1
|
Singh KD, Karnik SS. Implications of β-Arrestin biased signaling by angiotensin II type 1 receptor for cardiovascular drug discovery and therapeutics. Cell Signal 2024; 124:111410. [PMID: 39270918 DOI: 10.1016/j.cellsig.2024.111410] [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: 07/31/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Angiotensin II receptors, Type 1 (AT1R) and Type 2 (AT2R) are 7TM receptors that play critical roles in both the physiological and pathophysiological regulation of the cardiovascular system. While AT1R blockers (ARBs) have proven beneficial in managing cardiac, vascular and renal maladies they cannot completely halt and reverse the progression of pathologies. Numerous experimental and animal studies have demonstrated that β-arrestin biased AT1R-ligands (such as SII-AngII, S1I8, TRV023, and TRV027) offer cardiovascular benefits by blocking the G protein signaling while retaining the β-arrestin signaling. However, these ligands failed to show improvement in heart-failure outcome over the placebo in a phase IIb clinical trial. One major limitation of current β-arrestin biased AT1R-ligands is that they are peptides with short half-lives, limiting their long-term efficacy in patients. Additionally, β-arrestin biased AT1R-ligand peptides, may inadvertently block AT2R, a promiscuous receptor, potentially negating its beneficial effects in post-myocardial infarction (MI) patients. Therefore, developing a small molecule β-arrestin biased AT1R-ligand with a longer half-life and specificity to AT1R could be more effective in treating heart failure. This approach has the potential to revolutionize the treatment of cardiovascular diseases by offering more sustained and targeted therapeutic effects.
Collapse
Affiliation(s)
- Khuraijam Dhanachandra Singh
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, USA.
| | - Sadashiva S Karnik
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland Clinic, USA.
| |
Collapse
|
2
|
Sauge E, White Z, Lizotte F, Yuen C, Atmuri NDP, Ciufolini MA, Geraldes P, Bernatchez P. Losartan and metabolite EXP3179 activate endothelial function without lowering blood pressure in AT2 receptor KO mice. Eur J Pharmacol 2024; 977:176663. [PMID: 38815786 DOI: 10.1016/j.ejphar.2024.176663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND We have documented profound release of nitric oxide (NO) and endothelium-derived hyperpolarization factor (EDHF) by angiotensin II (ANGII) receptor 1 (AT1) blocker (ARB) losartan and its unique metabolite EXP3179, a pleiotropic effect that may help rationalize the protective properties of ARBs. Since blood pressure (BP) lowering by ARBs likely require an ANGII-dependent switch from AT1 to ANGII receptor 2 (AT2) signaling, a receptor known to stimulate endothelial NO release, we investigated the contribution of AT1 and AT2 to losartan and EXP3179's endothelial function-activating properties. EXPERIMENTAL APPROACH Two AT1 ligands were used in an attempt to block the AT1-dependent endothelium-enhancing effects of EXP3179. AT2-null mice were used to evaluate the acute ex vivo and chronic in vivo effects of EXP3179 (20μM) and losartan (0.6 g/l), respectively, on endothelial function, BP and aortic stiffness. KEY RESULTS Ex vivo blockade of AT1 receptors did not attenuate EXP3179's effects on NO and EDHF-dependent endothelial function activation. We observed significant reductions in PE-induced contractility with EXP3179 in both WT and AT2 knockout (KO) aortic rings. In vivo, a 1-month chronic treatment with losartan did not affect pulse wave velocity (PWV) but decreased PE-induced contraction by 74.9 % in WT (p < 0.0001) and 47.3 % in AT2 KO (p < 0.05). Presence of AT2 was critical to losartan's BP lowering activity. CONCLUSION In contrast to BP lowering, the endothelial function-enhancing effects of losartan and EXP3179 are mostly independent of the classic ANGII/AT1/AT2 pathway, which sheds light on ARB pleiotropism.
Collapse
MESH Headings
- Animals
- Losartan/pharmacology
- Blood Pressure/drug effects
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Mice, Knockout
- Mice
- Receptor, Angiotensin, Type 2/metabolism
- Receptor, Angiotensin, Type 2/genetics
- Male
- Nitric Oxide/metabolism
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 1/genetics
- Imidazoles/pharmacology
- Mice, Inbred C57BL
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Vascular Stiffness/drug effects
- Sulfonamides
- Thiophenes
Collapse
Affiliation(s)
- Elodie Sauge
- Department of Anesthesiology, Pharmacology & Therapeutics, D Department of Chemistry, University of British Columbia (UBC), Vancouver, Canada; Centre for Heart Lung Innovation, University of British Columbia (UBC), Vancouver, Canada
| | - Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, D Department of Chemistry, University of British Columbia (UBC), Vancouver, Canada; Centre for Heart Lung Innovation, University of British Columbia (UBC), Vancouver, Canada
| | - Farah Lizotte
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada
| | - Christopher Yuen
- Department of Anesthesiology, Pharmacology & Therapeutics, D Department of Chemistry, University of British Columbia (UBC), Vancouver, Canada; Centre for Heart Lung Innovation, University of British Columbia (UBC), Vancouver, Canada
| | - N D Prasad Atmuri
- Department of Medicine, Endocrinology Division, Université de Sherbrooke, Québec, Canada
| | - Marco A Ciufolini
- Department of Medicine, Endocrinology Division, Université de Sherbrooke, Québec, Canada
| | - Pedro Geraldes
- Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Québec, Canada; Department of Medicine, Endocrinology Division, Université de Sherbrooke, Québec, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, D Department of Chemistry, University of British Columbia (UBC), Vancouver, Canada; Centre for Heart Lung Innovation, University of British Columbia (UBC), Vancouver, Canada.
| |
Collapse
|
3
|
Samuel CS, Li Y, Wang Y, Widdop RE. Functional crosstalk between angiotensin receptors (types 1 and 2) and relaxin family peptide receptor 1 (RXFP1): Implications for the therapeutic targeting of fibrosis. Br J Pharmacol 2024; 181:2302-2318. [PMID: 36560925 DOI: 10.1111/bph.16019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
Class A, rhodopsin-like, G-protein-coupled receptors (GPCRs) are by far the largest class of GPCRs and are integral membrane proteins used by various cells to convert extracellular signals into intracellular responses. Initially, class A GPCRs were believed to function as monomers, but a growing body of evidence has emerged to suggest that these receptors can function as homodimers and heterodimers and can undergo functional crosstalk to influence the actions of agonists or antagonists acting at each receptor. This review will focus on the angiotensin type 1 (AT1) and type 2 (AT2) receptors, as well as the relaxin family peptide receptor 1 (RXFP1), each of which have their unique characteristics but have been demonstrated to undergo some level of interaction when appropriately co-expressed, which influences the function of each receptor. In particular, this receptor functional crosstalk will be discussed in the context of fibrosis, the tissue scarring that results from a failed wound-healing response to injury, and which is a hallmark of chronic disease and related organ dysfunction. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Yifang Li
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Yan Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
4
|
Somanader DVN, Zhao P, Widdop RE, Samuel CS. The involvement of the Wnt/β-catenin signaling cascade in fibrosis progression and its therapeutic targeting by relaxin. Biochem Pharmacol 2024; 223:116130. [PMID: 38490518 DOI: 10.1016/j.bcp.2024.116130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/06/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Organ scarring, referred to as fibrosis, results from a failed wound-healing response to chronic tissue injury and is characterised by the aberrant accumulation of various extracellular matrix (ECM) components. Once established, fibrosis is recognised as a hallmark of stiffened and dysfunctional tissues, hence, various fibrosis-related diseases collectively contribute to high morbidity and mortality in developed countries. Despite this, these diseases are ineffectively treated by currently-available medications. The pro-fibrotic cytokine, transforming growth factor (TGF)-β1, has emerged as the master regulator of fibrosis progression, owing to its ability to promote various factors and processes that facilitate rapid ECM synthesis and deposition, whilst negating ECM degradation. TGF-β1 signal transduction is tightly controlled by canonical (Smad-dependent) and non-canonical (MAP kinase- and Rho-associated protein kinase-dependent) intracellular protein activity, whereas its pro-fibrotic actions can also be facilitated by the Wnt/β-catenin pathway. This review outlines the pathological sequence of events and contributing roles of TGF-β1 in the progression of fibrosis, and how the Wnt/β-catenin pathway contributes to tissue repair in acute disease settings, but to fibrosis and related tissue dysfunction in synergy with TGF-β1 in chronic diseases. It also outlines the anti-fibrotic and related signal transduction mechanisms of the hormone, relaxin, that are mediated via its negative modulation of TGF-β1 and Wnt/β-catenin signaling, but through the promotion of Wnt/β-catenin activity in acute disease settings. Collectively, this highlights that the crosstalk between TGF-β1 signal transduction and the Wnt/β-catenin cascade may provide a therapeutic target that can be exploited to broadly treat and reverse established fibrosis.
Collapse
Affiliation(s)
- Deidree V N Somanader
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Peishen Zhao
- Drug Discovery Biology Program, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, Victoria 3052, Australia.
| |
Collapse
|
5
|
Naik N, Patel M, Sen R. Developmental Impacts of Epigenetics and Metabolism in COVID-19. J Dev Biol 2024; 12:9. [PMID: 38390960 PMCID: PMC10885083 DOI: 10.3390/jdb12010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Developmental biology is intricately regulated by epigenetics and metabolism but the mechanisms are not completely understood. The situation becomes even more complicated during diseases where all three phenomena are dysregulated. A salient example is COVID-19, where the death toll exceeded 6.96 million in 4 years, while the virus continues to mutate into different variants and infect people. Early evidence during the pandemic showed that the host's immune and inflammatory responses to COVID-19 (like the cytokine storm) impacted the host's metabolism, causing damage to the host's organs and overall physiology. The involvement of angiotensin-converting enzyme 2 (ACE2), the pivotal host receptor for the SARS-CoV-2 virus, was identified and linked to epigenetic abnormalities along with other contributing factors. Recently, studies have revealed stronger connections between epigenetics and metabolism in COVID-19 that impact development and accelerate aging. Patients manifest systemic toxicity, immune dysfunction and multi-organ failure. Single-cell multiomics and other state-of-the-art high-throughput studies are only just beginning to demonstrate the extent of dysregulation and damage. As epigenetics and metabolism directly impact development, there is a crucial need for research implementing cutting-edge technology, next-generation sequencing, bioinformatics analysis, the identification of biomarkers and clinical trials to help with prevention and therapeutic interventions against similar threats in the future.
Collapse
Affiliation(s)
- Noopur Naik
- Department of Molecular, Cellular & Developmental Biology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Mansi Patel
- Institute of Genomics and Integrative Biology, Delhi 110007, India
| | - Rwik Sen
- Active Motif, Inc., Carlsbad, CA 92008, USA
| |
Collapse
|
6
|
Young ON, Bourke JE, Widdop RE. Catch your breath: The protective role of the angiotensin AT 2 receptor for the treatment of idiopathic pulmonary fibrosis. Biochem Pharmacol 2023; 217:115839. [PMID: 37778444 DOI: 10.1016/j.bcp.2023.115839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/28/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease whereby excessive deposition of extracellular matrix proteins (ECM) ultimately leads to respiratory failure. While there have been advances in pharmacotherapies for pulmonary fibrosis, IPF remains an incurable and irreversible disease. There remains an unmet clinical need for treatments that reverse fibrosis, or at the very least have a more tolerable side effect profile than currently available treatments. Transforming growth factor β1(TGFβ1) is considered the main driver of fibrosis in IPF. However, as our understanding of the role of the pulmonary renin-angiotensin system (PRAS) in the pathogenesis of IPF increases, it is becoming clear that targeting angiotensin receptors represents a potential novel treatment strategy for IPF - in particular, via activation of the anti-fibrotic angiotensin type 2 receptor (AT2R). This review describes the current understanding of the pathophysiology of IPF and the mediators implicated in its pathogenesis; focusing on TGFβ1, angiotensin II and related peptides in the PRAS and their contribution to fibrotic processes in the lung. Preclinical and clinical assessment of currently available AT2R agonists and the development of novel, highly selective ligands for this receptor will also be described, with a focus on compound 21, currently in clinical trials for IPF. Collectively, this review provides evidence of the potential of AT2R as a novel therapeutic target for IPF.
