1
|
Chen X, Yan X, Yu C, Chen QH, Bi L, Shan Z. PTSD Increases Risk for Hypertension Development Through PVN Activation and Vascular Dysfunction in Sprague Dawley Rats. Antioxidants (Basel) 2024; 13:1423. [PMID: 39594564 PMCID: PMC11590931 DOI: 10.3390/antiox13111423] [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: 09/06/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
This study investigates the impact of single prolonged stress (SPS), a model of post-traumatic stress disorder (PTSD), on cardiovascular responses, hypothalamic paraventricular nucleus (PVN) activity, and vascular function to elucidate the mechanisms linking traumatic stress to hypertension. Although SPS did not directly cause chronic hypertension in male Sprague Dawley (SD) rats, it induced acute but transient increases in blood pressure and heart rate and significantly altered the expression of hypertension-associated genes, such as vasopressin, angiotensin II type 1 receptor (AT1R), and FOSL1 in the PVN. Notably, mitochondrial reactive oxygen species (mtROS) were predominantly elevated in the pre-autonomic regions of the PVN, colocalizing with AT1R- and FOSL1-expressing cells, suggesting that oxidative stress may amplify sympathetic activation and stress responses. SPS also increased mRNA levels of pro-inflammatory cytokines (TNFα and IL1β) and inducible nitric oxide synthase (iNOS) in the aorta, and impaired vascular reactivity to vasoconstrictor and vasodilator stimuli, reflecting compromised vascular function. These findings suggest that SPS-sensitize neuroendocrine, autonomic, and vascular pathways create a state of cardiovascular vulnerability that could predispose individuals to hypertension when exposed to additional stressors. Understanding these mechanisms provides critical insights into the pathophysiology of stress-related cardiovascular disorders and underscores the need for targeted therapeutic interventions that address oxidative stress and modulate altered PVN pathways to mitigate the cardiovascular impact of PTSD and related conditions.
Collapse
Affiliation(s)
- Xinqian Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA; (X.C.); (Q.-h.C.)
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
| | - Xin Yan
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA
| | - Chunxiu Yu
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Qing-hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA; (X.C.); (Q.-h.C.)
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
| | - Lanrong Bi
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
- Department of Chemistry, Michigan Technological University, Houghton, MI 49931, USA
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Michigan Technological University, Houghton, MI 49931, USA; (X.C.); (Q.-h.C.)
- Health Research Institute, Michigan Technological University, Houghton, MI 49931, USA; (X.Y.); (C.Y.)
| |
Collapse
|
2
|
Li X, He Y, Wang D, Momeni MR. Chronobiological disruptions: unravelling the interplay of shift work, circadian rhythms, and vascular health in the context of stroke risk. Clin Exp Med 2024; 25:6. [PMID: 39541048 PMCID: PMC11564290 DOI: 10.1007/s10238-024-01514-w] [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: 06/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Shift work, particularly night shifts, disrupts circadian rhythms and increases stroke risk. This manuscript explores the mechanisms connecting shift work with stroke, focusing on circadian rhythms, hypertension, and diabetes. The circadian system, controlled by different mechanisms including central and peripheral clock genes, suprachiasmatic nuclei (SCN), and pineal gland (through melatonin production), regulates body functions and responds to environmental signals. Disruptions in this system affect endothelial cells, leading to blood pressure issues. Type 2 diabetes mellitus (T2DM) is significantly associated with night shifts, with circadian disturbances affecting glucose metabolism, insulin sensitivity, and hormone regulation. The manuscript examines the relationship between melatonin, insulin, and glucose balance, highlighting pathways that link T2DM to stroke risk. Additionally, dyslipidemia, particularly reduced HDL-c levels, results from shift work and contributes to stroke development. High lipid levels cause oxidative stress, inflammation, and endothelial dysfunction, increasing cerebrovascular risks. The manuscript details the effects of dyslipidemia on brain functions, including disruptions in blood flow, blood-brain barrier integrity, and neural cell death. This comprehensive analysis emphasizes the complex interplay of circadian disruption, hypertension, diabetes, and dyslipidemia in increasing stroke risk among shift workers. Understanding these mechanisms is essential for developing targeted interventions to reduce stroke susceptibility and improve cerebrovascular health in this vulnerable population.
Collapse
Affiliation(s)
- Xiaohong Li
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yanjin He
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Dawu Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | | |
Collapse
|
3
|
Anderson HA, Robilotto GL, Mickle AD. Role of local angiotensin II signaling in bladder function. Am J Physiol Renal Physiol 2024; 327:F726-F738. [PMID: 39265080 PMCID: PMC11563647 DOI: 10.1152/ajprenal.00204.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/14/2024] Open
Abstract
Angiotensin II signaling plays a crucial role in many different diseases. Although it has been known for several decades that local angiotensin II signaling molecules are present in the bladder, the understanding of their functions there is still limited, especially compared with other organ systems such as cardiovascular and respiratory systems. This article reviews current literature regarding local angiotensin II signaling in the urinary bladder. By reviewing several decades of literature, the field has provided strong evidence to support the presence of local angiotensin II signaling in the bladder, including the expression of angiotensin type 1 receptor and angiotensin type 2 receptor in both human and animal tissues. In addition, evidence suggests a functional role of angiotensin type 1 receptor in mediating bladder contractions. In bladder disease models, angiotensin II signaling can be upregulated, and angiotensin type 1 receptor activity is associated with increases in inflammation, fibrosis, and oxidative stress. We also address the gaps in knowledge that remain in understanding local angiotensin II signaling in the bladder, including limitations on clinical translatability. Although there is a strong foundation regarding the local presence and role of angiotensin II signaling in the bladder, further research is needed to support translational applications.
Collapse
Affiliation(s)
- Hannah A Anderson
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Gabriella L Robilotto
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States
| | - Aaron D Mickle
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, Florida, United States
- Department of Neurosurgery, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Urology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
4
|
Ashraf N, Van Nostrand JL. Fine-tuning AMPK in physiology and disease using point-mutant mouse models. Dis Model Mech 2024; 17:dmm050798. [PMID: 39136185 PMCID: PMC11340815 DOI: 10.1242/dmm.050798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Abstract
AMP-activated protein kinase (AMPK) is an evolutionarily conserved serine/threonine kinase that monitors the cellular energy status to adapt it to the fluctuating nutritional and environmental conditions in an organism. AMPK plays an integral part in a wide array of physiological processes, such as cell growth, autophagy and mitochondrial function, and is implicated in diverse diseases, including cancer, metabolic disorders, cardiovascular diseases and neurodegenerative diseases. AMPK orchestrates many different physiological outcomes by phosphorylating a broad range of downstream substrates. However, the importance of AMPK-mediated regulation of these substrates in vivo remains an ongoing area of investigation to better understand its precise role in cellular and metabolic homeostasis. Here, we provide a comprehensive overview of our understanding of the kinase function of AMPK in vivo, as uncovered from mouse models that harbor phosphorylation mutations in AMPK substrates. We discuss some of the inherent limitations of these mouse models, highlight the broader implications of these studies for understanding human health and disease, and explore the valuable insights gained that could inform future therapeutic strategies for the treatment of metabolic and non-metabolic disorders.
Collapse
Affiliation(s)
- Naghmana Ashraf
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jeanine L. Van Nostrand
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Sabarathinam S. Deciphering the gut microbiota's (Coprococcus and Subdoligranulum) impact on depression: Network pharmacology and molecular dynamics simulation. Pharmacol Biochem Behav 2024; 241:173805. [PMID: 38848976 DOI: 10.1016/j.pbb.2024.173805] [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: 04/16/2024] [Revised: 05/24/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
Depression, a prevalent mental health condition, significantly impacts global mental impairment rates. While antidepressants are commonly used, treatment-resistant depression (TRD) poses a challenge. Emerging research highlights the role of the gut microbiota in depression through the gut-brain axis. This study identifies key genes associated with depression influenced by specific gut microbiota, Coprococcus and Subdoligranulum. Using bioinformatics tools, potential targets were elucidated, and molecular docking studies were performed. Furthermore, gene expression analysis identified hub-genes related to depression, intersecting with metabolite targets. Protein-protein interaction analysis revealed pivotal targets such as PTGS2 and MMP9. Molecular docking demonstrated 3-Indolepropionic acid's superior affinity over (R)-3-(4-Hydroxyphenyl)lactate. Physicochemical properties and toxicity profiles were compared, suggesting favorable attributes for 3-Indolepropionic acid. Molecular dynamics simulations confirmed stability and interactions of compounds with target proteins. This comprehensive approach sheds light on the complex interplay between gut microbiota, genes, and depression, emphasizing the potential for microbiota-targeted interventions in mental health management.
Collapse
Affiliation(s)
- Sarvesh Sabarathinam
- Interdisciplinary Institute of Indian System of Medicine (IIISM), SRM Institute of Science & Technology, Kattankulathur, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
6
|
Szczepanska-Sadowska E, Czarzasta K, Bogacki-Rychlik W, Kowara M. The Interaction of Vasopressin with Hormones of the Hypothalamo-Pituitary-Adrenal Axis: The Significance for Therapeutic Strategies in Cardiovascular and Metabolic Diseases. Int J Mol Sci 2024; 25:7394. [PMID: 39000501 PMCID: PMC11242374 DOI: 10.3390/ijms25137394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
A large body of evidence indicates that vasopressin (AVP) and steroid hormones are frequently secreted together and closely cooperate in the regulation of blood pressure, metabolism, water-electrolyte balance, and behavior, thereby securing survival and the comfort of life. Vasopressin cooperates with hormones of the hypothalamo-pituitary-adrenal axis (HPA) at several levels through regulation of the release of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and multiple steroid hormones, as well as through interactions with steroids in the target organs. These interactions are facilitated by positive and negative feedback between specific components of the HPA. Altogether, AVP and the HPA cooperate closely as a coordinated functional AVP-HPA system. It has been shown that cooperation between AVP and steroid hormones may be affected by cellular stress combined with hypoxia, and by metabolic, cardiovascular, and respiratory disorders; neurogenic stress; and inflammation. Growing evidence indicates that central and peripheral interactions between AVP and steroid hormones are reprogrammed in cardiovascular and metabolic diseases and that these rearrangements exert either beneficial or harmful effects. The present review highlights specific mechanisms of the interactions between AVP and steroids at cellular and systemic levels and analyses the consequences of the inappropriate cooperation of various components of the AVP-HPA system for the pathogenesis of cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | | | | | | |
Collapse
|
7
|
Kango G, Malek R, Mannuel H, Hussain A. Targeting androgen biosynthesis in prostate cancer: implications on endocrine physiology. Curr Opin Oncol 2024; 36:195-201. [PMID: 38573209 DOI: 10.1097/cco.0000000000001032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
PURPOSE OF REVIEW Targeting specific steroidogenic enzymes is effective in decreasing testosterone synthesis, resulting in significant antitumor effects in prostate cancer. Such treatments result in disruptions of complicated and intertwining pathways with systemic physiologic consequences via effects on the adrenal gland and renin-angiotensin-aldosterone axis. This review highlights some of these aspects that need to be taken into consideration when treating patients with androgen biosynthesis inhibitors. RECENT FINDINGS Targeting CYP17A1, a key enzyme involved in androgen biosynthesis, is a well established treatment in prostate cancer. More recently, efforts are underway to target a gatekeeper enzyme of steroidogenesis, CYP11A1. This enzyme mediates conversion of cholesterol to pregnenolone, the first step in steroid hormone biogenesis. Studies are beginning to demonstrate antitumor effects of ODM-208, a CYP11A1 inhibitor in prostate cancer. Although anticipated to have a therapeutic role in prostate cancer, there are potential downstream effects of CYP11A1 targeting arising from suppression of the entire adrenal cortex, including long-term adrenal insufficiency and possibly cardiovascular dysregulation. SUMMARY Agents targeting androgen biosynthesis can have systemic implications. Balancing management of prostate cancer with better understanding of the mechanisms associated with potential side effects will allow for patients to obtain improved antitumor benefit while mitigating against treatment-associated adverse effects.
Collapse
Affiliation(s)
- Ghazal Kango
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
| | - Rana Malek
- Department of Medicine University of Maryland School of Medicine
- Division of Endocrinology
| | - Heather Mannuel
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
- Baltimore VA Medical Center, Baltimore, Maryland, USA
| | - Arif Hussain
- University of Maryland Greenebaum Comprehensive Cancer Center
- Department of Medicine University of Maryland School of Medicine
- Department of Pathology
- Department of Biochemistry and Molecular Biology
- Baltimore VA Medical Center, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Xu T, Chen Z, Zhou X, Wang L, Zhou F, Yao D, Zhou B, Becker B. The central renin-angiotensin system: A genetic pathway, functional decoding, and selective target engagement characterization in humans. Proc Natl Acad Sci U S A 2024; 121:e2306936121. [PMID: 38349873 PMCID: PMC10895353 DOI: 10.1073/pnas.2306936121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 01/02/2024] [Indexed: 02/15/2024] Open
Abstract
Accumulating evidence suggests that the brain renin angiotensin system (RAS) plays a pivotal role in the regulation of cognition and behavior as well as in the neuropathology of neurological and mental disorders. The angiotensin II type 1 receptor (AT1R) mediates most functional and neuropathology-relevant actions associated with the central RAS. However, an overarching comprehension to guide translation and utilize the therapeutic potential of the central RAS in humans is currently lacking. We conducted a comprehensive characterization of the RAS using an innovative combination of transcriptomic gene expression mapping, image-based behavioral decoding, and pre-registered randomized controlled discovery-replication pharmacological resting-state functional magnetic resonance imaging (fMRI) trials (N = 132) with a selective AT1R antagonist. The AT1R exhibited a particular dense expression in a subcortical network encompassing the thalamus, striatum, and amygdalo-hippocampal formation. Behavioral decoding of the AT1R gene expression brain map showed an association with memory, stress, reward, and motivational processes. Transient pharmacological blockade of the AT1R further decreased neural activity in subcortical systems characterized by a high AT1R expression, while increasing functional connectivity in the cortico-basal ganglia-thalamo-cortical circuitry. Effects of AT1R blockade on the network level were specifically associated with the transcriptomic signatures of the dopaminergic, opioid, acetylcholine, and corticotropin-releasing hormone signaling systems. The robustness of the results was supported in an independent pharmacological fMRI trial. These findings present a biologically informed comprehensive characterization of the central AT1R pathways and their functional relevance on the neural and behavioral level in humans.
