1
|
Devasia AG, Shanmugham M, Ramasamy A, Bellanger S, Parry LJ, Leo CH. Therapeutic potential of relaxin or relaxin mimetics in managing cardiovascular complications of diabetes. Biochem Pharmacol 2024; 229:116507. [PMID: 39182735 DOI: 10.1016/j.bcp.2024.116507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Diabetes mellitus is a metabolic disease with an escalating global prevalence. Despite the abundance and relative efficacies of current therapeutic approaches, they primarily focus on attaining the intended glycaemic targets, but patients ultimately still suffer from various diabetes-associated complications such as retinopathy, nephropathy, cardiomyopathy, and atherosclerosis. There is a need to explore innovative and effective diabetic treatment strategies that not only address the condition itself but also combat its complications. One promising option is the reproductive hormone relaxin, an endogenous ligand of the RXFP1 receptor. Relaxin is known to exert beneficial actions on the cardiovascular system through its vasoprotective, anti-inflammatory and anti-fibrotic effects. Nevertheless, the native relaxin peptide exhibits a short biological half-life, limiting its therapeutic potential. Recently, several relaxin mimetics and innovative delivery technologies have been developed to extend its biological half-life and efficacy. The current review provides a comprehensive landscape of the cardiovascular effects of relaxin, focusing on its potential therapeutic applications in managing complications associated with diabetes. The latest advancements in the development of relaxin mimetics and delivery methods for the treatment of cardiometabolic disorders are also discussed.
Collapse
Affiliation(s)
- Arun George Devasia
- Science, Math & Technology, Singapore University of Technology & Design, Singapore 487372, Singapore; Genome Institute of Singapore (GIS), Agency for Science Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Meyammai Shanmugham
- Science, Math & Technology, Singapore University of Technology & Design, Singapore 487372, Singapore; A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Adaikalavan Ramasamy
- Genome Institute of Singapore (GIS), Agency for Science Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Sophie Bellanger
- A*STAR Skin Research Labs (A*SRL), Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #06-06 Immunos, Singapore 138648, Singapore
| | - Laura J Parry
- School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Chen Huei Leo
- Department of Biomedical Engineering, College of Design & Engineering, National University of Singapore, Singapore 117576, Singapore.
| |
Collapse
|
2
|
Romero G, Martin B, Gabris B, Salama G. Relaxin suppresses atrial fibrillation, reverses fibrosis and reduces inflammation in aged hearts. Biochem Pharmacol 2024; 227:116407. [PMID: 38969298 DOI: 10.1016/j.bcp.2024.116407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/18/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Healthy aging results in cardiac structural and electrical remodeling that increase susceptibility to cardiovascular diseases. Relaxin has shown broad cardioprotective effects including anti-fibrotic, anti-arrhythmic and anti-inflammatory outcomes in multiple models. This paper focuses on the cardioprotective effects of Relaxin in a rat model of aging. Sustained atrial or ventricular fibrillation are readily induced in the hearts of aged but not young control animals. Treatment with Relaxin suppressed this arrhythmogenic response by increasing conduction velocity, decreasing fibrosis and promoting substantial cardiac remodeling. Relaxin treatment resulted in a significant increase in the levels of: Nav1.5, Cx43, βcatenin and Wnt1 in rat hearts. In isolated cardiomyocytes, Relaxin increased Nav1.5 expression. These effects were mimicked by CHIR 99021, a pharmacological activator of canonical Wnt signaling, but blocked by the canonical Wnt inhibitor Dickkopf1. Relaxin prevented TGF-β-dependent differentiation of cardiac fibroblasts into myofibroblasts while increasing the expression of Wnt1; the effects of Relaxin on cardiac fibroblast differentiation were blocked by Dickkopf1. RNASeq studies demonstrated reduced expression of pro-inflammatory cytokines and an increase in the expression of α- and β-globin in Relaxin-treated aged males. Relaxin reduces arrhythmogenicity in the hearts of aged rats by reduction of fibrosis and increased conduction velocity. These changes are accompanied by substantial remodeling of the cardiac tissue and appear to be mediated by increased canonical Wnt signaling. Relaxin also exerts significant anti-inflammatory and anti-oxidant effects in the hearts of aged rodents. The mechanisms by which Relaxin increases the expression of Wnt ligands, promotes Wnt signaling and reprograms gene expression remain to be determined.