Collapse
Affiliation(s)
- Olivia N Young
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Jane E Bourke
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Robert E Widdop
- Department of Pharmacology and Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
7
|
Yi W, Chen F, Zhang H, Tang P, Yuan M, Wen J, Wang S, Cai Z. Role of angiotensin II in aging. Front Aging Neurosci 2022; 14:1002138. [PMID: 36533172 PMCID: PMC9755866 DOI: 10.3389/fnagi.2022.1002138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 11/08/2022] [Indexed: 10/29/2023] Open
Abstract
Aging is an inevitable progressive decline in physiological organ function that increases the chance of disease and death. The renin-angiotensin system (RAS) is involved in the regulation of vasoconstriction, fluid homeostasis, cell growth, fibrosis, inflammation, and oxidative stress. In recent years, unprecedented advancement has been made in the RAS study, particularly with the observation that angiotensin II (Ang II), the central product of the RAS, plays a significant role in aging and chronic disease burden with aging. Binding to its receptors (Ang II type 1 receptor - AT1R in particular), Ang II acts as a mediator in the aging process by increasing free radical production and, consequently, mitochondrial dysfunction and telomere attrition. In this review, we examine the physiological function of the RAS and reactive oxygen species (ROS) sources in detail, highlighting how Ang II amplifies or drives mitochondrial dysfunction and telomere attrition underlying each hallmark of aging and contributes to the development of aging and age-linked diseases. Accordingly, the Ang II/AT1R pathway opens a new preventive and therapeutic direction for delaying aging and reducing the incidence of age-related diseases in the future.
Collapse
Affiliation(s)
- Wenmin Yi
- Department of Neurology, Chongqing Medical University, Chongqing, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Fei Chen
- Department of Neurology, Chongqing Medical University, Chongqing, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Huiji Zhang
- Department of Neurology, Chongqing Medical University, Chongqing, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
| | - Peng Tang
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
| | - Minghao Yuan
- Department of Neurology, Chongqing Medical University, Chongqing, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Jie Wen
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
- Department and Institute of Neurology, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shengyuan Wang
- Department of Neurology, Chongqing Medical University, Chongqing, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Zhiyou Cai
- Department of Neurology, Chongqing Medical University, Chongqing, China
- Chongqing Institute Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, China
- Chongqing School, University of Chinese Academy of Sciences, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| |
Collapse
|
8
|
Steckelings UM, Widdop RE, Sturrock ED, Lubbe L, Hussain T, Kaschina E, Unger T, Hallberg A, Carey RM, Sumners C. The Angiotensin AT 2 Receptor: From a Binding Site to a Novel Therapeutic Target. Pharmacol Rev 2022; 74:1051-1135. [PMID: 36180112 PMCID: PMC9553111 DOI: 10.1124/pharmrev.120.000281] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/19/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022] Open
Abstract
Discovered more than 30 years ago, the angiotensin AT2 receptor (AT2R) has evolved from a binding site with unknown function to a firmly established major effector within the protective arm of the renin-angiotensin system (RAS) and a target for new drugs in development. The AT2R represents an endogenous protective mechanism that can be manipulated in the majority of preclinical models to alleviate lung, renal, cardiovascular, metabolic, cutaneous, and neural diseases as well as cancer. This article is a comprehensive review summarizing our current knowledge of the AT2R, from its discovery to its position within the RAS and its overall functions. This is followed by an in-depth look at the characteristics of the AT2R, including its structure, intracellular signaling, homo- and heterodimerization, and expression. AT2R-selective ligands, from endogenous peptides to synthetic peptides and nonpeptide molecules that are used as research tools, are discussed. Finally, we summarize the known physiological roles of the AT2R and its abundant protective effects in multiple experimental disease models and expound on AT2R ligands that are undergoing development for clinical use. The present review highlights the controversial aspects and gaps in our knowledge of this receptor and illuminates future perspectives for AT2R research. SIGNIFICANCE STATEMENT: The angiotensin AT2 receptor (AT2R) is now regarded as a fully functional and important component of the renin-angiotensin system, with the potential of exerting protective actions in a variety of diseases. This review provides an in-depth view of the AT2R, which has progressed from being an enigma to becoming a therapeutic target.
Collapse
Affiliation(s)
- U Muscha Steckelings
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert E Widdop
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Edward D Sturrock
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Lizelle Lubbe
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Tahir Hussain
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Elena Kaschina
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Thomas Unger
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Anders Hallberg
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Robert M Carey
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| | - Colin Sumners
- Institute of Molecular Medicine, Department of Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark (U.M.S.); Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Clayton, Victoria, Australia (R.E.W.); Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Republic of South Africa (E.D.S., L.L.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (T.H.); Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal (CMR) Research Center, DZHK (German Centre for Cardiovascular Research), Berlin, Germany (E.K.); CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands (T.U.); Department of Medicinal Chemistry, Faculty of Pharmacy, Uppsala University, Uppsala, Sweden (A.H.); Division of Endocrinology and Metabolism, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia (R.M.C.); and Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida (C.S.)
| |
Collapse
|
9
|
Tada AM, Hamezah HS, Pahrudin Arrozi A, Abu Bakar ZH, Yanagisawa D, Tooyama I. Pharmaceutical Potential of Casein-Derived Tripeptide Met-Lys-Pro: Improvement in Cognitive Impairments and Suppression of Inflammation in APP/PS1 Mice. J Alzheimers Dis 2022; 89:835-848. [PMID: 35964178 PMCID: PMC9535549 DOI: 10.3233/jad-220192] [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] [Indexed: 11/23/2022]
Abstract
Background: Tripeptide Met-Lys-Pro (MKP), a component of casein hydrolysates, has effective angiotensin-converting enzyme (ACE) inhibitory activity. Brain angiotensin II enzyme activates the NADPH oxidase complex via angiotensin II receptor type 1 (AT1) and enhances oxidative stress injury. ACE inhibitors improved cognitive function in Alzheimer’s disease (AD) mouse models and previous clinical trials. Thus, although undetermined, MKP may be effective against pathological amyloid-β (Aβ) accumulation-induced cognitive impairment. Objective: The current study aimed to investigate the potential of MKP as a pharmaceutical against AD by examining MKP’s effect on cognitive function and molecular changes in the brain using double transgenic (APP/PS1) mice. Methods: Experimental procedures were conducted in APP/PS1 mice (n = 38) with a C57BL/6 background. A novel object recognition test was used to evaluate recognition memory. ELISA was used to measure insoluble Aβ40, Aβ42, and TNF-α levels in brain tissue. Immunohistochemical analysis allowed the assessment of glial cell activation in MKP-treated APP/PS1 mice. Results: The novel object recognition test revealed that MKP-treated APP/PS1 mice showed significant improvement in recognition memory. ELISA of brain tissue showed that MKP significantly reduced insoluble Aβ40, Aβ42, and TNF-α levels. Immunohistochemical analysis indicated the suppression of the marker for microglia and reactive astrocytes in MKP-treated APP/PS1 mice. Conclusion: Based on these results, we consider that MKP could ameliorate pathological Aβ accumulation-induced cognitive impairment in APP/PS1 mice. Furthermore, our findings suggest that MKP potentially contributes to preventing cognitive decline in AD.
Collapse
Affiliation(s)
- Asuka Matsuzaki Tada
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan.,Functional Food Ingredients Group, Food Ingredients and Technology Institute, R&D Division, Morinaga Milk Industry Co., Ltd., Zama, Japan
| | - Hamizah Shahirah Hamezah
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan.,Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, UKM Bangi, Selangor, Malaysia
| | - Aslina Pahrudin Arrozi
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | | | - Daijiro Yanagisawa
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
10
|
Samuel CS, Bennett RG. Relaxin as an anti-fibrotic treatment: Perspectives, challenges and future directions. Biochem Pharmacol 2021; 197:114884. [PMID: 34968489 DOI: 10.1016/j.bcp.2021.114884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Fibrosis refers to the scarring and hardening of tissues, which results from a failed immune system-coordinated wound healing response to chronic organ injury and which manifests from the aberrant accumulation of various extracellular matrix components (ECM), primarily collagen. Despite being a hallmark of prolonged tissue damage and related dysfunction, and commonly associated with high morbidity and mortality, there are currently no effective cures for its regression. An emerging therapy that meets several criteria of an effective anti-fibrotic treatment, is the recombinant drug-based form of the human hormone, relaxin (also referred to as serelaxin, which is bioactive in several other species). This review outlines the broad anti-fibrotic and related organ-protective roles of relaxin, mainly from studies conducted in preclinical models of ageing and fibrotic disease, including its ability to ameliorate several aspects of fibrosis progression and maturation, from immune cell infiltration, pro-inflammatory and pro-fibrotic cytokine secretion, oxidative stress, organ hypertrophy, cell apoptosis, myofibroblast differentiation and ECM production, to its ability to facilitate established ECM degradation. Studies that have compared and/or combined these therapeutic effects of relaxin with current standard of care medication have also been discussed, along with the main challenges that have hindered the translation of the anti-fibrotic efficacy of relaxin to the clinic. The review then outlines the future directions as to where scientists and several pharmaceutical companies that have recognized the therapeutic potential of relaxin are working towards, to progress its development as a treatment for human patients suffering from various fibrotic diseases.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Robert G Bennett
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, Division of Diabetes, Endocrinology & Metabolism, University of Nebraska Medical Center, Omaha, NE 68198-4130, USA.
| |
Collapse
|
11
|
Association Between the Angiotensin II/Angiotensin (1-7) Imbalance and Left Ventricular Hypertrophy in Patients with Heart Failure. ACTA MEDICA BULGARICA 2021. [DOI: 10.2478/amb-2021-0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Introduction: Angiotensin II (AngII) and angiotensin-(1-7) [Ang-(1-7)] are key components of the renin angiotensin system (RAS). They exhibit counter-regulatory effects in the systemic circulation, as well as in tissues important for cardiovascular regulation.
Aim: To investigate the association between the AngII/Ang-(1-7) balance and left ventricular hypertrophy (LVH) in patients with heart failure and mid-range ejection fraction (HFmrEF).
Material and methods: 56 patients with HFmrEF were included, with a mean age of 65.62 ± 9.69 years and 22 age- and sex-matched healthy subjects, their mean age - 56.4 ± 5.53 years. The patients were divided in two subgroups: subjects with left ventricular hypertrophy (n = 32); (HFmrEF+LVH) and subjects without left ventricular hypertrophy (n = 24); (HFmrEFLVH). AngII and Ang-(1-7) levels were measured with an ELISA kit.
Results: Patients with HFmrEF+LVH showed significantly higher levels of AngII: 8.53 pg/mL (1.47 ÷ 13.0) than HFmrEF-LVH – 1.33 pg/mL (0.47 ÷ 6.93) and healthy controls – 1.53 pg/mL (0.27 ÷ 5.21); (KW = 3.48; p = 0.04). There was no significant difference between Ang-(1-7) levels in all patients compared to controls (p > 0.05). AngII/Ang-(1-7) ratio was significantly higher in all patients than controls: 3.81 (2.03 ÷ 6.66) vs. 1.5 (0.93 ÷ 2.06) (KW = 18.68; p < 0.001). Patients with HFmrEF+LVH showed statistically higher AngII/Ang-(1-7) ratio compared with controls (4.7 vs. 1.5). Moreover, subjects with LVH showed the highest AngII/Ang-(1-7) ratio among all particpants in the study. The AngII/Ang-(1-7) ratio correlated with LVH (r = -0.39; p = 0.03), DBP (r = 0.25; p = 0.04), HDL (r = 0.33; p = 0.01), SBP (r = 0.34; p = 0.01).
Conclusion: Our study showed an association between AngII/Ang-(1-7) ratio, blood pressure and LVH. The imbalance between Ang-II and Ang-(1-7) could contribute to the mechanisms determining LVH in HFmrEF. Further studies are warranted to clarify whether evaluation of serum Ang-II/Ang-(1-7) ratio could predict LVH development in patients with HFmrEF.
Collapse
|
12
|
Qi Z, Wang T, Chen X, Wong CK, Ding Q, Sauer H, Chen ZF, Long C, Yao X, Cai Z, Tsang SY. Extracellular and Intracellular Angiotensin II Regulate the Automaticity of Developing Cardiomyocytes via Different Signaling Pathways. Front Mol Biosci 2021; 8:699827. [PMID: 34513920 PMCID: PMC8425478 DOI: 10.3389/fmolb.2021.699827] [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: 04/24/2021] [Accepted: 07/27/2021] [Indexed: 12/02/2022] Open
Abstract
Angiotensin II (Ang II) plays an important role in regulating various physiological processes. However, little is known about the existence of intracellular Ang II (iAng II), whether iAng II would regulate the automaticity of early differentiating cardiomyocytes, and the underlying mechanism involved. Here, iAng II was detected by immunocytochemistry and ultra-high performance liquid chromatography combined with electrospray ionization triple quadrupole tandem mass spectrometry in mouse embryonic stem cell–derived cardiomyocytes (mESC-CMs) and neonatal rat ventricular myocytes. Expression of AT1R-YFP in mESC-CMs revealed that Ang II type 1 receptors were located on the surface membrane, while immunostaining of Ang II type 2 receptors (AT2R) revealed that AT2R were predominately located on the nucleus and the sarcoplasmic reticulum. While extracellular Ang II increased spontaneous action potentials (APs), dual patch clamping revealed that intracellular delivery of Ang II or AT2R activator C21 decreased spontaneous APs. Interestingly, iAng II was found to decrease the caffeine-induced increase in spontaneous APs and caffeine-induced calcium release, suggesting that iAng II decreased spontaneous APs via the AT2R- and ryanodine receptor–mediated pathways. This is the first study that provides evidence of the presence and function of iAng II in regulating the automaticity behavior of ESC-CMs and may therefore shed light on the role of iAng II in fate determination.
