with millisecond temporal precision. In addition, neurons can be manipulated chronically to simulate physiological aberrations by transfecting designer G-protein-coupled receptors exclusively activated by designer drugs. In this review, we elaborate on the fundamental concepts and applications of these techniques in research.
Collapse
|
46
|
Merz T, Lukaschewski B, Wigger D, Rupprecht A, Wepler M, Gröger M, Hartmann C, Whiteman M, Szabo C, Wang R, Waller C, Radermacher P, McCook O. Interaction of the hydrogen sulfide system with the oxytocin system in the injured mouse heart. Intensive Care Med Exp 2018; 6:41. [PMID: 30341744 PMCID: PMC6195501 DOI: 10.1186/s40635-018-0207-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/07/2018] [Indexed: 02/08/2023] Open
Abstract
Background Both the hydrogen sulfide/cystathionine-γ-lyase (H2S/CSE) and oxytocin/oxytocin receptor (OT/OTR) systems have been reported to be cardioprotective. H2S can stimulate OT release, thereby affecting blood volume and pressure regulation. Systemic hyper-inflammation after blunt chest trauma is enhanced in cigarette smoke (CS)-exposed CSE−/− mice compared to wildtype (WT). CS increases myometrial OTR expression, but to this point, no data are available on the effects CS exposure on the cardiac OT/OTR system. Since a contusion of the thorax (Txt) can cause myocardial injury, the aim of this post hoc study was to investigate the effects of CSE−/− and exogenous administration of GYY4137 (a slow release H2S releasing compound) on OTR expression in the heart, after acute on chronic disease, of CS exposed mice undergoing Txt. Methods This study is a post hoc analysis of material obtained in wild type (WT) homozygous CSE−/− mice after 2-3 weeks of CS exposure and subsequent anesthesia, blast wave-induced TxT, and surgical instrumentation for mechanical ventilation (MV) and hemodynamic monitoring. CSE−/− animals received a 50 μg/g GYY4137-bolus after TxT. After 4h of MV, animals were exsanguinated and organs were harvested. The heart was cut transversally, formalin-fixed, and paraffin-embedded. Immunohistochemistry for OTR, arginine-vasopressin-receptor (AVPR), and vascular endothelial growth factor (VEGF) was performed with naïve animals as native controls. Results CSE−/− was associated with hypertension and lower blood glucose levels, partially and significantly restored by GYY4137 treatment, respectively. Myocardial OTR expression was reduced upon injury, and this was aggravated in CSE−/−. Exogenous H2S administration restored myocardial protein expression to WT levels. Conclusions This study suggests that cardiac CSE regulates cardiac OTR expression, and this effect might play a role in the regulation of cardiovascular function.
Collapse
Affiliation(s)
- Tamara Merz
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.
| | - Britta Lukaschewski
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Daniela Wigger
- Clinic for Psychsomatic Medicine and Psychotherapy, University Medical Center, Ulm, Germany
| | - Aileen Rupprecht
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Martin Wepler
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.,Department of Anesthesiology, University Medical Center, Ulm, Germany
| | - Michael Gröger
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Clair Hartmann
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.,Department of Anesthesiology, University Medical Center, Ulm, Germany
| | - Matthew Whiteman
- University of Exeter Medical School, St. Luke's Campus, Exeter, England, UK
| | - Csaba Szabo
- Chair of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.,Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rui Wang
- Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Christiane Waller
- Clinic for Psychsomatic Medicine and Psychotherapy, University Medical Center, Ulm, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Paracelsus Medical University, Nuremberg General Hospital, Nuremberg, Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Oscar McCook
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| |
Collapse
|
47
|