Collapse
Affiliation(s)
- Ting Xu
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Zhiyi Chen
- Experimental Research Center for Medical and Psychological Science, School of Psychology, Third Military Medical University, Chongqing400037, People’s Republic of China
- Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
| | - Xinqi Zhou
- Institute of Brain and Psychological Sciences, Sichuan Normal University, Chengdu 610066, People’s Republic of China
| | - Lan Wang
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Feng Zhou
- Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
- Key Laboratory of Cognition and Personality, Ministry of Education, Faculty of Psychology, Southwest University, Chongqing400715, People’s Republic of China
| | - Dezhong Yao
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
| | - Bo Zhou
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
| | - Benjamin Becker
- The Center of Psychosomatic Medicine, Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu610054, People’s Republic of China
- Ministry of Education Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology, Chengdu610054, People’s Republic of China
- The State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong999077, People’s Republic of China
- Department of Psychology, The University of Hong Kong, Hong Kong999077, People’s Republic of China
| |
Collapse
|
9
|
Bhullar SK, Dhalla NS. Adaptive and maladaptive roles of different angiotensin receptors in the development of cardiac hypertrophy and heart failure. Can J Physiol Pharmacol 2024; 102:86-104. [PMID: 37748204 DOI: 10.1139/cjpp-2023-0226] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
Angiotensin II (Ang II) is formed by the action of angiotensin-converting enzyme (ACE) in the renin-angiotensin system. This hormone is known to induce cardiac hypertrophy and heart failure and its actions are mediated by the interaction of both pro- and antihypertrophic Ang II receptors (AT1R and AT2R). Ang II is also metabolized by ACE 2 to Ang-(1-7), which elicits the activation of Mas receptors (MasR) for inducing antihypertrophic actions. Since heart failure under different pathophysiological situations is preceded by adaptive and maladaptive cardiac hypertrophy, we have reviewed the existing literature to gain some information regarding the roles of AT1R, AT2R, and MasR in both acute and chronic conditions of cardiac hypertrophy. It appears that the activation of AT1R may be involved in the development of adaptive and maladaptive cardiac hypertrophy as well as subsequent heart failure because both ACE inhibitors and AT1R antagonists exert beneficial effects. On the other hand, the activation of both AT2R and MasR may prevent the occurrence of maladaptive cardiac hypertrophy and delay the progression of heart failure, and thus therapy with different activators of these antihypertrophic receptors under chronic pathological stages may prove beneficial. Accordingly, it is suggested that a great deal of effort should be made to develop appropriate activators of both AT2R and MasR for the treatment of heart failure subjects.
Collapse
Affiliation(s)
- Sukhwinder K Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Naranjan S Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre and Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
10
|
Szczepanska-Sadowska E. Interplay of Angiotensin Peptides, Vasopressin, and Insulin in the Heart: Experimental and Clinical Evidence of Altered Interactions in Obesity and Diabetes Mellitus. Int J Mol Sci 2024; 25:1310. [PMID: 38279313 PMCID: PMC10816525 DOI: 10.3390/ijms25021310] [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: 12/20/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
The present review draws attention to the specific role of angiotensin peptides [angiotensin II (Ang II), angiotensin-(1-7) (Ang-(1-7)], vasopressin (AVP), and insulin in the regulation of the coronary blood flow and cardiac contractions. The interactions of angiotensin peptides, AVP, and insulin in the heart and in the brain are also discussed. The intracardiac production and the supply of angiotensin peptides and AVP from the systemic circulation enable their easy access to the coronary vessels and the cardiomyocytes. Coronary vessels and cardiomyocytes are furnished with AT1 receptors, AT2 receptors, Ang (1-7) receptors, vasopressin V1 receptors, and insulin receptor substrates. The presence of some of these molecules in the same cells creates good conditions for their interaction at the signaling level. The broad spectrum of actions allows for the engagement of angiotensin peptides, AVP, and insulin in the regulation of the most vital cardiac processes, including (1) cardiac tissue oxygenation, energy production, and metabolism; (2) the generation of the other cardiovascular compounds, such as nitric oxide, bradykinin (Bk), and endothelin; and (3) the regulation of cardiac work by the autonomic nervous system and the cardiovascular neurons of the brain. Multiple experimental studies and clinical observations show that the interactions of Ang II, Ang(1-7), AVP, and insulin in the heart and in the brain are markedly altered during heart failure, hypertension, obesity, and diabetes mellitus, especially when these diseases coexist. A survey of the literature presented in the review provides evidence for the belief that very individualized treatment, including interactions of angiotensins and vasopressin with insulin, should be applied in patients suffering from both the cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
11
|
Cosentino N, Marenzi G, Muratori M, Magrì D, Cattadori G, Agostoni P. Fluid balance in heart failure. Eur J Prev Cardiol 2023; 30:ii9-ii15. [PMID: 37819223 DOI: 10.1093/eurjpc/zwad166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 10/13/2023]
Abstract
Fluid retention is a major determinant of symptoms in patients with heart failure (HF), and it is closely associated with prognosis. Hence, congestion represents a critical therapeutic target in this clinical setting. The first therapeutic strategy in HF patients with fluid overload is optimization of diuretic intervention to maximize water and sodium excretion. When diuretic therapy fails to relieve congestion, renal replacement therapy represents the only alternative option for fluid removal, as well as a way to restore diuretic responsiveness. On this background, the pathophysiology of fluid balance in HF is complex, with heart, kidney, and lung being deeply involved in volume regulation and management. Therefore, the interplay between these organs should be appreciated and considered when fluid overload in HF patients is targeted.
Collapse
Affiliation(s)
- Nicola Cosentino
- Centro Cardiologico Monzino, I.R.C.C.S., Via Parea 4, Milan 20138, Italy
- Cardiovascular Section, Department of Clinical Sciences and Community Health, University of Milan, Milan 20122, Italy
| | - Giancarlo Marenzi
- Centro Cardiologico Monzino, I.R.C.C.S., Via Parea 4, Milan 20138, Italy
| | - Manuela Muratori
- Centro Cardiologico Monzino, I.R.C.C.S., Via Parea 4, Milan 20138, Italy
| | - Damiano Magrì
- Department of Clinical and Molecular Medicine, Azienda Ospedaliera Sant'Andrea, 'Sapienza' Università degli Studi di Roma, Roma 00198, Italy
| | - Gaia Cattadori
- Unità Operativa Cardiologia Riabilitativa, IRCCS Multimedica, Milan 20123, Italy
| | | |
Collapse
|
12
|
Bo X, Liu Y, Hao C, Qian H, Zhao Y, Hu Y, Zhang Y, Kharbuja N, Ju C, Chen L, Ma G. Risk stratification and predictive value of serum sodium fluctuation for adverse prognosis in acute coronary syndrome patients. Clin Chim Acta 2023; 548:117491. [PMID: 37454722 DOI: 10.1016/j.cca.2023.117491] [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: 03/03/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Serum sodium fluctuation (SF) as an indicator of the extent of changes in serum sodium is associated with increased mortality in hospitalized patients. However, there is no consensus on diagnostic criteria for SF, and its impact on the outcome of patients with acute coronary syndrome (ACS) remains uncertain. We defined SF and assessed its association with adverse prognosis in hospitalized ACS patients. METHODS Patients diagnosed with ACS were consecutively recruited. The serum SF rate (SFR) was defined as the ratio of the difference between the highest and lowest serum sodium levels during hospitalization to the initial serum sodium level on admission. The Cox proportional hazards model was performed to evaluate the association between SFR and mortality. The dose-response relationships of SFR with mortality was characterized by restricted cubic splines (RCS) model. The predictive performance of SF for mortality was assessed by the area under the receiver operating characteristic curves (AUCs). RESULTS The study retrospectively enrolled 1856 ACS patients, of which 36 (1.94%) patients dead within 1 year. Multivariate Cox analysis showed that SFR was independently associated with higher risk of 1-year mortality (HR = 1.17, 95% CI: 1.111-1.244, P < 0.001). RCS analysis showed the optimal threshold for SFR was 5%, and the 1-year cumulative mortality was higher in the abnormal SF group (SFR ≥ 5%) compared with the normal SF group (SFR < 5%, P < 0.01). The AUCs of SF for predicting mortality within 1 month, 6 months, and 1 year were 0.842 (95% CI: 0.781-0.904), 0.830 (95% CI:0.736-0.926), 0.703 (95% CI:0.595--0.811), respectively. Even in patients with normal baseline serum sodium, abnormal SF group demonstrated a significantly higher 1-year mortality compared to normal SF group (HR = 4.955, 95% CI: 1.919-12.795). CONCLUSION The SFR during hospitalization is an adequate predictor of adverse outcomes in ACS patients, independent of serum sodium level at admission. Additional research is warranted to ascertain whether interventions targeting SF confer measurable clinical benefits.
Collapse
Affiliation(s)
- Xiangwei Bo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Yang Liu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Chunshu Hao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Hao Qian
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Yuanyuan Zhao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Ya Hu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | | | - Chengwei Ju
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| | - Lijuan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China.
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, PR China; School of Medicine, Southeast University, Nanjing, 210009, PR China
| |
Collapse
|
13
|
Ali F, Wang D, Cheng Y, Wu M, Saleem MZ, Wei L, Xie Y, Yan M, Chu J, Yang Y, Shen A, Peng J. Quercetin attenuates angiotensin II-induced proliferation of vascular smooth muscle cells and p53 pathway activation in vitro and in vivo. Biofactors 2023; 49:956-970. [PMID: 37296538 DOI: 10.1002/biof.1959] [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: 01/10/2023] [Accepted: 04/23/2023] [Indexed: 06/12/2023]
Abstract
Quercetin is an essential flavonoid mostly found in herbal plants, fruits, and vegetables, which exhibits anti-hypertension properties. However, its pharmacological impact on angiotensin II (Ang II) induced the increase of blood pressure along with in-depth mechanism needs further exploration. The present study pointed out the anti-hypertensive role of quercetin and its comprehensive fundamental mechanisms. Our data showed that quercetin treatment substantially reduced the increase in blood pressure, pulse wave velocity, and aortic thickness of abdominal aorta in Ang II-infused C57BL/6 mice. RNA sequencing revealed that quercetin treatment reversed 464 differentially expressed transcripts in the abdominal aorta of Ang II-infused mice. Moreover, overlapping KEGG-enriched signaling pathways identified multiple common pathways between the comparison of Ang II versus control and Ang II + quercetin versus Ang II. Likewise, these pathways included cell cycle as well as p53 pathways. Transcriptome was further validated by immunohistochemistry, indicating that quercetin treatment significantly decreased the Ang II-induced expression of proliferating cell nuclear antigen (PCNA), cyclin-dependent kinase-4 (CDK4), and cyclin D1, while increased protein expression of p53, and p21 in abdominal aortic tissues of mice. In vitro, quercetin treatment meaningfully decreased the cell viability, arrested cell cycle at G0/G1 phase, and up-regulated the p53 and p21 proteins expression, as well as down-regulated the protein expression of cell cycle-related markers, for example, CDK4, cyclin D1 in Ang II stimulated vascular smooth muscle cells (VSMCs). This study addresses pharmacologic and mechanistic perspectives of quercetin against Ang-II-induced vascular injury and the increase of blood pressure.