Collapse
Affiliation(s)
- Zenghua Qi
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,Institute of Environmental Health and Pollution Control, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Tao Wang
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, SAR China
| | - Xiangmao Chen
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Chun Kit Wong
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Qianqian Ding
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Heinrich Sauer
- Department of Physiology, Justus Liebig University Giessen, Giessen, Germany
| | - Zhi-Feng Chen
- Institute of Environmental Health and Pollution Control, School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou, China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiaoqiang Yao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| | - Zongwei Cai
- State Key Laboratory of Environmental and Biological Analysis, Department of Chemistry, Hong Kong Baptist University, Kowloon, Hong Kong, SAR China
| | - Suk Ying Tsang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China.,State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR China
| |
Collapse
|
13
|
Shen J, Fan Z, Sun G, Qi G. Sacubitril/valsartan (LCZ696) reduces myocardial injury following myocardial infarction by inhibiting NLRP3‑induced pyroptosis via the TAK1/JNK signaling pathway. Mol Med Rep 2021; 24:676. [PMID: 34296299 PMCID: PMC8335743 DOI: 10.3892/mmr.2021.12315] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to investigate the protective effects of sacubitril/valsartan (LCZ696) on ventricular remodeling in myocardial infarction (MI) and the effects of the inflammasome‑mediated inflammatory response. First, a rat model was established. Animals were then treated with LCZ696 so that the histopathological changes associated with ventricular remodeling could be investigated. The serum levels of the inflammatory factors IL‑18 and IL‑1β were also determined by ELISA. Immunofluorescence was used to investigate the ratio of pyroptosis following MI modelling. Western blotting and reverse transcription‑quantitative PCR were used to detect the relative expression levels of proteins and mRNAs in the transforming growth factor β‑activated kinase‑1 (TAK1)/JNK pathway and those associated with the NLR pyrin family domain containing 3 (NLRP3) inflammasome, respectively. The present study also investigated the regulatory mechanisms and associations between the TAK1 and JNK pathways, NOD‑, leucine‑rich repeat‑ and the NLRP3 inflammasome, in H9C2 cells and myocardial cells from the rat model of MI. LCZ696 improved MI‑induced myocardial fibrosis, rescued myocardial injury and suppressed the release of inflammatory factors. With regards to myocardial cell damage, pyroptosis in cardiomyocytes was observed. The in vitro experiments demonstrated that the overexpression of TAK1 promoted lysis of the N‑terminal of GSDMD, thereby activating the NLRP3 inflammasome and promoting the conversion of pro‑IL‑1β and pro‑IL‑18 into mature IL‑1β and IL‑18, respectively. In contrast, the silencing of TAK1 inhibited the expression levels of the NLRP3 inflammasome. In summary, LCZ696 reduced the expression levels of the NLRP3 inflammasome, suppressed inflammatory responses, improved the ventricular remodeling and exhibited protective effects in the MI heart by inhibiting the TAK1/JNK signaling pathway.
Collapse
Affiliation(s)
- Jianfen Shen
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhongbao Fan
- Department of Hepatobiliary Surgery, People's Hospital of China Medical University, Liaoning Provincial People's Hospital, Shenyang, Liaoning 110016, P.R. China
| | - Guang Sun
- Department of Geriatric Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Guoxian Qi
- Department of Geriatric Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
14
|
Yu J, Wang S, Shi W, Zhou W, Niu Y, Huang S, Zhang Y, Zhang A, Jia Z. Roxadustat (FG-4592) prevents Ang II hypertension by targeting angiotensin receptors and eNOS. JCI Insight 2021; 6:e133690. [PMID: 34403364 PMCID: PMC8492313 DOI: 10.1172/jci.insight.133690] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 08/11/2021] [Indexed: 11/28/2022] Open
Abstract
The prevalence of hypertension is increasing globally, while strategies for prevention and treatment of hypertension remain limited. FG-4592 (Roxadustat) is a potentially novel, orally active small-molecule hypoxia-inducible factor (HIF) stabilizer and is being used clinically to treat chronic kidney disease (CKD) anemia. In the present study, we evaluate the effects of FG-4592 on hypertension. In an angiotensin II (Ang II) hypertension model, FG-4592 abolished hypertensive responses; prevented vascular thickening, cardiac hypertrophy, and kidney injury; downregulated AGTR1 expression; and enhanced AGTR2, endothelial NO synthase (eNOS), and HIF1α protein levels in the aortas of mice. Additionally, the levels of thiobarbituric acid reactive substances (TBARs) in blood and urine were diminished by FG-4592 treatment. In vascular smooth muscle cells, FG-4592 treatment reduced angiotensin receptor type 1 (AGTR1) and increased AGTR2 levels, while preventing Ang II–induced oxidative stress. In vascular endothelial cells, FG-4592 upregulated total and phosphorylated eNOS. Moreover, FG-4592 treatment was hypotensive in L-NAME–induced hypertension. In summary, FG-4592 treatment remarkably ameliorated hypertension and organ injury, possibly through stabilizing HIF1α and subsequently targeting eNOS, AGTR1, AGTR2, and oxidative stress. Therefore, in addition to its role in treating CKD anemia, FG-4592 could be explored as a treatment for hypertension associated with high renin angiotensin system (RAS) activity or eNOS defects.
Collapse
Affiliation(s)
- Jing Yu
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Shuqin Wang
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Wei Shi
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Yujia Niu
- Department of Nephrology, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Kocic G, Gajic M, Tomovic K, Hadzi-Djokic J, Anderluh M, Smelcerovic A. Purine adducts as a presumable missing link for aristolochic acid nephropathy-related cellular energy crisis, potential anti-fibrotic prevention and treatment. Br J Pharmacol 2021; 178:4411-4427. [PMID: 34235731 DOI: 10.1111/bph.15618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 12/20/2022] Open
Abstract
Aristolochic acid nephropathy is a progressive exposome-induced disease characterized by tubular atrophy and fibrosis culminating in end-stage renal disease and malignancies. The molecular mechanisms of the energy crisis as a putative cause of fibrosis have not yet been elucidated. In light of the fact that aristolochic acid forms DNA and RNA adducts by covalent binding of aristolochic acid metabolites to exocyclic amino groups of (deoxy)adenosine and (deoxy)guanosine, we hypothesize here that similar aristolochic acid adducts may exist with other purine-containing molecules. We also provide new insights into the aristolochic acid-induced energy crisis and presumably a link between already known mechanisms. In addition, an overview of potential targets in fibrosis treatment is provided, which is followed by recommendations on possible preventive measures that could be taken to at least postpone or partially alleviate aristolochic acid nephropathy.
Collapse
Affiliation(s)
- Gordana Kocic
- Department of Biochemistry, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Mihajlo Gajic
- Department of Pharmacy, Faculty of Medicine, University of Nis, Nis, Serbia
| | - Katarina Tomovic
- Department of Pharmacy, Faculty of Medicine, University of Nis, Nis, Serbia
| | | | - Marko Anderluh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Andrija Smelcerovic
- Department of Chemistry, Faculty of Medicine, University of Nis, Nis, Serbia
| |
Collapse
|
16
|
Tufiño C, Vanegas M, Velázquez Nevárez R, Villanueva López C, Bobadilla Lugo RA. Divergent impact of gestational diabetes mellitus between the thoracic and abdominal rat aorta: Influence of endothelium and angiotensin II receptors. Eur J Pharmacol 2021; 899:173981. [PMID: 33689706 DOI: 10.1016/j.ejphar.2021.173981] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/15/2021] [Accepted: 02/23/2021] [Indexed: 01/23/2023]
Abstract
Gestational diabetes mellitus (GDM) affects 5-10% of pregnancies and increases the risk of fetal and maternal adverse outcomes. Interestingly, the vascular response to AngII is decreased by pregnancy while the response is increased by diabetes. It remains unclear how GDM affects vascular tone and how angiotensin II receptors contribute to these changes. In this work, we sought to establish the vascular impact of a hypercaloric diet-induced GDM through changes in AT1 and AT2 receptor's expression. Female rats fed for 7 weeks with standard (SD) or hypercaloric (HD) diet were divided at week 4. Half of the rats of each group were mated to become pregnant and those fed with a HD developed GDM. AngII-induced vasoconstriction was measured in thoracic or abdominal aorta rings using a conventional isolated organ bath and AT1 and AT2 receptors were searched by immunohistochemistry. Experiments where conducted on the pregnant standard diet group (PSD) and the pregnant hypercaloric-gestational diabetes mellitus group (PHD-GDM). Vasoconstriction was reduced in the thoracic aorta (P < 0.05 vs PSD) but increased in the abdominal aorta of PHD-GDM rats (P < 0.05 vs PSD). Blockade of AT2 receptors using PD123319 decreased vasoconstriction, particularly in the abdominal aorta of PHD-GDM animals (P < 0.05 vs PSD). PHD-GDM increased AT1 receptors expression (P < 0.05 vs PSD). Also, PHD-GDM reverted physiologic hypoglycemia and hypotension of healthy pregnancy. Findings provide new insight into the hypercaloric diet induced damage on the vasculature during pregnancy.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Angiotensin Receptor Antagonists/pharmacology
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/physiopathology
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/physiopathology
- Diabetes, Gestational/metabolism
- Diabetes, Gestational/physiopathology
- Disease Models, Animal
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Female
- Pregnancy
- Rats, Wistar
- Receptor, Angiotensin, Type 1/agonists
- Receptor, Angiotensin, Type 1/metabolism
- Receptor, Angiotensin, Type 2/agonists
- Receptor, Angiotensin, Type 2/metabolism
- Signal Transduction
- Vasoconstriction/drug effects
- Vasoconstrictor Agents/pharmacology
- Rats
Collapse
Affiliation(s)
- Cecilia Tufiño
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Santo Tomás, México, 11340, D.F, Mexico
| | - Miriam Vanegas
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Santo Tomás, México, 11340, D.F, Mexico
| | - Ruth Velázquez Nevárez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Santo Tomás, México, 11340, D.F, Mexico
| | - Cleva Villanueva López
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Santo Tomás, México, 11340, D.F, Mexico
| | - Rosa Amalia Bobadilla Lugo
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Col. Santo Tomás, México, 11340, D.F, Mexico.
| |
Collapse
|
17
|
Pavo N, Prausmüller S, Spinka G, Goliasch G, Bartko PE, Wurm R, Arfsten H, Strunk G, Poglitsch M, Domenig O, Mascherbauer J, Uyanik-Ünal K, Hengstenberg C, Zuckermann A, Hülsmann M. Myocardial Angiotensin Metabolism in End-Stage Heart Failure. J Am Coll Cardiol 2021; 77:1731-1743. [PMID: 33832600 DOI: 10.1016/j.jacc.2021.01.052] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND The myocardium exhibits an adaptive tissue-specific renin-angiotensin system (RAS), and local dysbalance may circumvent the desired effects of pharmacologic RAS inhibition, a mainstay of heart failure with reduced ejection fraction (HFrEF) therapy. OBJECTIVES This study sought to investigate human myocardial tissue RAS regulation of the failing heart in the light of current therapy. METHODS Fifty-two end-stage HFrEF patients undergoing heart transplantation (no RAS inhibitor: n = 9; angiotensin-converting enzyme [ACE] inhibitor: n = 28; angiotensin receptor blocker [ARB]: n = 8; angiotensin receptor neprilysin-inhibitor [ARNi]: n = 7) were enrolled. Myocardial angiotensin metabolites and enzymatic activities involved in the metabolism of the key angiotensin peptides angiotensin 1-8 (AngII) and Ang1-7 were determined in left ventricular samples by mass spectrometry. Circulating angiotensin concentrations were assessed for a subgroup of patients. RESULTS AngII and Ang2-8 (AngIII) were the dominant peptides in the failing heart, while other metabolites, especially Ang1-7, were below the detection limit. Patients receiving an ARB component (i.e., ARB or ARNi) had significantly higher levels of cardiac AngII and AngIII (AngII: 242 [interquartile range (IQR): 145.7 to 409.9] fmol/g vs 63.0 [IQR: 19.9 to 124.1] fmol/g; p < 0.001; and AngIII: 87.4 [IQR: 46.5 to 165.3] fmol/g vs 23.0 [IQR: <5.0 to 59.3] fmol/g; p = 0.002). Myocardial AngII concentrations were strongly related to circulating AngII levels. Myocardial RAS enzyme regulation was independent from the class of RAS inhibitor used, particularly, a comparable myocardial neprilysin activity was observed for patients with or without ARNi. Tissue chymase, but not ACE, is the main enzyme for cardiac AngII generation, whereas AngII is metabolized to Ang1-7 by prolyl carboxypeptidase but not to ACE2. There was no trace of cardiac ACE2 activity. CONCLUSIONS The failing heart contains considerable levels of classical RAS metabolites, whereas AngIII might be an unrecognized mediator of detrimental effects on cardiovascular structure. The results underline the importance of pharmacologic interventions reducing circulating AngII actions, yet offer room for cardiac tissue-specific RAS drugs aiming to limit myocardial AngII/AngIII peptide accumulation and actions.