Ruyle BC, Klutho PJ, Baines CP, Heesch CM, Hasser EM. Hypoxia activates a neuropeptidergic pathway from the paraventricular nucleus of the hypothalamus to the nucleus tractus solitarii. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1167-R1182. [PMID: 30230933 DOI: 10.1152/ajpregu.00244.2018] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The paraventricular nucleus of the hypothalamus (PVN) contributes to both autonomic and neuroendocrine function. PVN lesion or inhibition blunts cardiorespiratory responses to peripheral chemoreflex activation, suggesting that the PVN is required for full expression of these effects. However, the role of efferent projections to cardiorespiratory nuclei and the neurotransmitters/neuromodulators that are involved is unclear. The PVN sends dense projections to the nucleus tractus solitarii (nTS), a region that displays neuronal activation following hypoxia. We hypothesized that acute hypoxia activates nTS-projecting PVN neurons. Using a combination of retrograde tracing and immunohistochemistry, we determined whether hypoxia activates PVN neurons that project to the nTS and examined the phenotype of these neurons. Conscious rats underwent 2 h normoxia (21% O2, n = 5) or hypoxia (10% O2, n = 6). Hypoxia significantly increased Fos immunoreactivity in nTS-projecting neurons, primarily in the caudal PVN. The majority of activated nTS-projecting neurons contained corticotropin-releasing hormone (CRH). In the nTS, fibers expressing the CRH receptor corticotropin-releasing factor receptor 2 (CRFR2) were colocalized with oxytocin (OT) fibers and were closely associated with hypoxia-activated nTS neurons. A separate group of animals that received a microinjection of adeno-associated virus type 2-hSyn-green fluorescent protein (GFP) into the PVN exhibited GFP-expressing fibers in the nTS; a proportion of these fibers displayed OT immunoreactivity. Thus, nTS CRFR2s appear to be located on the fibers of PVN OT neurons that project to the nTS. Taken together, our findings suggest that PVN CRH projections to the nTS may modulate nTS neuronal activation, possibly via OTergic mechanisms, and thus contribute to chemoreflex cardiorespiratory responses.
Collapse
Affiliation(s)
- Brian C Ruyle
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Paula J Klutho
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri
| | - Christopher P Baines
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri
| | - Cheryl M Heesch
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri
| | - Eileen M Hasser
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri.,Dalton Cardiovascular Research Center, University of Missouri , Columbia, Missouri.,Department of Medical Pharmacology and Physiology, University of Missouri , Columbia, Missouri
| |
Collapse
|
48
|
Park S, Choi NK. Breastfeeding and Maternal Age-related Cataract. Am J Ophthalmol 2018; 192:124-130. [PMID: 29806992 DOI: 10.1016/j.ajo.2018.05.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE No studies addressed the influence of breastfeeding on cataract formation. The objective of this study was to address the relationship between breastfeeding and maternal age-related cataract. DESIGN A nationwide cross-sectional study. METHODS This study analyzed data for 3821 parous women aged 50 years or above in the Korea National Health and Nutrition Examination Survey 2010-2012. Participants were aggregated into quartiles according to the number of breastfed children and duration of breastfeeding. Logistic regression analysis was used to examine the relationships between less or short duration of breastfeeding and increased risks of cataract. RESULTS A total of 2197 women (57.5%) were classified as having age-related cataract. Women who breastfed 4-12 children (odds ratio [OR] = 0.56; 95% confidence interval [CI] = 0.35-0.89) had significantly lower risks for cortical cataract, compared to those who breastfed no or 1 child (P for trend across quartiles = .010). Women who breastfed for 36-60 months (OR = 0.61, 95% CI = 0.42-0.90) or 61-324 months (OR = 0.53, 95% CI = 0.33-0.83) had lower risks for cortical cataract compared to those who breastfed 16 months or less (P for trend across tertiles = .003). The population-attributable fractions of cortical cataract induced by number of children breastfed less than 3 and duration of breastfeeding less than 36 months were 9.4% (95% CI = 1.3%-17.6%) and 10.7% (95% CI = 3.0%-18.4%), respectively. CONCLUSIONS Breastfeeding more children and long-term breastfeeding were associated with lower risk of cortical cataract formation in parous women.