Collapse
Affiliation(s)
- Farman Ali
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Di Wang
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Ying Cheng
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Meizhu Wu
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Muhammad Zubair Saleem
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| | - Lihui Wei
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- Center for Innovation and Transformation of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yi Xie
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Mengchao Yan
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Jiangfeng Chu
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Yanyan Yang
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- Center for Innovation and Transformation of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Aling Shen
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- Center for Innovation and Transformation of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jun Peng
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| |
Collapse
|
14
|
Maciejewska M, Stec A, Zaremba M, Maciejewski C, Rudnicka L, Sikora M. Copeptin as a Biomarker of Microcirculation Alterations in Systemic Sclerosis. Clin Cosmet Investig Dermatol 2023; 16:1351-1361. [PMID: 37255624 PMCID: PMC10226486 DOI: 10.2147/ccid.s409490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/03/2023] [Indexed: 06/01/2023]
Abstract
Background Systemic sclerosis is a connective tissue disease characterized by vasculopathy and progressive fibrosis, leading to multiorgan dysfunction. Given the complex and not fully elucidated pathogenesis, biomarkers of rapid disease progression and therapeutic response are lacking. Copeptin, which reflects vasopressin activity in serum, is used in diagnosing or prognosing different cardiometabolic conditions. Objective The aim of study was to investigate the concentration of copeptin in patients with systemic sclerosis and correlate it with specific clinical symptoms. Patients and Methods Serum copeptin was measured in patients with systemic sclerosis (34 women and 3 men; mean age 57.6 years) and in healthy individuals (n=30) using commercially available ELISA kits. According to the criteria of LeRoy our systemic sclerosis cohort consisted of 17 patients with limited cutaneous systemic sclerosis (45.9%) and 20 diffuse cutaneous systemic sclerosis patients (54.1%). According to the criteria of LeRoy our systemic sclerosis cohort consisted of 17 patients with limited cutaneous systemic sclerosis (45.9%) and 20 diffuse cutaneous systemic sclerosis patients (54.1%). The median duration of the disease was 10 [4-14] years. Results We found significantly higher copeptin concentration in patients with systemic sclerosis (4.21 pmol/L [3.04-5.42]) in comparison to control group (3.40 pmol/L [2.38-3.76], p<0.01). Copeptin significantly correlated with Raynaud's condition score (r=0.801, p<0.05). Patients with "late" capillaroscopic patterns had higher copeptin concentrations (5.37 pmol/L [4.29-8.06]) than patients with "early" (2.43 pmol/L [2.25-3.20], p<0.05) and "active" patterns (3.93 pmol/L [2.92-5.16], p<0.05]). Copeptin was found to be significantly higher in SSc patients with DUs (5.71 pmol/L [IQR 4.85-8.06]) when compared to SSc patients without DUs (3.31 pmol/L, [2.28-4.30], p<0.05). Additionally, copeptin concentration had good diagnostic accuracy in discriminating between patients with and without digital ulcers (AUC=0.863). Alprostadil decreased copeptin concentration from 4.96 [4.02-6.01] to 3.86 pmol/L [3.17-4.63] (p<0.01) after 4-6 cycles of administration. Conclusion Our findings suggest that copeptin may be a promising biomarker of microcirculation alterations in systemic sclerosis.
Collapse
Affiliation(s)
- Magdalena Maciejewska
- Department of Dermatology, Doctoral School of Medical University of Warsaw, Warsaw, Poland
| | - Albert Stec
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Michał Zaremba
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Cezary Maciejewski
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Sikora
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
15
|
Swiderski J, Gadanec LK, Apostolopoulos V, Moore GJ, Kelaidonis K, Matsoukas JM, Zulli A. Role of Angiotensin II in Cardiovascular Diseases: Introducing Bisartans as a Novel Therapy for Coronavirus 2019. Biomolecules 2023; 13:787. [PMID: 37238657 PMCID: PMC10216788 DOI: 10.3390/biom13050787] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the main contributors to global morbidity and mortality. Major pathogenic phenotypes of CVDs include the development of endothelial dysfunction, oxidative stress, and hyper-inflammatory responses. These phenotypes have been found to overlap with the pathophysiological complications of coronavirus disease 2019 (COVID-19). CVDs have been identified as major risk factors for severe and fatal COVID-19 states. The renin-angiotensin system (RAS) is an important regulatory system in cardiovascular homeostasis. However, its dysregulation is observed in CVDs, where upregulation of angiotensin type 1 receptor (AT1R) signaling via angiotensin II (AngII) leads to the AngII-dependent pathogenic development of CVDs. Additionally, the interaction between the spike protein of severe acute respiratory syndrome coronavirus 2 with angiotensin-converting enzyme 2 leads to the downregulation of the latter, resulting in the dysregulation of the RAS. This dysregulation favors AngII/AT1R toxic signaling pathways, providing a mechanical link between cardiovascular pathology and COVID-19. Therefore, inhibiting AngII/AT1R signaling through angiotensin receptor blockers (ARBs) has been indicated as a promising therapeutic approach to the treatment of COVID-19. Herein, we review the role of AngII in CVDs and its upregulation in COVID-19. We also provide a future direction for the potential implication of a novel class of ARBs called bisartans, which are speculated to contain multifunctional targeting towards COVID-19.
Collapse
Affiliation(s)
- Jordan Swiderski
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
- Immunology Program, Australian Institute for Musculoskeletal Science, Melbourne, VIC 3021, Australia
| | - Graham J. Moore
- Pepmetics Incorporated, 772 Murphy Place, Victoria, BC V8Y 3H4, Canada;
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - John M. Matsoukas
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- NewDrug PC, Patras Science Park, 26500 Patras, Greece;
- Department of Chemistry, University of Patras, 26504 Patras, Greece
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, VIC 3030, Australia; (J.S.); (L.K.G.); (V.A.)
| |
Collapse
|
16
|
Zhu C, Gu W, Sun D, Wei W. The mechanism underlying fluoride-induced low-renin hypertension is related to an imbalance in the circulatory and local renin-angiotensin systems. Toxicol Lett 2023; 381:36-47. [PMID: 37105417 DOI: 10.1016/j.toxlet.2023.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/13/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
The renin-angiotensin system (RAS) is an important fluid regulation system in the body, and excessive activation of the circulatory or local RAS can increase blood pressure (BP). Excess fluoride can increase BP, although the underlying mechanism related to activation of the RAS remains unclear. Thus, the aim of this study was to elucidate the role of the RAS in fluoride-induced hypertension. Markers of the circulating and local RASs related to pathological changes to the kidneys, myocardium, and aorta were measured. Fluoride reduced serum levels of renin, angiotensin II (Ang II), and angiotensin (1-7) [Ang (1-7)], and dysregulated plasma levels of aldosterone and potassium levels. Excess fluoride can damage the kidneys, myocardium, and aorta, overactivate the renal angiotensin converting enzyme (ACE)-Ang II-angiotensin type 1 receptor axis, and inhibit activation of the ACE2-Ang (1-7)-Mas axis, leading to dysregulation of alpha epithelial sodium channels and significantly increased expression of Ang II in the myocardium and aorta. Hence, excess fluoride can cause low-renin hypertension via an imbalance between the circulatory and local RASs.
Collapse
Affiliation(s)
- Chenpeng Zhu
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, Heilongjiang, 150081, China
| | - Weikuan Gu
- Department of Orthopedic Surgery and BME-Campbell Clinic, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Dianjun Sun
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, Heilongjiang, 150081, China.
| | - Wei Wei
- Institute for Endemic Fluorosis Control, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, 150081, China; Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618504), Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
17
|
Shan M, Li S, Zhang Y, Chen Y, Zhou Y, Shi L. Maternal exercise upregulates the DNA methylation of Agtr1a to enhance vascular function in offspring of hypertensive rats. Hypertens Res 2023; 46:654-666. [PMID: 36539461 DOI: 10.1038/s41440-022-01124-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/08/2022] [Accepted: 11/14/2022] [Indexed: 12/25/2022]
Abstract
The angiotensin II signaling system regulates vascular dysfunction and is involved in the programming of hypertension. Maternal exercise has been linked to both short-term and long-term benefits for the mother and fetus. However, the impacts of maternal exercise on the intravascular renin-angiotensin system (RAS) in hypertensive offspring remain unexamined. This study examined whether maternal exercise has an epigenetic effect in repressing angiotensin II type 1 receptor (AT1R) expression, which leads to favorable alterations in the mesenteric artery (MA) function of spontaneously hypertensive offspring. Spontaneously hypertensive rats (SHRs) and Wistar-Kyoto (WKY) pregnant rats were randomly divided into an exercise group and a control group. Blood pressure, vascular tone, AT1R protein and mRNA expression, and AT1R gene (Agtr1a) promoter methylation status were examined in the MAs of 3-month-old male offspring. Maternal exercise significantly reduced the resting blood pressure and cardiovascular reactivity of offspring from SHRs. Furthermore, Ang II-AT1R activity in regulating vascular tone and AT1R expression was decreased in the MAs of the SHR offspring from the exercise groups. Importantly, exercise during gestation suppressed AT1R expression via hypermethylation of the Agtr1a promoter region and upregulated DNA methyltransferase (DNMT) expression in MAs of SHR offspring. These results suggest that maternal exercise upregulates DNMT expression, resulting in hypermethylation and repression of the Agtr1a gene, which may prevent MA dysfunction in the offspring of SHRs. A mechanistic model on the epigenetics of exercise during pregnancy. Maternal exercise during pregnancy triggers hypermethylation and transcriptional suppression of the Agtr1a gene via increased DNMT1 and DNMT3B expression in MAs of SHR offspring. Downregulation of AT1R expression reduces the contribution of Ang II to vascular tone, ultimately improving vascular structure and function. VSMC vascular smooth muscle cell; Ang II angiotensin II; AT1aR angiotensin type 1 receptor (AT1R) alpha subtypes; Agtr1a AT1R alpha isoform gene; MAs mesenteric arteries; BP blood pressure.
Collapse
Affiliation(s)
- Meiling Shan
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Shanshan Li
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Yanyan Zhang
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China.,Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, 100084, China
| | - Yu Chen
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Yang Zhou
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Lijun Shi
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China. .,Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
18
|
Li R, Li L, Zhang J, Wang D, Cui X, Bai L, Zhao L, Yang X. Alleviation of renal injury in rabbits by allisartan. J Investig Med 2023; 71:92-100. [PMID: 36733996 DOI: 10.1177/10815589221144850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The objective of this study was to determine the relationship between renal injury and inflammatory response induced by high-fat diet in rabbits and the interventional effect of allisartan. Fifteen 6-week-old healthy male rabbits were randomly divided into three groups: normal control (NC) group, high-lipid diet (HLD) group, high-lipid diet and allisartan (HLD+ALST) group. After allisartan treatment for 12 weeks, changes in total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), serum creatinine (Scr) and blood urea nitrogen (BUN) were measured enzymatically in the three groups. The left side of the kidney tissue was kept for paraffin section, and HE staining, periodic acid-Schiff (PAS) staining and Masson staining were used to observe the renal pathologic changes. TC, TG, LDL-C, Scr and BUN levels were all higher and HDL-C levels were lower in the HLD group compared with the NC group. Compared with the HLD group, Scr and BUN levels were significantly decreased in the HLD+ALST group. The results of HE staining showed that allisartan improved the changes of renal tissue morphology in rabbits on high-fat diet, reduced glomerular mesangial cell proliferation and improved glomerulosclerosis; PAS staining showed that glomerular glycogen deposition was reduced and glomerular red staining was significantly lighter; Masson staining showed that renal tubular blue-stained collagen fibers were reduced. In conclusion, hyperlipidemia can lead to aberrant expression of multiple cellular proteins and kidney tissue morphological damage in rabbits. On the other hand, allisartan attenuated renal injury and the mechanism may be related to the downregulation of the inflammatory response.
Collapse
Affiliation(s)
- Ruibin Li
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li Li
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jidong Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dong Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoran Cui
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Long Bai
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lei Zhao
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaohong Yang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Berton AM, Parasiliti-Caprino M, Prencipe N, Bioletto F, Lopez C, Bona C, Caputo M, Rumbolo F, Ponzetto F, Settanni F, Gasco V, Mengozzi G, Ghigo E, Grottoli S, Maccario M, Benso AS. Copeptin adaptive response to SGLT2 inhibitors in patients with type 2 diabetes mellitus: The GliRACo study. Front Neurosci 2023; 17:1098404. [PMID: 37021137 PMCID: PMC10067557 DOI: 10.3389/fnins.2023.1098404] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/03/2023] [Indexed: 04/07/2023] Open
Abstract
Introduction In type 2 diabetes mellitus (T2DM), the antidiuretic system participates in the adaptation to osmotic diuresis further increasing urinary osmolality by reducing the electrolyte-free water clearance. Sodium glucose co-transporter type 2 inhibitors (SGLT2i) emphasize this mechanism, promoting persistent glycosuria and natriuresis, but also induce a greater reduction of interstitial fluids than traditional diuretics. The preservation of osmotic homeostasis is the main task of the antidiuretic system and, in turn, intracellular dehydration the main drive to vasopressin (AVP) secretion. Copeptin is a stable fragment of the AVP precursor co-secreted with AVP in an equimolar amount. Aim To investigate the copeptin adaptive response to SGLT2i, as well as the induced changes in body fluid distribution in T2DM patients. Methods The GliRACo study was a prospective, multicenter, observational research. Twenty-six consecutive adult patients with T2DM were recruited and randomly assigned to empagliflozin or dapagliflozin treatment. Copeptin, plasma renin activity, aldosterone and natriuretic peptides were evaluated at baseline (T0) and then 30 (T30) and 90 days (T90) after SGLT2i starting. Bioelectrical impedance vector analysis (BIVA) and ambulatory blood pressure monitoring were performed at T0 and T90. Results Among endocrine biomarkers, only copeptin increased at T30, showing subsequent stability (7.5 pmol/L at T0, 9.8 pmol/L at T30, 9.5 pmol/L at T90; p = 0.001). BIVA recorded an overall tendency to dehydration at T90 with a stable proportion between extra- and intracellular fluid volumes. Twelve patients (46.1%) had a BIVA overhydration pattern at baseline and 7 of them (58.3%) resolved this condition at T90. Total body water content, extra and intracellular fluid changes were significantly affected by the underlying overhydration condition (p < 0.001), while copeptin did not. Conclusion In patients with T2DM, SGLT2i promote the release of AVP, thus compensating for persistent osmotic diuresis. This mainly occurs because of a proportional dehydration process between intra and extracellular fluid (i.e., intracellular dehydration rather than extracellular dehydration). The extent of fluid reduction, but not the copeptin response, is affected by the patient's baseline volume conditions. Clinical trial registration Clinicaltrials.gov, identifier NCT03917758.