Collapse
Affiliation(s)
- Noemi Pavo
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria.
| | - Suriya Prausmüller
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Georg Spinka
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Georg Goliasch
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Philipp E Bartko
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Raphael Wurm
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Henrike Arfsten
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Guido Strunk
- Complexity Research, Vienna, Austria; Department of Statistics, Complexity Research, FH Campus Vienna, Vienna, Austria; Department of Entrepreneurship and Economic Education, Faculty of Business and Economics, Technical University Dortmund, Dortmund, Germany
| | | | | | - Julia Mascherbauer
- Karl Landsteiner University of Health Sciences, Department of Internal Medicine 3, University Hospital St. Pölten, Krems, Austria
| | - Keziban Uyanik-Ünal
- Clinical Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Christian Hengstenberg
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Andreas Zuckermann
- Clinical Division of Cardiac Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Martin Hülsmann
- Clinical Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
18
|
The Tissue Renin-Angiotensin System and Its Role in the Pathogenesis of Major Human Diseases: Quo Vadis? Cells 2021; 10:cells10030650. [PMID: 33804069 PMCID: PMC7999456 DOI: 10.3390/cells10030650] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 01/18/2023] Open
Abstract
Evidence has arisen in recent years suggesting that a tissue renin-angiotensin system (tRAS) is involved in the progression of various human diseases. This system contains two regulatory pathways: a pathological pro-inflammatory pathway containing the Angiotensin Converting Enzyme (ACE)/Angiotensin II (AngII)/Angiotensin II receptor type 1 (AGTR1) axis and a protective anti-inflammatory pathway involving the Angiotensin II receptor type 2 (AGTR2)/ACE2/Ang1–7/MasReceptor axis. Numerous studies reported the positive effects of pathologic tRAS pathway inhibition and protective tRAS pathway stimulation on the treatment of cardiovascular, inflammatory, and autoimmune disease and the progression of neuropathic pain. Cell senescence and aging are known to be related to RAS pathways. Further, this system directly interacts with SARS-CoV 2 and seems to be an important target of interest in the COVID-19 pandemic. This review focuses on the involvement of tRAS in the progression of the mentioned diseases from an interdisciplinary clinical perspective and highlights therapeutic strategies that might be of major clinical importance in the future.
Collapse
|
19
|
Katsi V, Pavlidis G, Charalambous G, Tousoulis D, Toutouzas K. COVID-19, Angiotensin-Converting Enzyme 2 and Renin-Angiotensin System Inhibition: Implications for Practice. Curr Hypertens Rev 2021; 18:3-10. [PMID: 33475077 DOI: 10.2174/1573402117666210121100201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Recent studies suggested that patients with coronavirus disease 2019 (COVID-19) who use renin-angiotensin system (RAS) inhibitors have an increased risk of respiratory failure and death. The hypothesis was that angiotensin-converting enzyme inhibitor (ACEIs) or angiotensin receptor blocker (ARBs) may up-regulate ACE2 expression that is used as receptor for viral entry into cells. OBJECTIVE The purpose of this review is to discuss the existing evidence on the interaction between COVID-19 infection, ACE2 and ACEIs or ARBs and to examine the main implications for clinical practice. In addition, novel therapeutic strategies for blocking ACE2-mediated COVID-19 infection will be displayed. METHODS We performed a comprehensive review of the literature to identify data from clinical and experimental studies for the association between COVID-19 infection, ACE2 and RAS inhibition. RESULTS The current clinical and experimental evidence for ACEIs or ARBs to facilitate severe acute respiratory distress syndrome-coronavirus-2 (SARS-CoV-2) is insufficient to suggest discontinuing these drugs. Several observational studies arrive at the conclusion that the continued use of RAS inhibitors is unlike to be harmful in COVID-19-positive patients. CONCLUSIONS Further randomized trials are needed to answer definitely the question of whether RAS inhibitors are harmful or beneficial to patients with COVID-19.
Collapse
Affiliation(s)
- Vasiliki Katsi
- 1 st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens. Greece
| | - George Pavlidis
- Emergency Department, 'Hippokration' General Hospital, Athens. Greece
| | | | - Dimitrios Tousoulis
- 1 st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens. Greece
| | - Konstantinos Toutouzas
- 1 st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens. Greece
| |
Collapse
|
20
|
Cismaru AC, Cismaru LG, Nabavi SF, Berindan-Neagoe I, Clementi E, Banach M, Nabavi SM. Game of "crowning" season 8: RAS and reproductive hormones in COVID-19 - can we end this viral series? Arch Med Sci 2021; 17:275-284. [PMID: 33747262 PMCID: PMC7959061 DOI: 10.5114/aoms.2020.96604] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/27/2020] [Indexed: 01/08/2023] Open
Abstract
The outbreak of a newly identified coronavirus, the SARS-CoV-2 (alternative name 2019-nCoV), capable of jumping across species causing zoonosis with severe acute respiratory syndromes (SARS), has alerted authorities worldwide. Soon after the epidemic was first detected in the city of Wuhan in the Hubei Province of China, starting in late December 2019, the virus spread over multiple countries in different continents, being declared a pandemic by March 2020. The demographic characteristics of the infected patients suggest that age, sex, and comorbidities are predictive factors for the fatality of the infection. The mechanisms of viral entry into the human host cells seem to be in a close relationship with the mechanisms of regulating the renin-angiotensin system (RAS), which may explain the pathogenesis associated with the infection. This brings new insights into the possibilities of exploiting viral entry mechanisms to limit associated complications by means of enhancing the resistance of the infected patients using methods of regulating the RAS and strategies of modulating ACE2 expression. In this perspective article we exploit the mechanisms of COVID-19 pathogenesis based on the demographic characteristics of the infected patients reported in the recent literature and explore several approaches of limiting the initial steps of viral entry and pathogenesis based on viral interactions with ACE2 and RAS. We further discuss the implications of reproductive hormones in the regulation of the RAS and investigate the premise of using endocrine therapy against COVID-19.
Collapse
Affiliation(s)
- Andrei Cosmin Cismaru
- Research Centre for functional Genomics, Biomedicine, and Translational Medicine, The “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Functional Sciences, Immunology, and Allergology, The “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca, Romania
| | - Laurentiu Gabriel Cismaru
- Department of Internal Medicine, Cardiology-Rehabilitation, The “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
- Division of Translational Medicine, Baqiyatallah Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ioana Berindan-Neagoe
- Research Centre for functional Genomics, Biomedicine, and Translational Medicine, The “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- MEDFUTURE – Research Centre for Advanced Medicine – The “Iuliu Hatieganu” University of Medicine and Pharmacy, Cluj-Napoca, Romania
- The Functional Genomics Department, The Oncology Institute “Prof. Dr. Ion Chiricuta”, Cluj-Napoca, Romania
| | - Emilio Clementi
- E. Medea Scientific Institute, Bosisio Parini, Italy
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, Luigi Sacco University Hospital, Università degli Studi di Milano, Milan, Italy
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Lodz, Poland
- Polish Mothers Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Centre, Baqiyatallah University of Medical Sciences, Tehran, Iran
- Division of Translational Medicine, Baqiyatallah Hospital, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Pulakat L, Sumners C. Angiotensin Type 2 Receptors: Painful, or Not? Front Pharmacol 2020; 11:571994. [PMID: 33424587 PMCID: PMC7785813 DOI: 10.3389/fphar.2020.571994] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Pain in response to various types of acute injury can be a protective stimulus to prevent the organism from using the injured part and allow tissue repair and healing. On the other hand, neuropathic pain, defined as ‘pain caused by a lesion or disease of the somatosensory nervous system’, is a debilitating pathology. The TRPA1 neurons in the Dorsal Root Ganglion (DRG) respond to reactive oxygen species (ROS) and induce pain. In acute nerve injury and inflammation, macrophages infiltrating the site of injury undergo an oxidative burst, and generate ROS that promote tissue repair and induce pain via TRPA1. The latter discourages using the injured limb, with a lack of movement helping wound healing. In chronic inflammation caused by diabetes, cancer etc., ROS levels increase systemically and modulate TRPA1 neuronal functions and cause debilitating neuropathic pain. It is important to distinguish between drug targets that elicit protective vs. debilitating pain when developing effective drugs for neuropathic pain. In this context, the connection of the Angiotensin type 2 receptor (AT2R) to neuropathic pain presents an interesting dilemma. Several lines of evidence show that AT2R activation promotes anti-inflammatory and anti-nociceptive signaling, tissue repair, and suppresses ROS in chronic inflammatory models. Conversely, some studies suggest that AT2R antagonists are anti-nociceptive and therefore AT2R is a drug target for neuropathic pain. However, AT2R expression in nociceptive neurons is lacking, indicating that neuronal AT2R is not involved in neuropathic pain. It is also important to consider that Novartis terminated their phase II clinical trial (EMPHENE) to validate that AT2R antagonist EMA401 mitigates post-herpetic neuralgia. This trial, conducted in Australia, United Kingdom, and a number of European and Asian countries in 2019, was discontinued due to pre-clinical drug toxicity data. Moreover, early data from the trial did not show statistically significant positive outcomes. These facts suggest that may AT2R not be the proper drug target for neuropathic pain in humans and its inhibition can be harmful.
Collapse
Affiliation(s)
- Lakshmi Pulakat
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States.,Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
22
|
Zoja C, Xinaris C, Macconi D. Diabetic Nephropathy: Novel Molecular Mechanisms and Therapeutic Targets. Front Pharmacol 2020; 11:586892. [PMID: 33519447 PMCID: PMC7845653 DOI: 10.3389/fphar.2020.586892] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the major microvascular complications of diabetes mellitus and the leading cause of end-stage kidney disease. The standard treatments for diabetic patients are glucose and blood pressure control, lipid lowering, and renin-angiotensin system blockade; however, these therapeutic approaches can provide only partial renoprotection if started late in the course of the disease. One major limitation in developing efficient therapies for DN is the complex pathobiology of the diabetic kidney, which undergoes a set of profound structural, metabolic and functional changes. Despite these difficulties, experimental models of diabetes have revealed promising therapeutic targets by identifying pathways that modulate key functions of podocytes and glomerular endothelial cells. In this review we will describe recent advances in the field, analyze key molecular pathways that contribute to the pathogenesis of the disease, and discuss how they could be modulated to prevent or reverse DN.
Collapse
Affiliation(s)
- Carlamaria Zoja
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Christodoulos Xinaris
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy.,University of Nicosia Medical School, Nicosia, Cyprus
| | - Daniela Macconi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| |
Collapse
|
23
|
Chaudhary M. Anti-Hypertensive Potential and Epigenetics of Angiotensin II type 2 Receptor (AT2R). Curr Hypertens Rev 2020; 17:176-180. [PMID: 33302839 DOI: 10.2174/1573402116999201209203015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/29/2020] [Accepted: 10/07/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Renin angiotensin system (RAS) is a critical pathway involved in blood pressure regulation. Octapeptide, angiotensin II (Ang II), is a biologically active compound of RAS pathway which mediates its action by binding to either angiotensin II type 1 receptor (AT1R) or angiotensin II type 2 receptor (AT2R). Binding of Ang II to AT1R facilitates blood pressure regulation, whereas AT2R is primarily involved in wound healing and tissue remodeling. OBJECTIVES Recent studies have highlighted the additional role of AT2R to counterbalance the detrimental effects of AT1R. Activation of angiotensin II type 2 receptor using AT2R agonist has shown the effect on natriuresis and release of nitric oxide. Additionally, AT2R activation has been found to inhibit angiotensin converting enzyme (ACE) and enhance angiotensin receptor blocker (ARB) activity. These findings highlight the potential of AT2R as a novel therapeutic target against hypertension. CONCLUSION The potential role of AT2R highlights the importance of exploring additional mechanisms that might be crucial for AT2R expression. Epigenetic mechanisms, including DNA methylation and histone modification, have been explored vastly with relation to cancer, but the role of such mechanisms in the expression of AT2R has recently gained interest.