Collapse
Affiliation(s)
- Sangshin Park
- Center for International Health Research, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA; Department of Pediatrics, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Nam-Kyong Choi
- Department of Health Convergence, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
49
|
Li T, Chen Y, Gua C, Wu B. Elevated Oxidative Stress and Inflammation in Hypothalamic Paraventricular Nucleus Are Associated With Sympathetic Excitation and Hypertension in Rats Exposed to Chronic Intermittent Hypoxia. Front Physiol 2018; 9:840. [PMID: 30026701 PMCID: PMC6041405 DOI: 10.3389/fphys.2018.00840] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/14/2018] [Indexed: 01/08/2023] Open
Abstract
Obstructive sleep apnea (OSA), characterized by recurrent collapse of the upper airway during sleep leading to chronic intermittent hypoxia (CIH), is an independent risk factor for hypertension. Sympathetic excitation has been shown to play a major role in the pathogenesis of OSA-associated hypertension. Accumulating evidence indicates that oxidative stress and inflammation in the hypothalamic paraventricular nucleus (PVN), a critical cardiovascular and autonomic center, mediate sympathetic excitation in many cardiovascular diseases. Here we tested the hypothesis that CIH elevates oxidative stress and inflammation in the PVN, which might be associated with sympathetic excitation and increased blood pressure in a rat model of CIH that mimics the oxygen profile in patients with OSA. Sprague-Dawley rats were pretreated with intracerebroventricular (ICV) infusion of vehicle or superoxide scavenger tempol, and then exposed to control or CIH for 7 days. Compared with control+vehicle rats, CIH+vehicle rats exhibited increased blood pressure, and increased sympathetic drive as indicated by the blood pressure response to ganglionic blockade and plasma norepinephrine levels. Pretreatment with ICV tempol prevented CIH-induced increases in blood pressure and sympathetic drive. Molecular studies revealed that expression of NAD(P)H oxidase subunits, production of reactive oxygen species, expression of proinflammatory cytokines and neuronal excitation in the PVN were elevated in CIH+vehicle rats, compared with control+vehicle rats, but were normalized or reduced in CIH rat pretreated with ICV tempol. Notably, CIH+vehicle rats also had increased systemic oxidative stress and inflammation, which were not altered by ICV tempol. The results suggest that CIH induces elevated oxidative stress and inflammation in the PVN, which lead to PVN neuronal excitation and are associated with sympathetic excitation and increased blood pressure. Central oxidative stress and inflammation may be novel targets for the prevention and treatment of hypertension in patients with OSA.
Collapse
Affiliation(s)
- Tiejun Li
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yanli Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, China
| | - Chaojun Gua
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Baogang Wu
- Department of Cardiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Jurek B, Neumann ID. The Oxytocin Receptor: From Intracellular Signaling to Behavior. Physiol Rev 2018; 98:1805-1908. [DOI: 10.1152/physrev.00031.2017] [Citation(s) in RCA: 408] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The many facets of the oxytocin (OXT) system of the brain and periphery elicited nearly 25,000 publications since 1930 (see FIGURE 1 , as listed in PubMed), which revealed central roles for OXT and its receptor (OXTR) in reproduction, and social and emotional behaviors in animal and human studies focusing on mental and physical health and disease. In this review, we discuss the mechanisms of OXT expression and release, expression and binding of the OXTR in brain and periphery, OXTR-coupled signaling cascades, and their involvement in behavioral outcomes to assemble a comprehensive picture of the central and peripheral OXT system. Traditionally known for its role in milk let-down and uterine contraction during labor, OXT also has implications in physiological, and also behavioral, aspects of reproduction, such as sexual and maternal behaviors and pair bonding, but also anxiety, trust, sociability, food intake, or even drug abuse. The many facets of OXT are, on a molecular basis, brought about by a single receptor. The OXTR, a 7-transmembrane G protein-coupled receptor capable of binding to either Gαior Gαqproteins, activates a set of signaling cascades, such as the MAPK, PKC, PLC, or CaMK pathways, which converge on transcription factors like CREB or MEF-2. The cellular response to OXT includes regulation of neurite outgrowth, cellular viability, and increased survival. OXTergic projections in the brain represent anxiety and stress-regulating circuits connecting the paraventricular nucleus of the hypothalamus, amygdala, bed nucleus of the stria terminalis, or the medial prefrontal cortex. Which OXT-induced patterns finally alter the behavior of an animal or a human being is still poorly understood, and studying those OXTR-coupled signaling cascades is one initial step toward a better understanding of the molecular background of those behavioral effects.
Collapse
Affiliation(s)
- Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Inga D. Neumann
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| |
Collapse
|