Collapse
Affiliation(s)
- Alessandro Maria Berton
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
- *Correspondence: Alessandro Maria Berton, ; orcid.org/0000-0002-4745-2624
| | - Mirko Parasiliti-Caprino
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
- Mirko Parasiliti-Caprino, ; orcid.org/0000-0002-6930-7073
| | - Nunzia Prencipe
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Fabio Bioletto
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Chiara Lopez
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Chiara Bona
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Marina Caputo
- Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Francesca Rumbolo
- Clinical Biochemistry Laboratory, Department of Laboratory Medicine, AOU Città della Salute e della Scienza di Torino, University Hospital, Turin, Italy
| | - Federico Ponzetto
- Clinical Biochemistry Laboratory, Department of Laboratory Medicine, AOU Città della Salute e della Scienza di Torino, University Hospital, Turin, Italy
| | - Fabio Settanni
- Clinical Biochemistry Laboratory, Department of Laboratory Medicine, AOU Città della Salute e della Scienza di Torino, University Hospital, Turin, Italy
| | - Valentina Gasco
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giulio Mengozzi
- Clinical Biochemistry Laboratory, Department of Laboratory Medicine, AOU Città della Salute e della Scienza di Torino, University Hospital, Turin, Italy
| | - Ezio Ghigo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Grottoli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Mauro Maccario
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Andrea Silvio Benso
- Division of Endocrinology, Diabetes and Metabolism, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Fargieva KR, Guseinova RM, Pigarova EA, Dzeranova LK. The role of the apelin/APJ system in water homeostasis regulation. OBESITY AND METABOLISM 2022. [DOI: 10.14341/omet12752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Water balance in the body is achieved by balancing renal and non-renal water losses with corresponding water intake. It is under the control of both the central nervous system, which integrates many parameters of water and electrolyte balance in the body, including inducing important adaptive behavioral responses, and three hormonal systems: vasopressinergic, renin-angiotensin-aldosterone and apelinergic. A lot of research is devoted to the regulation of water-electrolyte metabolism. However, this process is still quite difficult to understand, especially since more and more of its regulators are being discovered over time. One of them is the hormone apelin, an endogenous ligand for the APJ receptor. As is known, the receptor is highly expressed in many organs, such as the brain, heart, liver and kidneys, lungs, and has multidirectional effects.This literature review discusses the main characteristics and features of the regulation of these systems in relation to water-electrolyte metabolism, as well as issues of intersystem interaction and modulation of the effects of apelin.
Collapse
|
21
|
Castro LUC, Otsuki DA, Sanches TR, Souza FL, Santinho MAR, da Silva C, Noronha IDL, Duarte-Neto AN, Gomes SA, Malbouisson LMS, Andrade L. Terlipressin combined with conservative fluid management attenuates hemorrhagic shock-induced acute kidney injury in rats. Sci Rep 2022; 12:20443. [PMID: 36443404 PMCID: PMC9705717 DOI: 10.1038/s41598-022-24982-0] [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: 12/14/2021] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Hemorrhagic shock (HS), a major cause of trauma-related mortality, is mainly treated by crystalloid fluid administration, typically with lactated Ringer's (LR). Despite beneficial hemodynamic effects, such as the restoration of mean arterial pressure (MAP), LR administration has major side effects, including organ damage due to edema. One strategy to avoid such effects is pre-hospitalization intravenous administration of the potent vasoconstrictor terlipressin, which can restore hemodynamic stability/homeostasis and has anti-inflammatory effects. Wistar rats were subjected to HS for 60 min, at a target MAP of 30-40 mmHg, thereafter being allocated to receive LR infusion at 3 times the volume of the blood withdrawn (liberal fluid management); at 2 times the volume (conservative fluid management), plus terlipressin (10 µg/100 g body weight); and at an equal volume (conservative fluid management), plus terlipressin (10 µg/100 g body weight). A control group comprised rats not subjected to HS and receiving no fluid resuscitation or treatment. At 15 min after fluid resuscitation/treatment, the blood previously withdrawn was reinfused. At 24 h after HS, MAP was higher among the terlipressin-treated animals. Terlipressin also improved post-HS survival and provided significant improvements in glomerular/tubular function (creatinine clearance), neutrophil gelatinase-associated lipocalin expression, fractional excretion of sodium, aquaporin 2 expression, tubular injury, macrophage infiltration, interleukin 6 levels, interleukin 18 levels, and nuclear factor kappa B expression. In terlipressin-treated animals, there was also significantly higher angiotensin II type 1 receptor expression and normalization of arginine vasopressin 1a receptor expression. Terlipressin associated with conservative fluid management could be a viable therapy for HS-induced acute kidney injury, likely attenuating such injury by modulating the inflammatory response via the arginine vasopressin 1a receptor.
Collapse
Affiliation(s)
- Leticia Urbano Cardoso Castro
- grid.11899.380000 0004 1937 0722Laboratory of Basic Science in Renal Diseases, Division of Nephrology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º Andar, sala 3310, São Paulo, SP CEP 01246-903 Brazil
| | - Denise Aya Otsuki
- grid.11899.380000 0004 1937 0722Laboratory of Anesthesiology, Division of Anesthesiology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Talita Rojas Sanches
- grid.11899.380000 0004 1937 0722Laboratory of Basic Science in Renal Diseases, Division of Nephrology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º Andar, sala 3310, São Paulo, SP CEP 01246-903 Brazil
| | - Felipe Lima Souza
- grid.11899.380000 0004 1937 0722Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Mirela Aparecida Rodrigues Santinho
- grid.11899.380000 0004 1937 0722Laboratory of Basic Science in Renal Diseases, Division of Nephrology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º Andar, sala 3310, São Paulo, SP CEP 01246-903 Brazil
| | - Cleonice da Silva
- grid.11899.380000 0004 1937 0722Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Irene de Lourdes Noronha
- grid.11899.380000 0004 1937 0722Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Amaro Nunes Duarte-Neto
- grid.11899.380000 0004 1937 0722Department of Pathology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Samirah Abreu Gomes
- grid.11899.380000 0004 1937 0722Laboratory of Cellular, Genetic, and Molecular Nephrology, Renal Division, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Luiz-Marcelo Sá Malbouisson
- grid.11899.380000 0004 1937 0722Laboratory of Anesthesiology, Division of Anesthesiology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Lucia Andrade
- grid.11899.380000 0004 1937 0722Laboratory of Basic Science in Renal Diseases, Division of Nephrology, University of São Paulo School of Medicine, Av. Dr. Arnaldo, 455, 3º Andar, sala 3310, São Paulo, SP CEP 01246-903 Brazil
| |
Collapse
|
22
|
The Heart as a Target of Vasopressin and Other Cardiovascular Peptides in Health and Cardiovascular Diseases. Int J Mol Sci 2022; 23:ijms232214414. [PMID: 36430892 PMCID: PMC9699305 DOI: 10.3390/ijms232214414] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
The automatism of cardiac pacemaker cells, which is tuned, is regulated by the autonomic nervous system (ANS) and multiple endocrine and paracrine factors, including cardiovascular peptides. The cardiovascular peptides (CPs) form a group of essential paracrine factors affecting the function of the heart and vessels. They may also be produced in other organs and penetrate to the heart via systemic circulation. The present review draws attention to the role of vasopressin (AVP) and some other cardiovascular peptides (angiotensins, oxytocin, cytokines) in the regulation of the cardiovascular system in health and cardiovascular diseases, especially in post-infarct heart failure, hypertension and cerebrovascular strokes. Vasopressin is synthesized mostly by the neuroendocrine cells of the hypothalamus. There is also evidence that it may be produced in the heart and lungs. The secretion of AVP and other CPs is markedly influenced by changes in blood volume and pressure, as well as by other disturbances, frequently occurring in cardiovascular diseases (hypoxia, pain, stress, inflammation). Myocardial infarction, hypertension and cardiovascular shock are associated with an increased secretion of AVP and altered responsiveness of the cardiovascular system to its action. The majority of experimental studies show that the administration of vasopressin during ventricular fibrillation and cardiac arrest improves resuscitation, however, the clinical studies do not present consisting results. Vasopressin cooperates with the autonomic nervous system (ANS), angiotensins, oxytocin and cytokines in the regulation of the cardiovascular system and its interaction with these regulators is altered during heart failure and hypertension. It is likely that the differences in interactions of AVP with ANS and other CPs have a significant impact on the responsiveness of the cardiovascular system to vasopressin in specific cardiovascular disorders.
Collapse
|
23
|
Zuo X, Ren S, Zhang H, Tian J, Tian R, Han B, Liu H, Dong Q, Wang Z, Cui Y, Niu R, Zhang F. Chemotherapy induces ACE2 expression in breast cancer via the ROS-AKT-HIF-1α signaling pathway: a potential prognostic marker for breast cancer patients receiving chemotherapy. J Transl Med 2022; 20:509. [PMID: 36335375 PMCID: PMC9636712 DOI: 10.1186/s12967-022-03716-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Background Angiotensin-converting enzyme 2 (ACE2) is a key enzyme of the renin-angiotensin system and a well-known functional receptor for the entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into host cells. The COVID-19 pandemic has brought ACE2 into the spotlight, and ACE2 expression in tumors and its relationship with SARS-COV-2 infection and prognosis of cancer patients have received extensive attention. However, the association between ACE2 expression and tumor therapy and prognosis, especially in breast cancer, remains ambiguous and requires further investigation. We have previously reported that ACE2 is elevated in drug-resistant breast cancer cells, but the exact function of ACE2 in drug resistance and progression of this malignant disease has not been explored. Methods The expression of ACE2 and HIF-1α in parental and drug-resistant breast cancer cells under normoxic and hypoxic conditions was analyzed by Western blot and qRT-PCR methods. The protein levels of ACE2 in plasma samples from breast cancer patients were examined by ELISA. The relationship between ACE2 expression and breast cancer treatment and prognosis was analyzed using clinical specimens and public databases. The reactive oxygen species (ROS) levels in breast cancer cells were measured by using a fluorescent probe. Small interfering RNAs (siRNAs) or lentivirus-mediated shRNA was used to silence ACE2 and HIF-1α expression in cellular models. The effect of ACE2 knockdown on drug resistance in breast cancer was determined by Cell Counting Kit 8 (CCK-8)-based assay, colony formation assay, apoptosis and EdU assay. Results ACE2 expression is relatively low in breast cancer cells, but increases rapidly and specifically after exposure to anticancer drugs, and remains high after resistance is acquired. Mechanistically, chemotherapeutic agents increase ACE2 expression in breast cancer cells by inducing intracellular ROS production, and increased ROS levels enhance AKT phosphorylation and subsequently increase HIF-1α expression, which in turn upregulates ACE2 expression. Although ACE2 levels in plasma and cancer tissues are lower in breast cancer patients compared with healthy controls, elevated ACE2 in patients after chemotherapy is a predictor of poor treatment response and an unfavorable prognostic factor for survival in breast cancer patients. Conclusion ACE2 is a gene in breast cancer cells that responds rapidly to chemotherapeutic agents through the ROS-AKT-HIF-1α axis. Elevated ACE2 modulates the sensitivity of breast cancer cells to anticancer drugs by optimizing the balance of intracellular ROS. Moreover, increased ACE2 is not only a predictor of poor response to chemotherapy, but is also associated with a worse prognosis in breast cancer patients. Thus, our findings provide novel insights into the spatiotemporal differences in the function of ACE2 in the initiation and progression of breast cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03716-w.
Collapse
|
24
|
Szczepańska-Sadowska E, Żera T. Vasopressin: a possible link between hypoxia and hypertension. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Cardiovascular and respiratory diseases are frequently associated with transient and prolonged hypoxia, whereas hypoxia exerts pro-hypertensive effects, through stimulation of the sympathetic system and release of pressor endocrine factors. This review is focused on the role of arginine vasopressin (AVP) in dysregulation of the cardiovascular system during hypoxia associated with cardiovascular disorders. AVP is synthesized mainly in the neuroendocrine neurons of the hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON), which send axons to the posterior pituitary and various regions of the central nervous system (CNS). Vasopressinergic neurons are innervated by multiple neuronal projections releasing several neurotransmitters and other regulatory molecules. AVP interacts with V1a, V1b and V2 receptors that are present in the brain and peripheral organs, including the heart, vessels, lungs, and kidneys. Release of vasopressin is intensified during hypernatremia, hypovolemia, inflammation, stress, pain, and hypoxia which frequently occur in cardiovascular patients, and blood AVP concentration is markedly elevated in cardiovascular diseases associated with hypoxemia. There is evidence that hypoxia stimulates AVP release through stimulation of chemoreceptors. It is suggested that acting in the carotid bodies, AVP may fine-tune respiratory and hemodynamic responses to hypoxia and that this effect is intensified in hypertension. There is also evidence that during hypoxia, augmentation of pro-hypertensive effects of vasopressin may result from inappropriate interaction of this hormone with other compounds regulating the cardiovascular system (catecholamines, angiotensins, natriuretic peptides, steroids, nitric oxide). In conclusion, current literature indicates that abnormal mutual interactions between hypoxia and vasopressin may significantly contribute to pathogenesis of hypertension.