Collapse
Affiliation(s)
- Mayank Chaudhary
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala-133207, Haryana, India
| |
Collapse
|
24
|
Kaur U, Acharya K, Mondal R, Singh A, Saso L, Chakrabarti S, Chakrabarti SS. Should ACE2 be given a chance in COVID-19 therapeutics: A semi-systematic review of strategies enhancing ACE2. Eur J Pharmacol 2020; 887:173545. [PMID: 32926917 PMCID: PMC7485553 DOI: 10.1016/j.ejphar.2020.173545] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022]
Abstract
The severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) has resulted in almost 28 million cases of COVID-19 (Corona virus disease-2019) and more than 900000 deaths worldwide since December 2019. In the absence of effective antiviral therapy and vaccine, treatment of COVID-19 is largely symptomatic. By making use of its spike (S) protein, the virus binds to its primary human cell receptor, angiotensin converting enzyme 2 (ACE2) which is present in the pulmonary epithelial cells as well as other organs. SARS-CoV-2 may cause a downregulation of ACE2. ACE2 plays a protective role in the pulmonary system through its Mas-receptor and alamandine-MrgD-TGR7 pathways. Loss of this protective effect could be a major component of COVID-19 pathogenesis. An attractive strategy in SARS-CoV-2 therapeutics would be to augment ACE2 either directly by supplementation or indirectly through drugs which increase its levels or stimulate its downstream players. In this semi-systematic review, we have analysed the pathophysiological interplay between ACE and ACE2 in the cardiopulmonary system, the modulation of these two proteins by SARS-CoV-2, and potential therapeutic avenues targeting ACE-Ang II and ACE2-Ang (1-7) axes, that can be utilized against COVID-19 disease progression.
Collapse
Affiliation(s)
- Upinder Kaur
- Department of Pharmacology, All India Institute of Medical Sciences, Gorakhpur, UP, India
| | - Kumudini Acharya
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, UP, India
| | - Ritwick Mondal
- Department of Internal Medicine, Institute of Post Graduate Medical Education and Research, Kolkata, WB, India
| | - Amit Singh
- Department of Pharmacology, Institute of Medical Sciences, Banaras Hindu University, UP, India
| | - Luciano Saso
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar (deemed to be) University, Mullana, Ambala, Haryana, India.
| | | |
Collapse
|
25
|
Scialo F, Daniele A, Amato F, Pastore L, Matera MG, Cazzola M, Castaldo G, Bianco A. ACE2: The Major Cell Entry Receptor for SARS-CoV-2. Lung 2020; 198:867-877. [PMID: 33170317 PMCID: PMC7653219 DOI: 10.1007/s00408-020-00408-4] [Citation(s) in RCA: 294] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023]
Abstract
Despite the unprecedented effort of the scientific community, the novel SARS-CoV-2 virus has infected more than 46 million people worldwide, killing over one million two hundred thousand. Understanding the mechanisms by which some individuals are more susceptible to SARS-CoV-2 infection and why a subgroup of them are prone to experience severe pneumonia, and death should lead to a better approach and more effective treatments for COVID-19. Here, we focus our attention on ACE2, a primary receptor of SARS-CoV-2. We will discuss its biology, tissue expression, and post-translational regulation that determine its potential to be employed by SARS-CoV-2 for cell entry. Particular attention will be given to how the ACE2 soluble form can have a great impact on disease progression and thus be used in a potential therapeutic strategy. Furthermore, we will discuss repercussions that SARS-CoV-2/ACE2 binding has on the renin–angiotensin system and beyond. Indeed, although mostly neglected, ACE2 can also act on [des-Arg 937]-bradykinin of the kinin–kallikrein system regulating coagulation and inflammation. Thorough comprehension of the role that ACE2 plays in different pathways will be the key to assess the impact that SARS-CoV-2/ACE2 binding has on organismal physiology and will help us to find better therapies and diagnostic tools.
Collapse
Affiliation(s)
- Filippo Scialo
- Dipartimento di Scienze Mediche Traslazionali, University of Campania "L. Vanvitelli", Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Aurora Daniele
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, University of Campania "L. Vanvitelli", Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Felice Amato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli Federico II, Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Lucio Pastore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli Federico II, Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Maria Gabriella Matera
- Dipartimento di Medicina Sperimentale, University of Campania "L. Vanvitelli", Naples, Italy
| | - Mario Cazzola
- Dipartimento di Medicina Sperimentale, University of Rome "Tor Vergata", Rome, Italy.
| | - Giuseppe Castaldo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università Di Napoli Federico II, Naples, Italy
- CEINGE, Biotecnologie Avanzate, Naples, Italy
| | - Andrea Bianco
- Dipartimento di Scienze Mediche Traslazionali, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
26
|
Sriram K, Insel PA. A hypothesis for pathobiology and treatment of COVID-19: The centrality of ACE1/ACE2 imbalance. Br J Pharmacol 2020; 177:4825-4844. [PMID: 32333398 PMCID: PMC7572451 DOI: 10.1111/bph.15082] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 11/29/2022] Open
Abstract
Angiotensin Converting Enzyme2 is the cell surface binding site for the coronavirus SARS-CoV-2, which causes COVID-19. We propose that an imbalance in the action of ACE1- and ACE2-derived peptides, thereby enhancing angiotensin II (Ang II) signalling is primary driver of COVID-19 pathobiology. ACE1/ACE2 imbalance occurs due to the binding of SARS-CoV-2 to ACE2, reducing ACE2-mediated conversion of Ang II to Ang peptides that counteract pathophysiological effects of ACE1-generated ANG II. This hypothesis suggests several approaches to treat COVID-19 by restoring ACE1/ACE2 balance: (a) AT receptor antagonists; (b) ACE1 inhibitors (ACEIs); (iii) agonists of receptors activated by ACE2-derived peptides (e.g. Ang (1-7), which activates MAS1); (d) recombinant human ACE2 or ACE2 peptides as decoys for the virus. Reducing ACE1/ACE2 imbalance is predicted to blunt COVID-19-associated morbidity and mortality, especially in vulnerable patients. Importantly, approved AT antagonists and ACEIs can be rapidly repurposed to test their efficacy in treating COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of PharmacologyUniversity of California San DiegoLa JollaCAUSA
| | - Paul A. Insel
- Department of PharmacologyUniversity of California San DiegoLa JollaCAUSA
- Department of MedicineUniversity of California San DiegoLa JollaCAUSA
| |
Collapse
|
27
|
Sriram K, Insel PA. A hypothesis for pathobiology and treatment of COVID-19: The centrality of ACE1/ACE2 imbalance. Br J Pharmacol 2020. [PMID: 32333398 DOI: 10.1111/bph.15082.10.1111/bph.15082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Abstract
Angiotensin Converting Enzyme2 is the cell surface binding site for the coronavirus SARS-CoV-2, which causes COVID-19. We propose that an imbalance in the action of ACE1- and ACE2-derived peptides, thereby enhancing angiotensin II (Ang II) signalling is primary driver of COVID-19 pathobiology. ACE1/ACE2 imbalance occurs due to the binding of SARS-CoV-2 to ACE2, reducing ACE2-mediated conversion of Ang II to Ang peptides that counteract pathophysiological effects of ACE1-generated ANG II. This hypothesis suggests several approaches to treat COVID-19 by restoring ACE1/ACE2 balance: (a) AT receptor antagonists; (b) ACE1 inhibitors (ACEIs); (iii) agonists of receptors activated by ACE2-derived peptides (e.g. Ang (1-7), which activates MAS1); (d) recombinant human ACE2 or ACE2 peptides as decoys for the virus. Reducing ACE1/ACE2 imbalance is predicted to blunt COVID-19-associated morbidity and mortality, especially in vulnerable patients. Importantly, approved AT antagonists and ACEIs can be rapidly repurposed to test their efficacy in treating COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Paul A Insel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
28
|
Bragina ME, Costa-Fraga F, Sturny M, Ebadi B, Ruoccolo RT, Santos RAS, Fraga-Silva RA, Stergiopulos N. Characterization of the Renin-Angiotensin System in Aged Cavernosal Tissue and its Role in Penile Fibrosis. J Sex Med 2020; 17:2129-2140. [PMID: 32943375 DOI: 10.1016/j.jsxm.2020.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 06/24/2020] [Accepted: 08/03/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND The renin-angiotensin system (RAS) plays an important role in erectile function. The RAS contains 2 major axes: one deleterious, composed of ACE-Ang II-AT1 receptor, and another protective, composed of ACE2-Ang-(1-7)-Mas receptor. While aging is a well-known cause for development of male sexual disorders, little is known about local regulation of the RAS in age-related erectile dysfunction (ED). AIM The present study aimed to assess regulation of the RAS in aging-associated ED rat model and evaluate possible options for disease management through pharmacological modulation of the RAS. METHODS Penile tissues were harvested from 3-, 12-, and 24-month-old Wistar rats. Local expression of major RAS components and ED markers was measured by RT-PCR. Protein expression of RAS components was assessed by western blot. Collagen deposition was measured by Sirius Red and immunohistochemical staining. Evaluation of collagen content was also performed in penile sections of Mas-knockout mice by Sirius Red and Masson's trichrome stainings. Finally, the effect of Ang-(1-7) pretreatment on TGF-β-induced myofibroblast activation was studied in primary cavernosal and immortalized fibroblasts. OUTCOMES Experimental results highlighted the essential role of the RAS in modulation of cavernosal fibrosis. RESULTS The present study demonstrates local expression of angiotensinogen mRNA alongside with major RAS components, which suggests local autonomous functioning of the RAS within penile tissue. Gene expression analysis revealed strong positive correlation between ACE-Ang II-AT1 axis with markers for inflammation and fibrosis. While corpus cavernosum from 24-month-old rats was characterized by increased collagen deposition, protein expression of ACE, AT1, and Mas was shown to be upregulated in the penile tissue of this group. At the same time, penile sections from Mas-knockout mice (FVB/N background) were also shown to have increased collagen deposition. Finally, it was demonstrated that Ang-(1-7) treatment of primary cavernosal and immortalized fibroblasts was able to alleviate TGF-β-induced fibroblast-to-myofibroblast transition. CLINICAL TRANSLATION The present study suggests Ang-(1-7) treatment as a possible strategy for pharmacological management of fibrosis-associated ED in aging. STRENGTHS & LIMITATIONS The link between the RAS and penile fibrosis, indicated by a holistic screening of different ED markers, was confirmed by in vivo and in vitro data. However, results, presented in the manuscript, need to be further reinforced by human data. Important to note, the main goal of the study was to characterize RAS regulation in aging condition rather than state any causal relationships. CONCLUSION Present study characterizes RAS regulation in aging-associated ED and indicates its important role in cavernosal fibrosis. Bragina ME, Costa-Fraga F, Sturny M, et al. Characterization of the Renin-Angiotensin System in Aged Cavernosal Tissue and its Role in Penile Fibrosis. J Sex Med 2020;17:2129-2140.
Collapse
Affiliation(s)
- Maiia E Bragina
- Laboratory of Hemodynamics and Cardiovascular Technology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| | - Fabiana Costa-Fraga
- Laboratory of Hemodynamics and Cardiovascular Technology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| | - Mikaël Sturny
- Laboratory of Hemodynamics and Cardiovascular Technology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| | - Babak Ebadi
- Laboratory of Hemodynamics and Cardiovascular Technology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| | - Rafael T Ruoccolo
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Robson A S Santos
- Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo A Fraga-Silva
- Laboratory of Hemodynamics and Cardiovascular Technology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland.
| | - Nikolaos Stergiopulos
- Laboratory of Hemodynamics and Cardiovascular Technology, École polytechnique fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
29
|
Czick M, Shapter C, Shapter R. COVID's Razor: RAS Imbalance, the Common Denominator Across Disparate, Unexpected Aspects of COVID-19. Diabetes Metab Syndr Obes 2020; 13:3169-3192. [PMID: 32982349 PMCID: PMC7495349 DOI: 10.2147/dmso.s265518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/04/2020] [Indexed: 12/25/2022] Open
Abstract
A modern iteration of Occam's Razor posits that "the simplest explanation is usually correct." Coronavirus Disease 2019 involves widespread organ damage and uneven mortality demographics, deemed unexpected from what was originally thought to be "a straightforward respiratory virus." The simplest explanation is that both the expected and unexpected aspects of COVID-19 share a common mechanism. Silent hypoxia, atypical acute respiratory distress syndrome (ARDS), stroke, olfactory loss, myocarditis, and increased mortality rates in the elderly, in men, in African-Americans, and in patients with obesity, diabetes, and cancer-all bear the fingerprints of the renin-angiotensin system (RAS) imbalance, suggesting that RAS is the common culprit. This article examines what RAS is and how it works, then from that baseline, the article presents the evidence suggesting RAS involvement in the disparate manifestations of COVID-19. Understanding the deeper workings of RAS helps one make sense of severe COVID-19. In addition, recognizing the role of RAS imbalance suggests potential routes to mitigate COVID-19 severity.