Collapse
Affiliation(s)
- Ewa Szczepańska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Tymoteusz Żera
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
25
|
Dmitrieva NI, Liu D, Wu CO, Boehm M. Middle age serum sodium levels in the upper part of normal range and risk of heart failure. Eur Heart J 2022; 43:3335-3348. [PMID: 35348651 PMCID: PMC10263272 DOI: 10.1093/eurheartj/ehac138] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/01/2022] [Accepted: 03/03/2022] [Indexed: 07/29/2023] Open
Abstract
AIMS With increasing prevalence of heart failure (HF) owing to the ageing population, identification of modifiable risk factors is important. In a mouse model, chronic hypohydration induced by lifelong water restriction promotes cardiac fibrosis. Hypohydration elevates serum sodium. Here, we evaluate the association of serum sodium at middle age as a measure of hydration habits with risk to develop HF. METHODS AND RESULTS We analysed data from Atherosclerosis Risk in Communities study with middle age enrolment (45-66 years) and 25 years of follow-up. Participants without water balance dysregulation were selected: serum sodium within normal range (135-146 mmol/L), not diabetic, not obese and free of HF at baseline (N = 11 814). In time-to-event analysis, HF risk was increased by 39% if middle age serum sodium exceeded 143 mmol/L corresponding to 1% body weight water deficit [hazard ratio 1.39, 95% confidence interval (CI) 1.14-1.70]. In a retrospective case-control analysis performed on 70- to 90-year-old attendees of Visit 5 (N = 4961), serum sodium of 142.5-143 mmol/L was associated with 62% increase in odds of left ventricular hypertrophy (LVH) diagnosis [odds ratio (OR) 1.62, 95% CI 1.03-2.55]. Serum sodium above 143 mmol/L was associated with 107% increase in odds of LVH (OR 2.07, 95% CI 1.30-3.28) and 54% increase in odds of HF (OR 1.54, 95% CI 1.06-2.23). As a result, prevalence of HF and LVH was increased among 70- to 90-year-old participants with higher middle age serum sodium. CONCLUSION Middle age serum sodium above 142 mmol is a risk factor for LVH and HF. Maintaining good hydration throughout life may slow down decline in cardiac function and decrease prevalence of HF.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- The Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Delong Liu
- The Laboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Colin O Wu
- Office of Biostatistics Research, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| | - Manfred Boehm
- The Laboratory of Cardiovascular Regenerative Medicine, National Heart Lung and Blood Institute, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Yang XF, Wang H, Huang Y, Huang JH, Ren HL, Xu Q, Su XM, Wang AM, Ren F, Zhou MS. Myeloid Angiotensin II Type 1 Receptor Mediates Macrophage Polarization and Promotes Vascular Injury in DOCA/Salt Hypertensive Mice. Front Pharmacol 2022; 13:879693. [PMID: 35721173 PMCID: PMC9204513 DOI: 10.3389/fphar.2022.879693] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022] Open
Abstract
Activation of the renin–angiotensin system has been implicated in hypertension. Angiotensin (Ang) II is a potent proinflammatory mediator. The present study investigated the role of myeloid angiotensin type 1 receptor (AT1R) in control of macrophage phenotype in vitro and vascular injury in deoxycorticosterone acetate (DOCA)/salt hypertension. In human THP-1/macrophages, Ang II increased mRNA expressions of M1 cytokines and decreased M2 cytokine expressions. Overexpression of AT1R further increased Ang II-induced expressions of M1 cytokines and decreased M2 cytokines. Silenced AT1R reversed Ang II-induced changes in M1 and M2 cytokines. Ang II upregulated hypoxia-inducible factor (HIF)1α, toll-like receptor (TLR)4, and the ratio of pIκB/IκB, which were prevented by silenced AT1R. Silenced HIF1α prevented Ang II activation of the TLR4/NFκB pathway. Furthermore, Ang II increased HIF1α via reactive oxygen species-dependent reduction in prolyl hydroxylase domain protein 2 (PHD2) expression. The expressions of AT1R and HIF1α and the ratio of pIκB/IκB were upregulated in the peritoneal macrophages of DOCA hypertensive mice, and the specific deletion of myeloid AT1R attenuated cardiac and vascular injury and vascular oxidative stress, reduced the recruitment of macrophages and M1 cytokine expressions, and improved endothelial function without significant reduction in blood pressure. Our results demonstrate that Ang II/AT1R controls the macrophage phenotype via stimulating the HIF1α/NFκB pathway, and specific myeloid AT1R KO improves endothelial function, vascular inflammation, and injury in salt-sensitive hypertension. The results support the notion that myeloid AT1R plays an important role in the regulation of the macrophage phenotype, and dysfunction of this receptor may promote vascular dysfunction and injury in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Xue-Feng Yang
- Department of Physiology, Jinzhou Medical University, Jinzhou, China
| | - Huan Wang
- Department of Physiology, Shenyang Medical College, Shenyang, China
| | - Yue Huang
- The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, China
| | - Jian-Hua Huang
- The First Affiliated Hospital, Jinzhou Medical University, Jinzhou, China
| | - Hao-Lin Ren
- Radiology Department of the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Qian Xu
- Department of Physiology, Shenyang Medical College, Shenyang, China
| | - Xiao-Min Su
- Department of Physiology, Shenyang Medical College, Shenyang, China
| | - Ai-Mei Wang
- Department of Physiology, Jinzhou Medical University, Jinzhou, China
| | - Fu Ren
- Department of Anatomy, Shenyang Medical College, Shenyang, China
- *Correspondence: Ming-Sheng Zhou, ; Fu Ren,
| | - Ming-Sheng Zhou
- Department of Physiology, Shenyang Medical College, Shenyang, China
- *Correspondence: Ming-Sheng Zhou, ; Fu Ren,
| |
Collapse
|
27
|
Alsalemi N, Sadowski CA, Elftouh N, Louis M, Kilpatrick K, Houle SKD, Lafrance JP. The effect of renin-angiotensin-aldosterone system inhibitors on continuous and binary kidney outcomes in subgroups of patients with diabetes: a meta-analysis of randomized clinical trials. BMC Nephrol 2022; 23:161. [PMID: 35484505 PMCID: PMC9052620 DOI: 10.1186/s12882-022-02763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 03/29/2022] [Indexed: 12/02/2022] Open
Abstract
INTRODUCTION Diabetic nephropathy is the leading cause of kidney failure. Clinical practice guidelines recommend prescribing renin-angiotensin aldosterone system inhibitors (RAASi) to prevent diabetic nephropathy at any stage. We conducted this systematic review and meta-analysis to compare the effects of RAASi with placebo and other antihypertensive agents in adults with diabetes on continuous and binary kidney outcomes to provide a comprehensive review of the class effect of RAASi on several subgroups. METHODS A systematic electronic search to identify randomized clinical trials of a duration of ≥ 12 months that recruited ≥ 50 adult participants with type 1 or 2 diabetes with any stage of chronic kidney disease and proteinuria was conducted in MEDLINE, CINAHL, EMBASE, and Cochrane library with no language restriction. Studies were screened against the inclusion and exclusion criteria by two reviewers independently. RESULTS In this meta-analysis, evidence was drawn from 26,551 patients with diabetes from 46 studies. Our analysis shows that RAASi were better than placebo in reducing SrCr (the raw mean difference [RMD] = -13.4 μmol/L; 95%CI: -16.78; -10.01) and albuminuria levels (standardized mean difference [SMD] = -1; 95%CI: -1.57, -0.44, I2 = 96%). When compared to other active treatments, RAASi did not reduce SrCr (RMD = 0.03 μmol/L; 95%CI: -6.4, 6.10, I2 = 76%), caused a non-significant reduction of GFR levels (RMD = -1.21 mL/min; 95%CI: -4.52, 2.09, I2 = 86%), and resulted in modest reduction of albuminuria levels (SMD = -0.55; 95%CI: -0.95, -0.16, I2 = 90%). RAASi were superior to placebo in reducing the risks of kidney failure (OR = 0.74; 95%CI: 0.56, 0.97) and doubling of serum creatinine levels (SrCr; OR = 0.71; 95%CI: 0.55, 0.91), but not in promoting the regression of albuminuria (OR = 3.00; 95%CI: 0.96, 9.37). RAASi, however, were not superior to other antihypertensives in reducing the risks of these outcomes. Patients with type 2 diabetes, macroalbuminuria and longer duration of diabetes had less risk of developing kidney failure in placebo-controlled trials, while longer duration of diabetes, normal kidney function, and hypertension increased the probability of achieving regression of albuminuria in active-controlled trials. CONCLUSION While our findings revealed the non-superiority of RAASi over other antihypertensives and portrayed a class effect on several subgroups of study participants, it raised a challenging question on whether RAASi deserve their place as first-line therapy in managing diabetic nephropathy.
Collapse
Affiliation(s)
- Noor Alsalemi
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, Canada
- Centre de Recherche de L'Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Cheryl A Sadowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Naoual Elftouh
- Centre de Recherche de L'Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Maudeline Louis
- Centre de Recherche de L'Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Kelley Kilpatrick
- Centre de Recherche de L'Hôpital Maisonneuve-Rosemont, Montreal, Canada
- Ingram School of Nursing, McGill University, Montreal, Canada
| | | | - Jean-Philippe Lafrance
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, Canada.
- Centre de Recherche de L'Hôpital Maisonneuve-Rosemont, Montreal, Canada.
- Service de Néphrologie, CIUSSS de L'Est-de-L'Île-de-Montréal, Montreal, Canada.
| |
Collapse
|
28
|
Hasan HF, Galal SM, Ellethy RA. Mitigative impact of bradykinin potentiating factor isolated from Androctonus amorexi scorpion venom and low doses of γ-irradiation on doxorubicin induced hepatotoxicity through Ang II/AMPK crosstalk. Toxicol Mech Methods 2022; 32:518-529. [DOI: 10.1080/15376516.2022.2049941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Hesham Farouk Hasan
- Radiation Biology Department, National Center for Radiation Research and Technology (NCRRT), Atomic Energy Authority, Cairo, Egypt
| | - Shereen Mohamed Galal
- Health Radiation Research Department, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Rania A Ellethy
- Chemistry department, faculty of science, Helwan university, Cairo, Egypt
| |
Collapse
|
29
|
Szczepanska-Sadowska E, Wsol A, Cudnoch-Jedrzejewska A, Czarzasta K, Żera T. Multiple Aspects of Inappropriate Action of Renin-Angiotensin, Vasopressin, and Oxytocin Systems in Neuropsychiatric and Neurodegenerative Diseases. J Clin Med 2022; 11:908. [PMID: 35207180 PMCID: PMC8877782 DOI: 10.3390/jcm11040908] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
The cardiovascular system and the central nervous system (CNS) closely cooperate in the regulation of primary vital functions. The autonomic nervous system and several compounds known as cardiovascular factors, especially those targeting the renin-angiotensin system (RAS), the vasopressin system (VPS), and the oxytocin system (OTS), are also efficient modulators of several other processes in the CNS. The components of the RAS, VPS, and OTS, regulating pain, emotions, learning, memory, and other cognitive processes, are present in the neurons, glial cells, and blood vessels of the CNS. Increasing evidence shows that the combined function of the RAS, VPS, and OTS is altered in neuropsychiatric/neurodegenerative diseases, and in particular in patients with depression, Alzheimer's disease, Parkinson's disease, autism, and schizophrenia. The altered function of the RAS may also contribute to CNS disorders in COVID-19. In this review, we present evidence that there are multiple causes for altered combined function of the RAS, VPS, and OTS in psychiatric and neurodegenerative disorders, such as genetic predispositions and the engagement of the RAS, VAS, and OTS in the processes underlying emotions, memory, and cognition. The neuroactive pharmaceuticals interfering with the synthesis or the action of angiotensins, vasopressin, and oxytocin can improve or worsen the effectiveness of treatment for neuropsychiatric/neurodegenerative diseases. Better knowledge of the multiple actions of the RAS, VPS, and OTS may facilitate programming the most efficient treatment for patients suffering from the comorbidity of neuropsychiatric/neurodegenerative and cardiovascular diseases.
Collapse
Affiliation(s)
- Ewa Szczepanska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland; (A.W.); (A.C.-J.); (K.C.); (T.Ż.)
| | | | | | | | | |
Collapse
|
30
|
Nasiri Boroujeni S, Chehelcheraghi F, Khaksarian M, Sedighi M, Ghorbanzadeh V, Nazari A. Applying Vasopressin-Pre-Conditioned Human Adipose Mesenchymal Stem Cells Improves Heart Condition after Transplantation into Infarcted Myocardium. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2022; 11:207-222. [PMID: 37605740 PMCID: PMC10440004 DOI: 10.22088/ijmcm.bums.11.3.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 04/03/2023] [Accepted: 04/24/2023] [Indexed: 08/23/2023]
Abstract
Transplantation of H-AdMSCs may improve heart function after MI. AVP is a neurohypophyseal hormone that reduces cardiovascular damage. This study investigated the role of AVP preconditioning in the survival of MSCs and their effect on myocardial repair in the MI rats. H-AMSCs were isolated and incubated for 3 days. The expression of oxytocin and vasopressin receptors was evaluated by Real-time-PCR. Forty male Wistar rats were divided into 4 groups: control, sham, ASC and AVP-ASC. Ischemia was established by ligation of LAD coronary artery. Electrocardiography, fibrosis, angiogenesis, and apoptosis in myocardium were determined after 7 days. Results showed that preconditioned MSCs significantly increased cardiac function when compared with group that received non-preconditioned MSCs. This was associated with significantly reduced fibrosis, increased vascular density, and decreased resident myocyte apoptosis. Results indicate that AVP preconditioned MSCs can be consider a novel approach to management of MI.
Collapse
Affiliation(s)
| | - Farzaneh Chehelcheraghi
- Department of Anatomical Sciences, School of Medicine, Lorestan University of Medical Sciences, Khoramabad, Iran.
| | - Mojtaba Khaksarian
- Department of Physiology, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Mehrnoosh Sedighi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran.
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Vajihe Ghorbanzadeh
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran.
| | - Afshin Nazari
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran.
| |
Collapse
|
31
|
Bhullar S, Shah A, Dhalla N. Mechanisms for the development of heart failure and improvement of cardiac function by angiotensin-converting enzyme inhibitors. SCRIPTA MEDICA 2022. [DOI: 10.5937/scriptamed53-36256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Angiotensin-converting enzyme (ACE) inhibitors, which prevent the conversion of angiotensin I to angiotensin II, are well-known for the treatments of cardiovascular diseases, such as heart failure, hypertension and acute coronary syndrome. Several of these inhibitors including captopril, enalapril, ramipril, zofenopril and imidapril attenuate vasoconstriction, cardiac hypertrophy and adverse cardiac remodeling, improve clinical outcomes in patients with cardiac dysfunction and decrease mortality. Extensive experimental and clinical research over the past 35 years has revealed that the beneficial effects of ACE inhibitors in heart failure are associated with full or partial prevention of adverse cardiac remodeling. Since cardiac function is mainly determined by coordinated activities of different subcellular organelles, including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils, for regulating the intracellular concentration of Ca2+ and myocardial metabolism, there is ample evidence to suggest that adverse cardiac remodelling and cardiac dysfunction in the failing heart are the consequence of subcellular defects. In fact, the improvement of cardiac function by different ACE inhibitors has been demonstrated to be related to the attenuation of abnormalities in subcellular organelles for Ca2+-handling, metabolic alterations, signal transduction defects and gene expression changes in failing cardiomyocytes. Various ACE inhibitors have also been shown to delay the progression of heart failure by reducing the formation of angiotensin II, the development of oxidative stress, the level of inflammatory cytokines and the occurrence of subcellular defects. These observations support the view that ACE inhibitors improve cardiac function in the failing heart by multiple mechanisms including the reduction of oxidative stress, myocardial inflammation and Ca2+-handling abnormalities in cardiomyocytes.
Collapse
|
32
|
Heinrich NS, Theilade S, Winther SA, Tofte N, Ahluwalia TS, Jeppesen JL, Persson F, Hansen TW, Goetze JP, Rossing P. Copeptin and renal function decline, cardiovascular events and mortality in type 1 diabetes. Nephrol Dial Transplant 2021; 37:100-107. [PMID: 33367877 DOI: 10.1093/ndt/gfaa308] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Plasma copeptin is a surrogate of arginine vasopressin (AVP) secretion and is associated with a risk of renal and cardiovascular disease. We investigated associations between copeptin and renal events, cardiovascular events and mortality in type 1 diabetes (T1D). METHODS We conducted a prospective cohort study on 658 individuals with T1D from Steno Diabetes Center Copenhagen. Plasma copeptin concentrations and conventional risk factors were assessed at baseline. The five endpoints were traced through national registries and electronic laboratory records. RESULTS Baseline mean age was 55 ± 13 years and estimated glomerular filtration rate (eGFR) was 81 ± 26 mL/min/1.73 m2. The median follow-up was 6.2 years (interquartile range 5.8-6.7); 123 participants reached a combined renal endpoint [decline in eGFR ≥30%, end-stage kidney disease (ESKD) or all-cause mortality], 93 had a decrease in eGFR ≥30%, 21 developed ESKD, 94 experienced a combined cardiovascular endpoint and 58 died from all causes. Higher copeptin was associated with all endpoints in unadjusted Cox regression analyses. Upon adjustment for baseline eGFR, the associations were attenuated and remained significant only for the combined renal endpoint and decrease in eGFR ≥30%. Results were similar upon further adjustment for other risk factors, after which hazard ratios for the two renal endpoints were 2.27 (95% confidence interval 1.08-4.74) and 4.49 (1.77-11.4), respectively, for the highest versus the lowest quartile of copeptin. CONCLUSIONS Higher copeptin was an independent risk marker for a combined renal endpoint and decline in renal function. AVP may be a marker of renal damage or a factor whose contribution to renal and cardiovascular risk is partially mediated by renal damage.
Collapse
Affiliation(s)
| | - Simone Theilade
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Medicine, Herlev-Gentofte Hospital, Hellerup, Denmark
| | | | - Nete Tofte
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | | | - Jørgen L Jeppesen
- Department of Medicine, Amager Hvidovre Hospital, Glostrup, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Jens P Goetze
- Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Institute of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
33
|
Agbaraolorunpo F, Oloyo AK, Ogunnowo SA, Anigbogu CN, Sofola O. Effect of Angiotensin receptor blockade on Plasma Osmolality and Neurohumoral Responses to High Environmental Temperature in Rats Fed a High Salt Diet. Niger J Physiol Sci 2021; 36:149-157. [PMID: 35947735 DOI: 10.54548/njps.v36i2.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 06/15/2023]
Abstract
Plasma osmolality (pOsmol) and neurohumoral signals play important roles in the pathophysiology of cardiovascular diseases. Our study investigated the effect of high environmental temperature (HET) on neurohumoral responses and pOsmol in rats fed a high salt diet (HSD), with and without angiotensin II receptor blockade (ARB), using telmisartan. Fifty-six male 8-week old Sprague-Dawley rats (95-110g) were randomly assigned into seven groups of 8 rats. These included control rats (I) fed with 0.3% NaCl diet (normal diet, ND); salt-loaded rats (II) fed with 8% NaCl (high salt) diet; ND rats (III) exposed to HET (38.5±0.5oC ) 4 hours daily per week; rats (IV) fed with 8% NaCl diet and exposed to HET daily. Others included rats (V) fed with 8% NaCl diet and treated with telmisartan (30mg/kg); ND rats (VI) exposed to HET and treated with telmisartan; rats (VI) fed with 8% NaCl diet, exposed to HET and treated with telmisartan. Plasma angiotensin II, aldosterone, vasopressin and norepinephrine (NE) concentrations were determined by ELISA technique; pOsmol from plasma K+, Na+ and Urea. HSD combined with HET in rats synergistically increased pOsmol (P<0.001) with an associated non-synergistic rise in fluid intake (P<0.001), fluid balance (P<0.001), plasma angiotensin II (P<0.01) and aldosterone (P<0.05), NE (P<0.001) and vasopressin (P<0.05) concentrations compared to control. Telmisartan did not alter pOsmol in all the treated-rats, but normalized fluid intake levels and plasma vasopressin in the rats exposed to either HSD or HEt alone. Prolonged exposure of rats to hot environment exacerbated the effect of excess dietary salt on pOsmol, with no effect on angiotensin II-mediated neurohumoral responses.
Collapse
|
34
|
Proczka M, Przybylski J, Cudnoch-Jędrzejewska A, Szczepańska-Sadowska E, Żera T. Vasopressin and Breathing: Review of Evidence for Respiratory Effects of the Antidiuretic Hormone. Front Physiol 2021; 12:744177. [PMID: 34867449 PMCID: PMC8637824 DOI: 10.3389/fphys.2021.744177] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/27/2021] [Indexed: 12/17/2022] Open
Abstract
Vasopressin (AVP) is a key neurohormone involved in the regulation of body functions. Due to its urine-concentrating effect in the kidneys, it is often referred to as antidiuretic hormone. Besides its antidiuretic renal effects, AVP is a potent neurohormone involved in the regulation of arterial blood pressure, sympathetic activity, baroreflex sensitivity, glucose homeostasis, release of glucocorticoids and catecholamines, stress response, anxiety, memory, and behavior. Vasopressin is synthesized in the paraventricular (PVN) and supraoptic nuclei (SON) of the hypothalamus and released into the circulation from the posterior lobe of the pituitary gland together with a C-terminal fragment of pro-vasopressin, known as copeptin. Additionally, vasopressinergic neurons project from the hypothalamus to the brainstem nuclei. Increased release of AVP into the circulation and elevated levels of its surrogate marker copeptin are found in pulmonary diseases, arterial hypertension, heart failure, obstructive sleep apnoea, severe infections, COVID-19 due to SARS-CoV-2 infection, and brain injuries. All these conditions are usually accompanied by respiratory disturbances. The main stimuli that trigger AVP release include hyperosmolality, hypovolemia, hypotension, hypoxia, hypoglycemia, strenuous exercise, and angiotensin II (Ang II) and the same stimuli are known to affect pulmonary ventilation. In this light, we hypothesize that increased AVP release and changes in ventilation are not coincidental, but that the neurohormone contributes to the regulation of the respiratory system by fine-tuning of breathing in order to restore homeostasis. We discuss evidence in support of this presumption. Specifically, vasopressinergic neurons innervate the brainstem nuclei involved in the control of respiration. Moreover, vasopressin V1a receptors (V1aRs) are expressed on neurons in the respiratory centers of the brainstem, in the circumventricular organs (CVOs) that lack a blood-brain barrier, and on the chemosensitive type I cells in the carotid bodies. Finally, peripheral and central administrations of AVP or antagonists of V1aRs increase/decrease phrenic nerve activity and pulmonary ventilation in a site-specific manner. Altogether, the findings discussed in this review strongly argue for the hypothesis that vasopressin affects ventilation both as a blood-borne neurohormone and as a neurotransmitter within the central nervous system.
Collapse
Affiliation(s)
- Michał Proczka
- Department of Experimental and Clinical Physiology, Doctoral School, Medical University of Warsaw, Warsaw, Poland
| | - Jacek Przybylski
- Department of Biophysics, Physiology, and Pathophysiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Ewa Szczepańska-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Tymoteusz Żera
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
35
|
Soppert J, Frisch J, Wirth J, Hemmers C, Boor P, Kramann R, Vondenhoff S, Moellmann J, Lehrke M, Hohl M, van der Vorst EPC, Werner C, Speer T, Maack C, Marx N, Jankowski J, Roma LP, Noels H. A systematic review and meta-analysis of murine models of uremic cardiomyopathy. Kidney Int 2021; 101:256-273. [PMID: 34774555 DOI: 10.1016/j.kint.2021.10.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) triggers the risk of developing uremic cardiomyopathy as characterized by cardiac hypertrophy, fibrosis and functional impairment. Traditionally, animal studies are used to reveal the underlying pathological mechanism, although variable CKD models, mouse strains and readouts may reveal diverse results. Here, we systematically reviewed 88 studies and performed meta-analyses of 52 to support finding suitable animal models for future experimental studies on pathological kidney-heart crosstalk during uremic cardiomyopathy. We compared different mouse strains and the direct effect of CKD on cardiac hypertrophy, fibrosis and cardiac function in "single hit" strategies as well as cardiac effects of kidney injury combined with additional cardiovascular risk factors in "multifactorial hit" strategies. In C57BL/6 mice, CKD was associated with a mild increase in cardiac hypertrophy and fibrosis and marginal systolic dysfunction. Studies revealed high variability in results, especially regarding hypertrophy and systolic function. Cardiac hypertrophy in CKD was more consistently observed in 129/Sv mice, which express two instead of one renin gene and more consistently develop increased blood pressure upon CKD induction. Overall, "multifactorial hit" models more consistently induced cardiac hypertrophy and fibrosis compared to "single hit" kidney injury models. Thus, genetic factors and additional cardiovascular risk factors can "prime" for susceptibility to organ damage, with increased blood pressure, cardiac hypertrophy and early cardiac fibrosis more consistently observed in 129/Sv compared to C57BL/6 strains.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Janina Frisch
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Julia Wirth
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Hemmers
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany; Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Sonja Vondenhoff
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Mathias Hohl
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands; Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Werner
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Thimoteus Speer
- Translational Cardio-Renal Medicine, Saarland University, Homburg/Saar, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Würzburg, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
36
|
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: 6.0] [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
|
37
|
Bhullar SK, Shah AK, Dhalla NS. Role of angiotensin II in the development of subcellular remodeling
in heart failure. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00054] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The development of heart failure under various pathological conditions such as myocardial infarction (MI), hypertension and diabetes are accompanied by adverse cardiac remodeling and cardiac dysfunction. Since heart function is mainly determined by coordinated activities of different subcellular organelles including sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils for regulating the intracellular concentration of Ca2+, it has been suggested that the occurrence of heart failure is a consequence of subcellular remodeling, metabolic alterations and Ca2+-handling abnormalities in cardiomyocytes. Because of the elevated plasma levels of angiotensin II (ANG II) due to activation of the renin-angiotensin system (RAS) in heart failure, we have evaluated the effectiveness of treatments with angiotensin converting enzyme (ACE) inhibitors and ANG II type 1 receptor (AT1R) antagonists in different experimental models of heart failure. Attenuation of marked alterations in subcellular activities, protein content and gene expression were associated with improvement in cardiac function in MI-induced heart failure by treatment with enalapril (an ACE inhibitor) or losartan (an AT1R antagonist). Similar beneficial effects of ANG II blockade on subcellular remodeling and cardiac performance were also observed in failing hearts due to pressure overload, volume overload or chronic diabetes. Treatments with enalapril and losartan were seen to reduce the degree of RAS activation as well as the level of oxidative stress in failing hearts. These observations provide evidence which further substantiate to support the view that activation of RAS and high level of plasma ANG II play a critical role in inducing subcellular defects and cardiac dys-function during the progression of heart failure.