Collapse
Affiliation(s)
- Maureen Czick
- University of Connecticut, Department of Anesthesia, Farmington, CT, USA
| | | | - Robert Shapter
- Independent Consultant ( Medical Research, Medical Communications, and Medical Education), Hartford, CT, USA
| |
Collapse
|
30
|
Ozhan O, Parlakpinar H, Acet A. Comparison of the effects of losartan, captopril, angiotensin II type 2 receptor agonist compound 21, and MAS receptor agonist AVE 0991 on myocardial ischemia–reperfusion necrosis in rats. Fundam Clin Pharmacol 2020; 35:669-680. [DOI: 10.1111/fcp.12599] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Onural Ozhan
- Department of Pharmacology Medicine School Inonu University Malatya 44280 Turkey
| | - Hakan Parlakpinar
- Department of Pharmacology Medicine School Inonu University Malatya 44280 Turkey
| | - Ahmet Acet
- Department of Pharmacology Medicine School Inonu University Malatya 44280 Turkey
| |
Collapse
|
31
|
Nemoto T, Nakakura T, Kakinuma Y. Elevated blood pressure in high-fat diet-exposed low birthweight rat offspring is most likely caused by elevated glucocorticoid levels due to abnormal pituitary negative feedback. PLoS One 2020; 15:e0238223. [PMID: 32853260 PMCID: PMC7451543 DOI: 10.1371/journal.pone.0238223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/12/2020] [Indexed: 11/19/2022] Open
Abstract
Being delivered as a low birthweight (LBW) infant is a risk factor for elevated blood pressure and future problems with cardiovascular and cerebellar diseases. Although premature babies are reported to have low numbers of nephrons, some unclear questions remain about the mechanisms underlying elevated blood pressure in full-term LBW infants. We previously reported that glucocorticoids increased miR-449a expression, and increased miR-449a expression suppressed Crhr1 expression and caused negative glucocorticoid feedback. Therefore, we conducted this study to clarify the involvement of pituitary miR-449a in the increase in blood pressure caused by higher glucocorticoids in LBW rats. We generated a fetal low-carbohydrate and calorie-restricted model rat (60% of standard chow), and some individuals showed postnatal growth failure caused by growth hormone receptor expression. Using this model, we examined how a high-fat diet (lard-based 45kcal% fat)-induced mismatch between prenatal and postnatal environments could elevate blood pressure after growth. Although LBW rats fed standard chow had slightly higher blood pressure than control rats, their blood pressure was significantly higher than controls when exposed to a high-fat diet. Observation of glomeruli subjected to periodic acid methenamine silver (PAM) staining showed no difference in number or size. Aortic and cardiac angiotensin II receptor expression was altered with compensatory responses. Blood aldosterone levels were not different between control and LBW rats, but blood corticosterone levels were significantly higher in the latter with high-fat diet exposure. Administration of metyrapone, a steroid synthesis inhibitor, reduced blood pressure to levels comparable to controls. We showed that high-fat diet exposure causes impairment of the pituitary glucocorticoid negative feedback via miR-449a. These results clarify that LBW rats have increased blood pressure due to high glucocorticoid levels when they are exposed to a high-fat diet. These findings suggest a new therapeutic target for hypertension of LBW individuals.
Collapse
Affiliation(s)
- Takahiro Nemoto
- Department of Bioregulatory Science (Physiology), Nippon Medical School, Tokyo, Japan
- * E-mail:
| | - Takashi Nakakura
- Department of Anatomy, Graduate School of Medicine, Teikyo University, Tokyo, Japan
| | - Yoshihiko Kakinuma
- Department of Bioregulatory Science (Physiology), Nippon Medical School, Tokyo, Japan
| |
Collapse
|
32
|
Yao YY, Ling EA, Lu D. Microglia mediated neuroinflammation - signaling regulation and therapeutic considerations with special reference to some natural compounds. Histol Histopathol 2020; 35:1229-1250. [PMID: 32662061 DOI: 10.14670/hh-18-239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Neuroinflammation plays a central role in multiple neurodegenerative diseases and neurological disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), cerebral ischemic injury etc. In this connection, microglia, the key players in the central nervous system, mediate the inflammatory response process. In brain injuries, activated microglia can clear the cellular debris and invading pathogens and release neurotrophic factors; however, prolonged microglia activation may cause neuronal death through excessive release of inflammatory mediators. Therefore, it is of paramount importance to understand the underlying molecular mechanisms of microglia activation to design an effective therapeutic strategy to alleviate neuronal injury. Recent studies have shown that some natural compounds and herbal extracts possess anti-inflammatory properties that may suppress microglial activation and ameliorate neuroinflammation and hence are neuroprotective. In this review, we will update some of the common signaling pathways that regulate microglia activation. Among the various signaling pathways, the Notch-1, mitogen-activated protein kinases (MAPKs), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) have been reported to exacerbate microglia mediated neuroinflammation that is implicated in different neuropathological diseases. The search for natural compounds or agents, specifically those derived from natural herbal extracts such as Gastrodin, scutellarin, RG1 etc. has been the focus of many of our recent studies because they have been found to regulate microglia activation. The pharmacological effects of these agents and their potential mechanisms for regulating microglia activation are systematically reviewed here for a fuller understanding of their biochemical action and therapeutic potential for treatment of microglia mediated neuropathological diseases.
Collapse
Affiliation(s)
- Yue-Yi Yao
- Technology Transfer Center, Kunming Medical University, Kunming, China
| | - Eng-Ang Ling
- Department of Anatomy, Young Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Di Lu
- Technology Transfer Center, Kunming Medical University, Kunming, China.
| |
Collapse
|
33
|
Macedo LM, de Ávila RI, Pedrino GR, Colugnati DB, Valadares MC, Lima EM, Borges CL, Kitten GT, Gava E, Castro CH. Effect of angiotensin II and angiotensin-(1-7) on proliferation of stem cells from human dental apical papilla. J Cell Physiol 2020; 236:366-378. [PMID: 32519379 DOI: 10.1002/jcp.29862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
The effects of the renin-angiotensin system (RAS) on stem cells isolated from human dental apical papilla (SCAPs) are completely unknown. Therefore, the aim of this study was to identify RAS components expressed in SCAPs and the effects of angiotensin (Ang) II and Ang-(1-7) on cell proliferation. SCAPs were collected from third molar teeth of adolescents and maintained in cell culture. Messenger RNA expression and protein levels of angiotensin-converting enzyme (ACE), ACE2, and Mas, Ang II type I (AT1) and type II (AT2) receptors were detected in SCAPs. Treatment with either Ang II or Ang-(1-7) increased the proliferation of SCAPs. These effects were inhibited by PD123319, an AT2 antagonist. While Ang II augmented mTOR phosphorylation, Ang-(1-7) induced ERK1/2 phosphorylation. In conclusion, SCAPs produce the main RAS components and both Ang II and Ang-(1-7) treatments induced cell proliferation mediated by AT2 activation through different intracellular mechanisms.
Collapse
Affiliation(s)
- Larissa M Macedo
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Renato I de Ávila
- Laboratory of Education and Research in In Vitro Toxicology (Tox In), Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Gustavo R Pedrino
- Department of Physiological Sciences, Centre for Neuroscience and Cardiovascular Research, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Diego B Colugnati
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Marize C Valadares
- Laboratory of Education and Research in In Vitro Toxicology (Tox In), Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Eliana M Lima
- Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Clayton L Borges
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Gregory T Kitten
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Elisandra Gava
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Carlos H Castro
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| |
Collapse
|
34
|
Sabbatini AR, Kararigas G. Estrogen-related mechanisms in sex differences of hypertension and target organ damage. Biol Sex Differ 2020; 11:31. [PMID: 32487164 PMCID: PMC7268741 DOI: 10.1186/s13293-020-00306-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Hypertension (HTN) is a primary risk factor for cardiovascular (CV) events, target organ damage (TOD), premature death and disability worldwide. The pathophysiology of HTN is complex and influenced by many factors including biological sex. Studies show that the prevalence of HTN is higher among adults aged 60 and over, highlighting the increase of HTN after menopause in women. Estrogen (E2) plays an important role in the development of systemic HTN and TOD, exerting several modulatory effects. The influence of E2 leads to alterations in mechanisms regulating the sympathetic nervous system, renin-angiotensin-aldosterone system, body mass, oxidative stress, endothelial function and salt sensitivity; all associated with a crucial inflammatory state and influenced by genetic factors, ultimately resulting in cardiac, vascular and renal damage in HTN. In the present article, we discuss the role of E2 in mechanisms accounting for the development of HTN and TOD in a sex-specific manner. The identification of targets with therapeutic potential would contribute to the development of more efficient treatments according to individual needs.
Collapse
Affiliation(s)
| | - Georgios Kararigas
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.
| |
Collapse
|
35
|
Cao DY, Saito S, Veiras LC, Okwan-Duodu D, Bernstein EA, Giani JF, Bernstein KE, Khan Z. Role of angiotensin-converting enzyme in myeloid cell immune responses. Cell Mol Biol Lett 2020; 25:31. [PMID: 32508938 PMCID: PMC7249647 DOI: 10.1186/s11658-020-00225-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/05/2020] [Indexed: 02/07/2023] Open
Abstract
Angiotensin-converting enzyme (ACE), a dicarboxypeptidase, plays a major role in the regulation of blood pressure by cleaving angiotensin I into angiotensin II (Ang II), a potent vasoconstrictor. Because of its wide substrate specificity and tissue distribution, ACE affects many diverse biological processes. In inflammatory diseases, including granuloma, atherosclerosis, chronic kidney disease and bacterial infection, ACE expression gets upregulated in immune cells, especially in myeloid cells. With increasing evidences connecting ACE functions to the pathogenesis of these acquired diseases, it is suggested that ACE plays a vital role in immune functions. Recent studies with mouse models of bacterial infection and tumor suggest that ACE plays an important role in the immune responses of myeloid cells. Inhibition of ACE suppresses neutrophil immune response to bacterial infection. In contrast, ACE overexpression in myeloid cells strongly induced bacterial and tumor resistance in mice. A detailed biochemical understanding of how ACE activates myeloid cells and which ACE peptide(s) (substrate or product) mediate these effects could lead to the development of novel therapies for boosting immunity against a variety of stimuli, including bacterial infection and tumor.
Collapse
Affiliation(s)
- Duo-Yao Cao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Suguru Saito
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Luciana C Veiras
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Derick Okwan-Duodu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Ellen A Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Jorge F Giani
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Kenneth E Bernstein
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| | - Zakir Khan
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Davis Res. Bldg., Rm. 2014, 8700 Beverly Blvd, Los Angeles, CA 90048 USA
| |
Collapse
|
36
|
Sharma N, Belenchia AM, Toedebusch R, Pulakat L, Hans CP. AT2R agonist NP-6A4 mitigates aortic stiffness and proteolytic activity in mouse model of aneurysm. J Cell Mol Med 2020; 24:7393-7404. [PMID: 32420690 PMCID: PMC7339180 DOI: 10.1111/jcmm.15342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/16/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical and experimental studies show that angiotensin II (AngII) promotes vascular pathology via activation of AngII type 1 receptors (AT1Rs). We recently reported that NP-6A4, a selective peptide agonist for AngII type 2 receptor (AT2R), exerts protective effects on human vascular cells subjected to serum starvation or doxorubicin exposure. In this study, we investigated whether NP-6A4-induced AT2R activation could mitigate AngII-induced abdominal aortic aneurism (AAA) using AngII-treated Apoe-/- mice. Male Apoe-/- mice were infused with AngII (1 µg/kg/min) by implanting osmotic pumps subcutaneously for 28 days. A subset of mice was pre-treated subcutaneously with NP-6A4 (2.5 mg/kg/day) or vehicle for 14 days prior to AngII, and treatments were continued for 28 days. NP-6A4 significantly reduced aortic stiffness of the abdominal aorta induced by AngII as determined by ultrasound functional analyses and histochemical analyses. NP-6A4 also increased nitric oxide bioavailability in aortic tissues and suppressed AngII-induced increases in monocyte chemotactic protein-1, osteopontin and proteolytic activity of the aorta. However, NP-6A4 did not affect maximal intraluminal aortic diameter or AAA incidences significantly. These data suggest that the effects of AT2R agonist on vascular pathologies are selective, affecting the aortic stiffness and proteolytic activity without affecting the size of AAA.