Collapse
Affiliation(s)
- Sukhwinder K. Bhullar
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada
| | - Anureet K. Shah
- School of Kinesiology, Nutrition and Food Science, California State University, Los Angeles, CA 90032, USA
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, University of Manitoba, Winnipeg, Manitoba R2H 2A6, Canada; Department of Physiology and Pathophysiology, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 3P5, Canada
| |
Collapse
|
38
|
Capalonga L, de Araujo CLP, Hentschke VS, Rossato DD, Quagliotto E, Becker T, Rigatto K, Ferraresi C, Parizotto NA, Dal Lago P. Neuromuscular electrical stimulation but not photobiomodulation therapy improves cardiovascular parameters of rats with heart failure. Can J Physiol Pharmacol 2021; 99:720-728. [PMID: 33211546 DOI: 10.1139/cjpp-2020-0339] [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] [Indexed: 11/22/2022]
Abstract
The aim of the present study was to analyze the effect of neuromuscular electrical stimulation (NMES) and photobiomodulation (PBMT) on the cardiovascular parameters, hemodynamic function, arterial baroreflex sensitivity (BRS), and autonomic balance (ANS) of rats with heart failure (HF). Male Wistar rats (220-290 g) were organized into five groups: Sham (n = 6), Control-HF (n = 5), NMES-HF (n = 6), PBMT-HF (n = 6), and NMES + PBMT-HF (n = 6). Myocardial infarction (MI) was induced by left coronary artery ligation. Animals were subjected to an eight-week NMES and PBMT protocol. Statistical analysis included the General Linear Model (GLM) followed by a Bonferroni post-hoc test. Rats of the NMES-HF group showed a higher MI area than the Control-HF (P = 0.003), PBMT-HF (P = 0.002), and NMES + PBMT-HF (P = 0.012) groups. NMES-HF and NMES + PBMT-HF showed higher pulmonary congestion (P = 0.004 and P = 0.02) and lower systolic pressure (P = 0.019 and P = 0.002) than the Sham group. NMES + PBMT-HF showed lower mean arterial pressure (P = 0.02) than the Sham group. Control-HF showed a higher heart rate than the NMES-HF and NMES + PBMT-HF (P = 0.017 and P = 0.013) groups. There was no difference in the BRS and ANS variables between groups. In conclusion, eight-week NMES isolated or associated with PBMT protocol reduced basal heart rate, systolic and mean arterial pressure, without influence on baroreflex sensibility and autonomic control, and no effect of PBMT was seen in rats with HF.
Collapse
Affiliation(s)
- Lucas Capalonga
- Laboratory of Experimental Physiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Cintia Laura Pereira de Araujo
- Laboratory of Experimental Physiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências da Reabilitação, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | | | | | - Edson Quagliotto
- Universidade de Santa Cruz do Sul (UNISC), Santa Cruz do Sul, RS, Brazil
| | - Tiago Becker
- Departamento de Engenharia Mecânica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Katya Rigatto
- Laboratório de Fisiologia Translacional, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Cleber Ferraresi
- Biomedical Engineering, Universidade do Brasil, São Paulo, Brazil
| | - Nivaldo Antonio Parizotto
- Biomedical Engineering, Universidade do Brasil, São Paulo, Brazil
- Biotechnology in Regenerative Medicine and Medical Chemistry, Universidade de Araraquara, Araraquara, Brazil
- Physical Therapy Department, Universidade Federal de São Carlos, São Carlos, Brazil
| | - Pedro Dal Lago
- Laboratory of Experimental Physiology, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
- Programa de Pós-Graduação em Ciências da Reabilitação, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| |
Collapse
|
39
|
Barsha G, Mirabito Colafella KM, Walton SL, Gaspari TA, Spizzo I, Pinar AA, Hilliard Krause LM, Widdop RE, Samuel CS, Denton KM. In Aged Females, the Enhanced Pressor Response to Angiotensin II Is Attenuated By Estrogen Replacement via an Angiotensin Type 2 Receptor-Mediated Mechanism. Hypertension 2021; 78:128-137. [PMID: 33966450 DOI: 10.1161/hypertensionaha.121.17164] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Giannie Barsha
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Katrina M Mirabito Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Sarah L Walton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Tracey A Gaspari
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Iresha Spizzo
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Anita A Pinar
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Lucinda M Hilliard Krause
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Pharmacology (T.A.G., I.S., A.A.P., R.E.W., C.S.S.), Monash University, Melbourne, Victoria, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute (G.B., K.M.M.C., S.L.W., T.A.G., I.S., A.A.P., L.M.H.K., R.E.W., C.S.S., K.M.D.), Monash University, Melbourne, Victoria, Australia.,Department of Physiology (G.B., KM.M.C., S.L.W., L.M.H.K., K.M.D.), Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Clarke GD, Li J, Kuo AH, Moody AJ, Nathanielsz PW. Cardiac magnetic resonance imaging: insights into developmental programming and its consequences for aging. J Dev Orig Health Dis 2021; 12:203-219. [PMID: 33349289 PMCID: PMC7987688 DOI: 10.1017/s2040174420001233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVD) are important consequences of adverse perinatal conditions such as fetal hypoxia and maternal malnutrition. Cardiac magnetic resonance imaging (CMR) can produce a wealth of physiological information related to the development of the heart. This review outlines the current state of CMR technologies and describes the physiological biomarkers that can be measured. These phenotypes include impaired ventricular and atrial function, maladaptive ventricular remodeling, and the proliferation of myocardial steatosis and fibrosis. The discussion outlines the applications of CMR to understanding the developmental pathways leading to impaired cardiac function. The use of CMR, both in animal models of developmental programming and in human studies, is described. Specific examples are given in a baboon model of intrauterine growth restriction (IUGR). CMR offers great potential as a tool for understanding the sequence of dysfunctional adaptations of developmental origin that can affect the human cardiovascular system.
Collapse
Affiliation(s)
- G D Clarke
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - J Li
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - A H Kuo
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - A J Moody
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Research Imaging Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - P W Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
41
|
Shinoda R, Shinoda Y, Mori T, Yoshimura T. [Retrospective Observational Study on Predictors of Body Weight and BNP Teduction in Cases of Tolvaptan Induction for Heart Failure]. YAKUGAKU ZASSHI 2021; 141:281-288. [PMID: 33518649 DOI: 10.1248/yakushi.20-00221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tolvaptan (TLV) carries the risk of serious side effects, and its introduction requires hospitalization. Therefore, it is important from the viewpoints of safety and medical economics to predict in advance, the patients for whom it will be effective and introduce it. The purpose of this study was to investigate the noninvasive and simple predictors for identifying TLV responders. We conducted a retrospective observational study of patients with heart failure who had TLV introduced at our hospital from January 1, 2017, to December 31, 2018. By using the body weight and BNP reduction as the effect indices, predictors of body weight and BNP reduction were extracted by logistic analysis. The sensitivity and specificity at the cutoff value obtained by ROC analysis were also examined. Among 85 subjects, urine sodium concentration >63 mEq/L [odds ratio (OR): 6.11, 95% confidence interval (CI): 1.36-27.4] was detected as a predictor of body weight reduction. The sensitivity at this cutoff value was 81%, and the specificity was 70%. Serum osmolarity>291 mOsm/L (OR: 3.76, 95% CI: 1.00-14.2), urine potassium concentration<21 mEq/L (OR: 4.45, 95% CI: 1.09-18.2), and urine sodium concentration>71 mEq/L (OR: 7.38, 95% CI: 2.05-26.6) were detected as predictors of BNP reduction. The sensitivities were 62%, 53%, and 73%, and the specificities were 58%, 68%, and 68%, respectively. Therefore, it was suggested that urine sodium concentration may be useful as a predictor of body weight and BNP decrease after TLV induction.
Collapse
Affiliation(s)
- Rie Shinoda
- Department of Pharmacy, Ogaki Municipal Hospital
| | | | | | | |
Collapse
|
42
|
Hasegawa K, Yamaguchi Y, Tanaka M. Differential roles of VPS and RAAS in water homeostasis and a risk for kidney dysfunction in rats undergoing rapid fasting/dehydration with regular exercise. Physiol Rep 2021; 9:e14670. [PMID: 33400404 PMCID: PMC7785051 DOI: 10.14814/phy2.14670] [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: 08/04/2020] [Revised: 10/23/2020] [Accepted: 11/10/2020] [Indexed: 11/24/2022] Open
Abstract
Purpose We examined the effects of rapid restriction of food and fluid intake on the pathways of water homeostasis, the vasopressinergic system (VPS), and the renin–angiotensin–aldosterone system (RAAS), in rats with or without regular exercise. Methods Sprague Dawley rats were divided into the following groups: no intervention, rapid restriction, regular exercise, and rapid restriction combined with regular exercise. Rats in the exercise group performed climbing exercise for 4 weeks. All rats consumed food ad libitum, and those in the rapid restriction group fasted for the last 3 days with no water on the last 1 day. Results Despite no significant differences in body weight among the groups, the kidney weight was decreased when rapid restriction and regular exercise were combined. Rapid restriction reduced the urine volume and increased the urine osmolality, whereas regular exercise did not. Rapid restriction but not regular exercise increased the levels of circulating aldosterone and the renal expression levels of the ion channel SGK‐1 compared to those without rapid restriction, indicating the stimulation of RAAS. Conversely, VPS showed no significant response to these interventions. Moreover, rapid restriction combined with regular exercise induced the renal expression levels of proinflammatory cytokines and increased the active forms of apoptotic effector caspase‐3 compared with the no intervention group. Conclusions Functional significance may differ between VPS and RAAS in water homeostasis in response to rapid restriction. Moreover, the combination of rapid restriction and regular exercise has potentially deleterious effects on the kidney.
Collapse
Affiliation(s)
- KazuyA Hasegawa
- Faculty of Nutritional Sciences, Morioka University, Takizawa city, Japan
| | - Yuya Yamaguchi
- Department of Physiology, Faculty of Medicine, Toho University, Japan
| | - Masashi Tanaka
- Department of Physical Therapy, Health Science University, Minamitsuru-gun, Japan
| |
Collapse
|
43
|
De Souza AMA, Linares A, Speth RC, Campos GV, Ji H, Chianca D, Sandberg K, De Menezes RCA. Severe food restriction activates the central renin angiotensin system. Physiol Rep 2020; 8:e14338. [PMID: 31925945 PMCID: PMC6954120 DOI: 10.14814/phy2.14338] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We previously showed that 2 weeks of a severe food restricted (sFR) diet (40% of the caloric intake of the control (CT) diet) up‐regulated the circulating renin angiotensin (Ang) system (RAS) in female Fischer rats, most likely as a result of the fall in plasma volume. In this study, we investigated the role of the central RAS in the mean arterial pressure (MAP) and heart rate (HR) dysregulation associated with sFR. Although sFR reduced basal mean MAP and HR, the magnitude of the pressor response to intracerebroventricular (icv) microinjection of Ang‐[1‐8] was not affected; however, HR was 57 ± 13 bpm lower 26 min after Ang‐[1‐8] microinjection in the sFR rats and a similar response was observed after losartan was microinjected. The major catabolic pathway of Ang‐[1‐8] in the hypothalamus was via Ang‐[1‐7]; however, no differences were detected in the rate of Ang‐[1‐8] synthesis or degradation between CT and sFR animals. While sFR had no effect on the AT1R binding in the subfornical organ (SFO), the organum vasculosum laminae terminalis (OVLT) and median preoptic nucleus (MnPO) of the paraventricular anteroventral third ventricle, ligand binding increased 1.4‐fold in the paraventricular nucleus (PVN) of the hypothalamus. These findings suggest that sFR stimulates the central RAS by increasing AT1R expression in the PVN as a compensatory response to the reduction in basal MAP and HR. These findings have implications for people experiencing a period of sFR since an activated central RAS could increase their risk of disorders involving over activation of the RAS including renal and cardiovascular diseases.
Collapse
Affiliation(s)
| | - Andrea Linares
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Robert C Speth
- Division of Nephrology & Hypertension, Department of Medicine, Georgetown University, Washington, DC, USA.,Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Glenda V Campos
- Division of Nephrology & Hypertension, Department of Medicine, Georgetown University, Washington, DC, USA
| | - Hong Ji
- Division of Nephrology & Hypertension, Department of Medicine, Georgetown University, Washington, DC, USA
| | - Deoclécio Chianca
- Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade, Federal de Ouro Preto, Ouro Preto, Brazil
| | - Kathryn Sandberg
- Division of Nephrology & Hypertension, Department of Medicine, Georgetown University, Washington, DC, USA
| | - Rodrigo C A De Menezes
- Departamento de Ciências Biológicas, Instituto de Ciências Exatas e Biológicas, Universidade, Federal de Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
44
|
Adin D, Atkins C, Londoño L, Del Nero B. Correction of serum chloride concentration in dogs with congestive heart failure. J Vet Intern Med 2020; 35:51-57. [PMID: 33305873 PMCID: PMC7848309 DOI: 10.1111/jvim.15998] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 11/20/2020] [Accepted: 12/01/2020] [Indexed: 12/03/2022] Open
Abstract
Background Hypochloremia associated with congestive heart failure (CHF) in dogs is likely multifactorial. Loop diuretics cause 1:2 sodium [Na+]:chloride [Cl−] loss, whereas water retention causes a 1:1 [Na+]:[Cl−] dilution. Mathematical [Cl−] correction separates these effects on [Cl−]. Hypothesis We hypothesized that corrected [Cl−] (c[Cl−]) would not differ from measured [Cl−] (m[Cl−]) in dogs with controlled CHF because of loop diuretics, and dogs with refractory CHF would have higher c[Cl−] than m[Cl−], indicating relative water excess. Animals Seventy‐one client‐owned dogs with acquired heart disease, without CHF (NO‐CHF), 76 with Stage C CHF and 24 with Stage D CHF. Methods Clinicopathological data from a previous study were retrospectively analyzed. Corrected [Cl−], m[Cl−], and differences were compared among NO‐CHF, Stage C CHF, and Stage D CHF, using the formula: c[Cl−] = (mid‐reference range [Na+]/measured [Na+]) × m[Cl−]. Results Corrected [Cl−] and m[Cl−] were lower in Stage D vs Stage C and NO‐CHF (all P < .0001). The c[Cl−] was higher than m[Cl−] in Stage D (P < .0001) but not Stage C or NO‐CHF. Median difference between c[Cl−] and m[Cl−] was higher for Stage D vs Stage C (P = .0003). No hypochloremic Stage D dogs had normal c[Cl−], but 11/24 had [Cl−] that was increased by >2 mmol/L. Conclusions and Clinical Importance Serum [Cl−] increased after mathematical correction in Stage D CHF dogs but not in Stage C and NO‐CHF dogs. Although c[Cl−] was higher than m[Cl−] in Stage D dogs supportive of relative water excess, hypochloremia persisted, consistent with concurrent loop diuretic effects on electrolytes. Future study correlating c[Cl−] to antidiuretic hormone concentrations is warranted.
Collapse
Affiliation(s)
- Darcy Adin
- University of Florida, College of Veterinary Medicine, Gainesville, Florida, USA
| | - Clarke Atkins
- North Carolina State University, College of Veterinary Medicine, Raleigh, North Carolina, USA
| | - Leonel Londoño
- University of Florida, College of Veterinary Medicine, Gainesville, Florida, USA
| | - Bruna Del Nero
- University of Florida, College of Veterinary Medicine, Gainesville, Florida, USA
| |
Collapse
|
45
|
Jeon SJ, Hwang HB, Lee NY. Evaluation of Systemic Renin and Angiotensin II Levels in Normal Tension Glaucoma. J Clin Med 2020; 9:jcm9123838. [PMID: 33256138 PMCID: PMC7761439 DOI: 10.3390/jcm9123838] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
The purpose of this study was to investigate the function of the renin-angiotensin-aldosterone system (RAAS) in normal tension glaucoma (NTG) patients by measuring the level of renin and angiotensin II (AngII) in the plasma. Twenty-four patients with NTG and 38 control subjects were included in this study. Renin and AngII were measured in the blood samples of all subjects by enzyme-linked immunosorbent assay (ELISA). No significant differences were found in the complete blood count, fasting glucose, low-density lipoprotein (LDL), and high-sensitivity C-reactive protein (hs-CRP) levels between the control and NTG groups. The systemic concentration and variability of the renin concentration in the blood was significantly higher in the NTG group (p = 0.005 and 0.005, respectively). According to multivariate logistic regression analysis, the variability of the renin concentration was associated with NTG (p = 0.006). In conclusion, the systemic concentration and variability of renin levels were elevated in NTG patients. An altered renin concentration could represent a difference in RAAS function in NTG patients.
Collapse
Affiliation(s)
- Soo Ji Jeon
- Department of Ophthalmology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Korea;
| | - Hyung Bin Hwang
- Department of Ophthalmology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Incheon 21431, Korea;
| | - Na Young Lee
- Department of Ophthalmology, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03312, Korea;
- Correspondence: ; Tel.: +82-2-2030-2795; Fax: +82-2-599-7405
| |
Collapse
|
46
|
Abdel Ghafar MT. An overview of the classical and tissue-derived renin-angiotensin-aldosterone system and its genetic polymorphisms in essential hypertension. Steroids 2020; 163:108701. [PMID: 32717198 DOI: 10.1016/j.steroids.2020.108701] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 07/05/2020] [Accepted: 07/19/2020] [Indexed: 01/25/2023]
Abstract
The renin-angiotensin-aldosterone system (RAAS) is a specific hormonal cascade implicated in the blood pressure control and sodium balance regulation. Several components of this pathway have been identified including renin, angiotensinogen, angiotensin-converting enzyme, angiotensins with a wide range of distinct subtypes and receptors, and aldosterone. The RAAS is not only confined to the systemic circulation but also exists locally in specific tissues such as the heart, brain, and blood vessels with a particular paracrine action. Alteration of RAAS function can contribute to the development of hypertension and the emergence of its associated end-organ damage. Genotypic variations of the different genes of RAAS cascade have been linked to the susceptibility to essential hypertension. Accordingly, to understand the pathogenesis of essential hypertension and its related complications, deep insight into the physiological and genetic aspects of RAAS with its different components and pathways is necessary. In this review, we aimed to illustrate the physiological and genetic aspects of RAAS and the underlying mechanisms which link this system to the predisposition to essential hypertension.
Collapse
|
47
|
Wieruszewski PM, Wittwer ED, Kashani KB, Brown DR, Butler SO, Clark AM, Cooper CJ, Davison DL, Gajic O, Gunnerson KJ, Tendler R, Mara KC, Barreto EF. Angiotensin II Infusion for Shock: A Multicenter Study of Postmarketing Use. Chest 2020; 159:596-605. [PMID: 32882250 DOI: 10.1016/j.chest.2020.08.2074] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/11/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Vasodilatory shock refractory to catecholamine vasopressors and arginine vasopressin is highly morbid and responsible for significant mortality. Synthetic angiotensin II is a potent vasoconstrictor that may be suitable for use in these patients. RESEARCH QUESTION What is the safety and effectiveness of angiotensin II and what variables are associated with a favorable hemodynamic response? STUDY DESIGN AND METHODS We performed a multicenter, retrospective study at five tertiary medical centers in the United States. The primary end point of hemodynamic responsiveness to angiotensin II was defined as attainment of mean arterial pressure (MAP) of ≥ 65 mm Hg with a stable or reduced total vasopressor dosage 3 h after drug initiation. RESULTS Of 270 included patients, 181 (67%) demonstrated hemodynamic responsiveness to angiotensin II. Responders showed a greater increase in MAP (+10.3 mm Hg vs +1.6 mm Hg, P < .001) and reduction in vasopressor dosage (-0.20 μg/kg/min vs +0.04 μg/kg/min; P < .001) compared with nonresponders at 3 h. Variables associated with favorable hemodynamic response included lower lactate concentration (OR 1.11; 95% CI, 1.05-1.17, P < .001) and receipt of vasopressin (OR, 6.05; 95% CI, 1.98-18.6; P = .002). In severity-adjusted multivariate analysis, hemodynamic responsiveness to angiotensin II was associated with reduced likelihood of 30-day mortality (hazard ratio, 0.50; 95% CI, 0.35-0.71; P < .001). Arrhythmias occurred in 28 patients (10%) and VTE was identified in 4 patients. INTERPRETATION In postmarketing use for vasopressor-refractory shock, 67% of angiotensin II recipients demonstrated a favorable hemodynamic response. Patients with lower lactate concentrations and those receiving vasopressin were more likely to respond to angiotensin II. Patients who responded to angiotensin II experienced reduced mortality.
Collapse
Affiliation(s)
- Patrick M Wieruszewski
- Department of Pharmacy, Mayo Clinic, Rochester, MN; Multidisciplinary Epidemiology and Translational Research in Intensive Care, Mayo Clinic, Rochester, MN
| | - Erica D Wittwer
- Multidisciplinary Epidemiology and Translational Research in Intensive Care, Mayo Clinic, Rochester, MN; Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Kianoush B Kashani
- Multidisciplinary Epidemiology and Translational Research in Intensive Care, Mayo Clinic, Rochester, MN; Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Daniel R Brown
- Multidisciplinary Epidemiology and Translational Research in Intensive Care, Mayo Clinic, Rochester, MN; Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | | | - Angela M Clark
- Department of Pharmacy, Michigan Medicine, Ann Arbor, MI
| | - Craig J Cooper
- Department of Pharmacy, Northwestern Memorial Hospital, Chicago, IL
| | - Danielle L Davison
- Departments of Anesthesiology and Critical Care Medicine, George Washington University, Washington, DC
| | - Ognjen Gajic
- Multidisciplinary Epidemiology and Translational Research in Intensive Care, Mayo Clinic, Rochester, MN; Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - Kyle J Gunnerson
- Departments of Emergency Medicine, Anesthesiology, and Internal Medicine, Michigan Medicine, Ann Arbor, MI
| | | | - Kristin C Mara
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN
| | - Erin F Barreto
- Department of Pharmacy, Mayo Clinic, Rochester, MN; Multidisciplinary Epidemiology and Translational Research in Intensive Care, Mayo Clinic, Rochester, MN; Robert D. and Patricia E. Kern Center for the Science of Health Care Delivery, Mayo Clinic, Rochester, MN.
| |
Collapse
|
48
|
Chen F, Cao K, Zhang H, Yu H, Liu Y, Xue Q. Maternal high-fat diet increases vascular contractility in adult offspring in a sex-dependent manner. Hypertens Res 2020; 44:36-46. [PMID: 32719462 DOI: 10.1038/s41440-020-0519-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/25/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022]
Abstract
A maternal high-fat diet (HFD) is a risk factor for cardiovascular diseases in offspring. The aim of the study was to determine whether maternal HFD causes the epigenetic programming of vascular angiotensin II receptors (ATRs) and leads to heightened vascular contraction in adult male offspring in a sex-dependent manner. Pregnant rats were treated with HFD (60% kcal fat). Aortas were isolated from adult male and female offspring. Maternal HFD increased phenylephrine (PE)-and angiotensin II (Ang II)-induced contractions of the aorta in male but not female offspring. NG-nitro-L-arginine (ʟ-NNA; 100 μM) abrogated the maternal HFD-induced increase in PE-mediated contraction. HFD caused a decrease in endothelium-dependent relaxations induced by acetylcholine in male but not female offspring. However, it had no effect on sodium nitroprusside-induced endothelium-independent relaxations of aortas regardless of sex. The AT1 receptor (AT1R) antagonist losartan (10 μM), but not the AT2 receptor (AT2R) antagonist PD123319 (10 μM), blocked Ang II-induced contractions in both control and HFD offspring in both sexes. Maternal HFD increased AT1R but decreased AT2R, leading to an increased ratio of AT1R/AT2R in HFD male offspring, which was associated with selective decreases in DNA methylation at the AT1aR promoter and increases in DNA methylation at the AT2R promoter. The vascular ratio of AT1R/AT2R was not significantly different in HFD female offspring compared with the control group. Our results indicated that maternal HFD caused a differential regulation of vascular AT1R and AT2R gene expression through a DNA methylation mechanism, which may be involved in HFD-induced vascular dysfunction and the development of a hypertensive phenotype in adulthood in a sex-dependent manner.
Collapse
Affiliation(s)
- Fangyuan Chen
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Kaifang Cao
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Haichuan Zhang
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Haili Yu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yinghua Liu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Qin Xue
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China. .,Guangzhou Institute of Cardiovascular Disease, Guangzhou Key Laboratory of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, PR China.
| |
Collapse
|
49
|
Involvement of ACE2/Ang-(1-7)/MAS1 Axis in the Regulation of Ovarian Function in Mammals. Int J Mol Sci 2020; 21:ijms21134572. [PMID: 32604999 PMCID: PMC7369927 DOI: 10.3390/ijms21134572] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
In addition to the classic, endocrine renin-angiotensin system, local renin-angiotensin system (RAS) has been documented in many tissues and organs, including the ovaries. The localization and functional activity of the two opposing axes of the system, viz. ACE1/Ang II/AT1 and ACE2/Ang-(1-7)/MAS1, differs between animal species and varied according to the stage of follicle development. It appears that the angiotensin peptides and their receptors participate in reproductive processes such as folliculogenesis, steroidogenesis, oocyte maturation, and ovulation. In addition, changes in the constituent compounds of local RAS may contribute to pathological conditions, such as polycystic ovary syndrome, ovarian hyperstimulation syndrome, and ovarian cancer. This review article examines the expression, localization, metabolism, and activity of individual elements of the ACE2/Ang-(1-7)/MAS1 axis in the ovaries of various animal species. The manuscript also presents the relationship between the secretion of gonadotropins and sex hormones and expression of Ang-(1-7) and MAS1 receptors. It also summarizes current knowledge regarding the positive and negative impact of ACE2/Ang-(1-7)/MAS1 axis on ovarian function.
Collapse
|
50
|
Endogenous hydrogen sulfide improves vascular remodeling through PPARδ/SOCS3 signaling. J Adv Res 2020; 27:115-125. [PMID: 33318871 PMCID: PMC7728593 DOI: 10.1016/j.jare.2020.06.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/23/2020] [Accepted: 06/08/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction Mounting evidences demonstrated the deficiency of hydrogen sulfide (H2S) facilitated the progression of cardiovascular diseases. However, the exact effects of H2S on vascular remodeling are not consistent. Objectives This study aimed to investigate the beneficial role of endogenous H2S on vascular remodeling. Methods CSE inhibitor, DL-propargylglycine (PPG) was used to treat mice and vascular smooth muscle cells (VSMCs). Sodium hydrosulfide (NaHS) was given to provide hydrogen sulfide. Vascular tension, H&E staining, masson trichrome staining, western blot and CCK8 were used to determine the vascular remodeling, expressions of inflammatory molecules and proliferation of VSMCs. Results The deficiency of endogenous H2S generated vascular remodeling with aggravated active and passive contraction, thicken aortic walls, collagen deposition, increased phosphorylation of STAT3, decreased production of PPARδ and SOCS3 in aortas, which were reversed by NaHS. PPG inhibited expression of PPARδ and SOCS3, stimulated the phosphorylation of STAT3, increased inflammatory molecules production and proliferation rate of VSMCs which could all be corrected by NaHS supply. PPARδ agonist GW501516 offered protections similar to NaHS in PPG treated VSMCs. Aggravated active and passive contraction in PPG mice aortas, upregulated p-STAT3 and inflammatory molecules, downregulated SOCS3 and phenotype transformation in PPG treated VSMCs could be corrected by PPARδ agonist GW501516 treatment. On the contrary, PPARδ antagonist GSK0660 exhibited opposite effects on vascular contraction in aortas, expressions of p-STAT3 and SOCS3 in VSMCs compared with GW501516. Conclusion In a word, endogenous H2S protected against vascular remodeling through preserving PPARδ/SOCS3 anti-inflammatory signaling pathway. Deficiency of endogenous H2S should be considered as a risk factor for VSMCs dysfunction.
Collapse
|