Collapse
Affiliation(s)
- Neekun Sharma
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Anthony M Belenchia
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Ryan Toedebusch
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Lakshmi Pulakat
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.,Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.,Molecular Cardiology Research Institute, Department of Medicine, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA
| | - Chetan P Hans
- Department of Cardiovascular Medicine, University of Missouri, Columbia, MO, USA.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA.,Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
37
|
|
38
|
D’Ardes D, Boccatonda A, Rossi I, Guagnano MT, Santilli F, Cipollone F, Bucci M. COVID-19 and RAS: Unravelling an Unclear Relationship. Int J Mol Sci 2020; 21:E3003. [PMID: 32344526 PMCID: PMC7215550 DOI: 10.3390/ijms21083003] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/31/2022] Open
Abstract
The renin-angiotensin system (RAS) plays a main role in regulating blood pressure and electrolyte and liquid balance. Previous evidence suggests that RAS may represent an important target for the treatment of lung pathologies, especially for acute respiratory distress syndrome and chronic fibrotic disease. The scientific community has recently focused its attention on angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor 1 (AT1R) inhibitors and their possible benefit/harms for patients infected by Coronavirus disease (COVID-19) who experience pneumonia, but there are still some doubts about the effects of these drugs in this setting.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Santilli
- Clinica Medica Institute, European Center of Excellence on Atherosclerosis, Hypertension and Dyslipidemia, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy (A.B.); (I.R.); (M.T.G.); (F.C.); (M.B.)
| | | | | |
Collapse
|
39
|
Wang C, Pinar AA, Widdop RE, Hossain MA, Bathgate RAD, Denton KM, Kemp-Harper BK, Samuel CS. The anti-fibrotic actions of relaxin are mediated through AT 2 R-associated protein phosphatases via RXFP1-AT 2 R functional crosstalk in human cardiac myofibroblasts. FASEB J 2020; 34:8217-8233. [PMID: 32297670 DOI: 10.1096/fj.201902506r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 04/02/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022]
Abstract
Fibrosis is a hallmark of several cardiovascular diseases. The relaxin family peptide receptor 1 (RXFP1) agonist, relaxin, has rapidly occurring anti-fibrotic actions which are mediated through RXFP1 and angiotensin II receptor crosstalk on renal and cardiac myofibroblasts. Here, we investigated whether this would allow relaxin to indirectly activate angiotensin II type 2 receptor (AT2 R)-specific signal transduction in primary human cardiac myofibroblasts (HCMFs). The anti-fibrotic effects of recombinant human relaxin (RLX; 16.8 nM) or the AT2 R-agonist, Compound 21 (C21; 1 μM), were evaluated in TGF-β1-stimulated HCMFs, in the absence or presence of an RXFP1 antagonist (1 μM) or AT2 R antagonist (0.1 μM) to confirm RXFP1-AT2 R crosstalk. Competition binding for RXFP1 was determined. Western blotting was performed to determine which AT2 R-specific protein phosphatases were expressed by HCMFs; then, the anti-fibrotic effects of RLX and/or C21 were evaluated in the absence or presence of pharmacological inhibition (NSC95397 (1 μM) for MKP-1; okadaic acid (10 nM) for PP2A) or siRNA-knockdown of these phosphatases after 72 hours. The RLX- or C21-induced increase in ERK1/2 and nNOS phosphorylation, and decrease in α-SMA (myofibroblast differentiation) and collagen-I expression by HCMFs was abrogated by pharmacological blockade of RXFP1 or the AT2 R, confirming RXFP1-AT2 R crosstalk in these cells. HCMFs were found to express AT2 R-dependent MKP-1 and PP2A phosphatases, while pharmacological blockade or siRNA-knockdown of either phosphatase also abolished RLX and/or C21 signal transduction in HCMFs (all P < .05 vs RLX or C21 alone). These findings demonstrated that RLX can indirectly activate AT2 R-dependent phosphatase activity in HCMFs by signaling through RXFP1-AT2 R crosstalk, which have important therapeutic implications for its anti-fibrotic actions.
Collapse
Affiliation(s)
- Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Anita A Pinar
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Mohammed A Hossain
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Ross A D Bathgate
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, VIC, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Physiology, Monash University, Clayton, VIC, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia.,Department of Pharmacology, Monash University, Clayton, VIC, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
40
|
Wright JW, Harding JW. Contributions by the Brain Renin-Angiotensin System to Memory, Cognition, and Alzheimer's Disease. J Alzheimers Dis 2020; 67:469-480. [PMID: 30664507 DOI: 10.3233/jad-181035] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive neuron losses in memory-associated brain structures that rob patients of their dignity and quality of life. Five drugs have been approved by the FDA to treat AD but none modify or significantly slow disease progression. New therapies are needed to delay the course of this disease with the ultimate goal of preventing neuron losses and preserving memory functioning. In this review we describe the renin-angiotensin II (AngII) system (RAS) with specific regard to its deleterious contributions to hypertension, facilitation of neuroinflammation and oxidative stress, reduced cerebral blood flow, tissue remodeling, and disruption of memory consolidation and retrieval. There is evidence that components of the RAS, AngIV and Ang(1-7), are positioned to counter such damaging influences and these systems are detailed with the goal of drawing attention to their importance as drug development targets. Ang(1-7) binds at the Mas receptor, while AngIV binds at the AT4 receptor subtype, and these receptor numbers are significantly decreased in AD patients, accompanied by declines in brain aminopeptidases A and N, enzymes essential for the synthesis of AngIV. Potent analogs may be useful to counter these changes and facilitate neuronal functioning and reduce apoptosis in memory associated brain structures of AD patients.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| |
Collapse
|
41
|
Xue F, Yang J, Cheng J, Sui W, Cheng C, Li H, Zhang M, Zhang J, Xu X, Ma J, Lu L, Xu J, Zhang M, Zhang Y, Zhang C. Angiotensin-(1-7) mitigated angiotensin II-induced abdominal aortic aneurysms in apolipoprotein E-knockout mice. Br J Pharmacol 2020; 177:1719-1734. [PMID: 31658493 DOI: 10.1111/bph.14906] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/19/2019] [Accepted: 10/06/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE To test the hypothesis that angiotensin-(1-7) [Ang-(1-7)] may attenuate abdominal aortic aneurysm (AAA) via inhibiting vascular inflammation, extracellular matrix degradation, and smooth muscle cell (SMC) apoptosis, an animal model of AAA was established by angiotensin II (Ang II) infusion to apolipoprotein E-knockout (ApoE-/- ) mice. EXPERIMENTAL APPROACH All mice and cultured SMCs or macrophages were divided into control, Ang II-treated, Ang II + Ang-(1-7)-treated, Ang II + Ang-(1-7) + A779-treated and Ang II + Ang-(1-7) + PD123319-treated groups respectively. In vivo, aortic mechanics and serum lipids were assessed. Ex vivo, AAA were examined by histology, immunohistochemistry and zymography. Cultured cells were analysed by RT-PCR, western blots and TUNEL assays. KEY RESULTS In vivo, Ang-(1-7) reduced the incidence and severity of AAA induced by Ang II infusion, by inhibiting macrophage infiltration, attenuating expression of IL-6, TNF-α, CCL2 and MMP2, and decreasing SMC apoptosis in abdominal aortic tissues. Addition of A779 or PD123319 reversed Ang-(1-7)-mediated beneficial effects on AAA. In vitro, Ang-(1-7) decreased expression of mRNA for IL-6, TNF-α, and CCL2 induced by Ang II in macrophages. In addition, Ang-(1-7) suppressed apoptosis and MMP2 expression and activity in Ang II-treated SMCs. These effects were accompanied by inhibition of the ERK1/2 signalling pathways via Ang-(1-7) stimulation of Mas and AT2 receptors. CONCLUSION AND IMPLICATIONS Ang-(1-7) treatment attenuated Ang II-induced AAA via inhibiting vascular inflammation, extracellular matrix degradation, and SMC apoptosis. Ang-(1-7) may provide a novel and promising approach to the prevention and treatment of AAA.
Collapse
Affiliation(s)
- Fei Xue
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhai Sui
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Cheng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hongxuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jie Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xingli Xu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Ma
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Lu
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jinfeng Xu
- Department of Ultrasound, Shenzhen People's Hospital, Shenzhen, China
| | - Meng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
42
|
Chen TY, Li X, Hung CH, Bahudhanapati H, Tan J, Kass DJ, Zhang Y. The relaxin family peptide receptor 1 (RXFP1): An emerging player in human health and disease. Mol Genet Genomic Med 2020; 8:e1194. [PMID: 32100955 PMCID: PMC7196478 DOI: 10.1002/mgg3.1194] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/11/2020] [Indexed: 12/14/2022] Open
Abstract
Background Relaxin/relaxin family peptide receptor 1 (RXFP1) signaling is important for both normal physiology and disease. Strong preclinical evidence supports relaxin as a potent antifibrotic molecule. However, relaxin‐based therapy failed in clinical trial in patients with systemic sclerosis. We and others have discovered that aberrant expression of RXFP1 may contribute to the abnormal relaxin/RXFP1 signaling in different diseases. Reduced RXFP1 expression and alternative splicing transcripts with potential functional consequences have been observed in fibrotic tissues. A relative decrease in RXFP1 expression in fibrotic tissues—specifically lung and skin—may explain a potential insensitivity to relaxin. In addition, receptor dimerization also plays important roles in relaxin/RXFP1 signaling. Methods This review describes the tissue specific expression, characteristics of the splicing variants, and homo/heterodimerization of RXFP1 in both normal physiological function and human diseases. We discuss the potential implications of these molecular features for developing therapeutics to restore relaxin/RXFP1 signaling and to harness relaxin's potential antifibrotic effects. Results Relaxin/RXFP1 signaling is important in both normal physiology and in human diseases. Reduced expression of RXFP1 in fibrotic lung and skin tissues surrenders both relaxin/RXFP1 signaling and their responsiveness to exogenous relaxin treatments. Alternative splicing and receptor dimerization are also important in regulating relaxin/RXFP1 signaling. Conclusions Understanding the molecular mechanisms that drive aberrant expression of RXFP1 in disease and the functional roles of alternative splicing and receptor dimerization will provide insight into therapeutic targets that may restore the relaxin responsiveness of fibrotic tissues.
Collapse
Affiliation(s)
- Ting-Yun Chen
- Division of Pulmonary, Allergy and Critical Care Medicine and the Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Institute of Allied Health Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Xiaoyun Li
- Division of Pulmonary, Allergy and Critical Care Medicine and the Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ching-Hsia Hung
- Institute of Allied Health Sciences, National Cheng Kung University, Tainan, Taiwan
| | - Harinath Bahudhanapati
- Division of Pulmonary, Allergy and Critical Care Medicine and the Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jiangning Tan
- Division of Pulmonary, Allergy and Critical Care Medicine and the Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel J Kass
- Division of Pulmonary, Allergy and Critical Care Medicine and the Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yingze Zhang
- Division of Pulmonary, Allergy and Critical Care Medicine and the Dorothy P. and Richard P. Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Abstract
The active hormone of the renin-angiotensin system (RAS), angiotensin II (Ang II), is involved in several human diseases, driving the development and clinical use of several therapeutic drugs, mostly angiotensin I converting enzyme (ACE) inhibitors and angiotensin receptor type I (AT1R) antagonists. However, angiotensin peptides can also bind to receptors different from AT1R, in particular, angiotensin receptor type II (AT2R), resulting in biological and physiological effects different, and sometimes antagonistic, of their binding to AT1R. In the present Perspective, the components of the RAS and the therapeutic tools developed to control it will be reviewed. In particular, the characteristics of AT2R and tools to modulate its functions will be discussed. Agonists or antagonists to AT2R are potential therapeutics in cardiovascular diseases, for agonists, and in the control of pain, for antagonists, respectively. However, controlling their binding properties and their targeting to the target tissues must be optimized.
Collapse
Affiliation(s)
- Lucienne Juillerat-Jeanneret
- Transplantation Center, Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Chemin des Boveresses 155, CH1011 Lausanne, Switzerland
| |
Collapse
|
44
|
Wang Y, Han L, Shen M, Jones ES, Spizzo I, Walton SL, Denton KM, Gaspari TA, Samuel CS, Widdop RE. Serelaxin and the AT 2 Receptor Agonist CGP42112 Evoked a Similar, Nonadditive, Cardiac Antifibrotic Effect in High Salt-Fed Mice That Were Refractory to Candesartan Cilexetil. ACS Pharmacol Transl Sci 2020; 3:76-87. [PMID: 32259090 DOI: 10.1021/acsptsci.9b00095] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 12/29/2022]
Abstract
Fibrosis is involved in the majority of cardiovascular diseases and is a key contributor to end-organ dysfunction. In the current study, the antifibrotic effects of recombinant human relaxin-2 (serelaxin; RLX) and/or the AT2R agonist CGP42112 (CGP) were compared with those of the established AT1R antagonist, candesartan cilexetil (CAND), in a high salt-induced cardiac fibrosis model. High salt (HS; 5%) for 8 weeks did not increase systolic blood pressure in male FVB/N mice, but CAND treatment alone significantly reduced systolic blood pressure from HS-induced levels. HS significantly increased cardiac interstitial fibrosis, which was reduced by either RLX and/or CGP, which were not additive under the current experimental conditions, while CAND failed to reduce HS-induced cardiac fibrosis. The antifibrotic effects induced by RLX and/or CGP were associated with reduced myofibroblast differentiation. Additionally, all treatments inhibited the HS-induced elevation in tissue inhibitor of matrix metalloproteinases-1, together with trends for increased MMP-13 expression, that collectively would favor collagen degradation. Furthermore, these antifibrotic effects were associated with reduced cardiac inflammation. Collectively, these results highlight that either RXFP1 or AT2R stimulation represents novel therapeutic strategies to target fibrotic conditions, particularly in HS states that may be refractory to AT1R blockade.
Collapse
Affiliation(s)
- Yan Wang
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Lei Han
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Matthew Shen
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Emma S Jones
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Iresha Spizzo
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Sarah L Walton
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Tracey A Gaspari
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Biomedicine Discovery Institute, Department of Pharmacology, and Department of Physiology, Monash University, Clayton, Victoria 3800 Australia
| |
Collapse
|
45
|
Nigro E, Perrotta F, Polito R, D'Agnano V, Scialò F, Bianco A, Daniele A. Metabolic Perturbations and Severe COVID-19 Disease: Implication of Molecular Pathways. Int J Endocrinol 2020; 2020:8896536. [PMID: 33312199 PMCID: PMC7703458 DOI: 10.1155/2020/8896536] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease (COVID-19) is caused by SARS-CoV-2 virus, which can result in serious respiratory illnesses such as pneumonia leading to respiratory failure. It was first reported in Wuhan, Hubei, China, in December 2019 and rapidly spread globally, becoming a pandemic in March 2020. Among comorbidities observed in SARS-CoV-2 positive patients, hypertension (68.3%) and type 2-diabetes (30.1%) are the most frequent conditions. Although symptoms are highly heterogeneous (ranging from absence of symptoms to severe acute respiratory failure), patients with metabolic-associated diseases often experience worse COVID-19 outcomes. This review investigates the association between metabolic disorders and COVID-19 severity, exploring the molecular mechanisms potentially underlying this relationship and those that are responsible for more severe COVID-19 outcomes. In addition, the role of the main biological processes that may connect metabolic alterations to SARS-CoV-2 infection such as hyperglycemia, immune system deregulation, ACE-2 receptor modulation, and inflammatory response is described. The impact of metabolic disorders on the prognosis of COVID-19 has major implications in public health especially for countries affected by a high incidence of metabolic diseases.
Collapse
Affiliation(s)
- Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università Degli Studi Della Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, Caserta 81100, Italy
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| | - Fabio Perrotta
- Dipartimento di Medicina e Scienze Della Salute “V. Tiberio”, Università Del Molise, Campobasso 86100, Italy
| | - Rita Polito
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| | - Vito D'Agnano
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Filippo Scialò
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Andrea Bianco
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Aurora Daniele
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università Degli Studi Della Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, Caserta 81100, Italy
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| |
Collapse
|
46
|
Chow BSM, Kocan M, Shen M, Wang Y, Han L, Chew JY, Wang C, Bosnyak S, Mirabito-Colafella KM, Barsha G, Wigg B, Johnstone EKM, Hossain MA, Pfleger KDG, Denton KM, Widdop RE, Summers RJ, Bathgate RAD, Hewitson TD, Samuel CS. AT1R-AT2R-RXFP1 Functional Crosstalk in Myofibroblasts: Impact on the Therapeutic Targeting of Renal and Cardiac Fibrosis. J Am Soc Nephrol 2019; 30:2191-2207. [PMID: 31511361 DOI: 10.1681/asn.2019060597] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/29/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recombinant human relaxin-2 (serelaxin), which has organ-protective actions mediated via its cognate G protein-coupled receptor relaxin family peptide receptor 1 (RXFP1), has emerged as a potential agent to treat fibrosis. Studies have shown that serelaxin requires the angiotensin II (AngII) type 2 receptor (AT2R) to ameliorate renal fibrogenesis in vitro and in vivo. Whether its antifibrotic actions are affected by modulation of the AngII type 1 receptor (AT1R), which is expressed on myofibroblasts along with RXFP1 and AT2R, is unknown. METHODS We examined the signal transduction mechanisms of serelaxin when applied to primary rat renal and human cardiac myofibroblasts in vitro, and in three models of renal- or cardiomyopathy-induced fibrosis in vivo. RESULTS The AT1R blockers irbesartan and candesartan abrogated antifibrotic signal transduction of serelaxin via RXFP1 in vitro and in vivo. Candesartan also ameliorated serelaxin's antifibrotic actions in the left ventricle of mice with cardiomyopathy, indicating that candesartan's inhibitory effects were not confined to the kidney. We also demonstrated in a transfected cell system that serelaxin did not directly bind to AT1Rs but that constitutive AT1R-RXFP1 interactions could form. To potentially explain these findings, we also demonstrated that renal and cardiac myofibroblasts expressed all three receptors and that antagonists acting at each receptor directly or allosterically blocked the antifibrotic effects of either serelaxin or an AT2R agonist (compound 21). CONCLUSIONS These findings have significant implications for the concomitant use of RXFP1 or AT2R agonists with AT1R blockers, and suggest that functional interactions between the three receptors on myofibroblasts may represent new targets for controlling fibrosis progression.
Collapse
Affiliation(s)
- Bryna S M Chow
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Martina Kocan
- Florey Institute of Neuroscience and Mental Health.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Matthew Shen
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Yan Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Lei Han
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Jacqueline Y Chew
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Sanja Bosnyak
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Katrina M Mirabito-Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Giannie Barsha
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Belinda Wigg
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Elizabeth K M Johnstone
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | | | - Kevin D G Pfleger
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Pharmacology and Therapeutics, ARC Centre for Personalised Therapeutic Technologies, Melbourne, Australia; and.,Dimerix Limited, Nedlands, Western Australia, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Ross A D Bathgate
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Tim D Hewitson
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Chrishan S Samuel
- Department of Biochemistry and Molecular Biology, and .,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| |
Collapse
|
47
|
Benitez SG, Seltzer AM, Messina DN, Foscolo MR, Patterson SI, Acosta CG. Cutaneous inflammation differentially regulates the expression and function of Angiotensin-II types 1 and 2 receptors in rat primary sensory neurons. J Neurochem 2019; 152:675-696. [PMID: 31386177 DOI: 10.1111/jnc.14848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/25/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022]
Abstract
Neuropathic and inflammatory pain results from cellular and molecular changes in dorsal root ganglion (DRG) neurons. The type-2 receptor for Angiotensin-II (AT2R) has been involved in this type of pain. However, the underlying mechanisms are poorly understood, including the role of the type-1 receptor for Angiotensin-II (AT1R). Here, we used a combination of immunohistochemistry and immunocytochemistry, RT-PCR and in vitro and in vivo pharmacological manipulation to examine how cutaneous inflammation affected the expression of AT1R and AT2R in subpopulations of rat DRG neurons and studied their impact on inflammation-induced neuritogenesis. We demonstrated that AT2R-neurons express C- or A-neuron markers, primarily IB4, trkA, and substance-P. AT1R expression was highest in small neurons and co-localized significantly with AT2R. In vitro, an inflammatory soup caused significant elevation of AT2R mRNA, whereas AT1R mRNA levels remained unchanged. In vivo, we found a unique pattern of change in the expression of AT1R and AT2R after cutaneous inflammation. AT2R increased in small neurons at 1 day and in medium size neurons at 4 days. Interestingly, cutaneous inflammation increased AT1R levels only in large neurons at 4 days. We found that in vitro and in vivo AT1R and AT2R acted co-operatively to regulate DRG neurite outgrowth. In vivo, AT2R inhibition impacted more on non-peptidergic C-neurons neuritogenesis, whereas AT1R blockade affected primarily peptidergic nerve terminals. Thus, cutaneous-induced inflammation regulated AT1R and AT2R expression and function in different DRG neuronal subpopulations at different times. These findings must be considered when targeting AT1R and AT2R to treat chronic inflammatory pain. Cover Image for this issue: doi: 10.1111/jnc.14737.
Collapse
Affiliation(s)
- Sergio G Benitez
- Laboratorio de Neurobiología del Dolor, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Alicia M Seltzer
- Laboratorio de Neurobiología, Instituto de Embriología e Histología (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego N Messina
- Laboratorio de Neurobiología del Dolor, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Mabel R Foscolo
- Laboratorio de Neurobiología del Dolor, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Sean I Patterson
- Departamento de Morfofisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.,Instituto de Histología y Embriología - CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Cristian G Acosta
- Laboratorio de Neurobiología del Dolor, Instituto de Histología y Embriología de Mendoza (IHEM-CONICET), Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
48
|
Renin Activity in Heart Failure with Reduced Systolic Function-New Insights. Int J Mol Sci 2019; 20:ijms20133182. [PMID: 31261774 PMCID: PMC6651297 DOI: 10.3390/ijms20133182] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 12/27/2022] Open
Abstract
Regardless of the cause, symptomatic heart failure (HF) with reduced ejection fraction (rEF) is characterized by pathological activation of the renin–angiotensin–aldosterone system (RAAS) with sodium retention and extracellular fluid expansion (edema). Here, we review the role of active renin, a crucial, upstream enzymatic regulator of the RAAS, as a prognostic and diagnostic plasma biomarker of heart failure with reduced ejection fraction (HFrEF) progression; we also discuss its potential as a pharmacological bio-target in HF therapy. Clinical and experimental studies indicate that plasma renin activity is elevated with symptomatic HFrEF with edema in patients, as well as in companion animals and experimental models of HF. Plasma renin activity levels are also reported to be elevated in patients and animals with rEF before the development of symptomatic HF. Modulation of renin activity in experimental HF significantly reduces edema formation and the progression of systolic dysfunction and improves survival. Thus, specific assessment and targeting of elevated renin activity may enhance diagnostic and therapeutic precision to improve outcomes in appropriate patients with HFrEF.
Collapse
|
49
|
Wen J, Li P, Cheng J, Wang N, Mao L, Tan X, Zeng X, Xia D, Zhou Y, Yang Q, Yang Y. Downregulation of AT 2R decreases the responsiveness of BK Ca channels to angiotensin II in patients with hypertension. J Mol Cell Cardiol 2019; 131:20-28. [PMID: 30998981 DOI: 10.1016/j.yjmcc.2019.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/27/2019] [Accepted: 04/12/2019] [Indexed: 01/23/2023]
Abstract
Angiotensin II (Ang II) modulates blood pressure via Ang II type 1 receptor (AT1R) and type 2 receptor (AT2R). The activation of AT2R relaxes vascular tone through opening large-conductance Ca2+-activated potassium (BKCa) channels in vascular smooth muscle cells (SMCs). In the present study, we studied the role of the AT2R-BKCa pathway in patients with hypertension. The mesenteric arterial SMCs (MSMCs) were obtained from normotensive patients (NP) and hypertensive patients (HP). BKCa currents were recorded with patch clamp and the expressions of mRNAs and proteins of AT1R/AT2R were analyzed by RT-PCR and Western blotting, respectively. Ang II significantly increased the macroscopic BKCa currents at the whole cell level, while increased the open probability and decreased the mean close time of BKCa channels at the single channel level with AT1R blockade by valsartan in NP. However, Ang II had no effect on the BKCa currents at the same condition in HP. Furthermore, the expressions of mRNA and protein of AT2R but not AT1R were markedly decreased in the MSMCs of HP compared to that of NP. The data suggest that AT2R is well functioned in the MSMCs in NP but not in HP and deficiency in the AT2R-BKCa pathway may contribute to the development of hypertension.
Collapse
Affiliation(s)
- Jing Wen
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pengyun Li
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jun Cheng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Na Wang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Liang Mao
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaoqiu Tan
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiaorong Zeng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Dong Xia
- The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yejiang Zhou
- The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qingqiang Yang
- The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
50
|
ACE inhibitor suppresses cardiac remodeling after myocardial infarction by regulating dendritic cells and AT 2 receptor-mediated mechanism in mice. Biomed Pharmacother 2019; 114:108660. [PMID: 30974387 DOI: 10.1016/j.biopha.2019.108660] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 01/10/2023] Open
Abstract
Dendritic cells (DCs) play a complex role in the progression of myocardial infarction (MI). The impact of angiotensin-converting enzyme (ACE) inhibitor therapy, partly via affecting DCs maturation and recruitment, was tested on a MI mouse model. Furthermore, the cardioprotective effects of ACEI were enhanced through attenuating migration of DCs from the spleen into peripheral circulation, thereby inhibiting DCs maturation and tissue inflammation. ACEI repress DCs immune inflammatory response through down-regulating DCs maturation surface markers and regulating inflammatory cytokines, which led to a higher survival rate, improved function and remodeling through decreased inflammatory response after MI. However, inhibition of AT2R activation, resulted in a reduction of ACEI effects on DCs. The potent anti-inflammatory effect of ACEI can partially be attributed to its impact on DCs through activation of AT2R, which may provide a new target mechanism for ACEI therapy after MI.
Collapse
|