Collapse
Affiliation(s)
- Guillermo Romero
- Departments of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Brian Martin
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Beth Gabris
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guy Salama
- Departments of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Almeida-Pinto N, Dschietzig TB, Brás-Silva C, Adão R. Cardiovascular effects of relaxin-2: therapeutic potential and future perspectives. Clin Res Cardiol 2024; 113:1137-1150. [PMID: 37721595 PMCID: PMC11269324 DOI: 10.1007/s00392-023-02305-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/04/2023] [Indexed: 09/19/2023]
Abstract
The hormone relaxin-2 has emerged as a promising player in regulating the physiology of the cardiovascular system. Through binding to the relaxin family peptide receptor 1 (RXFP1), this hormone elicits multiple physiological responses including vasodilation induction, reduction of inflammation and oxidative stress, and angiogenesis stimulation. The role of relaxin-2, or its recombinant human form known as serelaxin, has been investigated in preclinical and clinical studies as a potential therapy for cardiovascular diseases, especially heart failure, whose current therapy is still unoptimized. However, evidence from past clinical trials has been inconsistent and further research is needed to fully understand the potential applications of relaxin-2. This review provides an overview of serelaxin use in clinical trials and discusses future directions in the development of relaxin-2 mimetics, which may offer new therapeutic options for patients with heart failure.
Collapse
Affiliation(s)
- Nísia Almeida-Pinto
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
| | | | - Carmen Brás-Silva
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, Porto, Portugal
| | - Rui Adão
- Cardiovascular R&D Centre-UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4200-319, Porto, Portugal.
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
- CIBER Enfermedades Respiratorias (Ciberes), Madrid, Spain.
| |
Collapse
|
4
|
Yuan S, Guo D, Liang X, Zhang L, Zhang Q, Xie D. Relaxin in fibrotic ligament diseases: Its regulatory role and mechanism. Front Cell Dev Biol 2023; 11:1131481. [PMID: 37123405 PMCID: PMC10134402 DOI: 10.3389/fcell.2023.1131481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 02/24/2023] [Indexed: 05/02/2023] Open
Abstract
Fibrotic ligament diseases (FLDs) are diseases caused by the pathological accumulation of periarticular fibrotic tissue, leading to functional disability around joint and poor life quality. Relaxin (RLX) has been reported to be involved in the development of fibrotic lung and liver diseases. Previous studies have shown that RLX can block pro-fibrotic process by reducing the excess extracellular matrix (ECM) formation and accelerating collagen degradation in vitro and in vivo. Recent studies have shown that RLX can attenuate connective tissue fibrosis by suppressing TGF-β/Smads signaling pathways to inhibit the activation of myofibroblasts. However, the specific roles and mechanisms of RLX in FLDs remain unclear. Therefore, in this review, we confirmed the protective effect of RLX in FLDs and summarized its mechanism including cells, key cytokines and signaling pathways involved. In this article, we outline the potential therapeutic role of RLX and look forward to the application of RLX in the clinical translation of FLDs.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Dong Guo
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xinzhi Liang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Luhui Zhang
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Qun Zhang
- Good Clinical Practice Development, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- *Correspondence: Denghui Xie, ; Qun Zhang,
| | - Denghui Xie
- Department of Joint Surgery and Sports Medicine, Center for Orthopedic Surgery, Orthopedic Hospital of Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Academy of Orthopedics, Guangdong Province, Guangzhou, Guangdong, China
- *Correspondence: Denghui Xie, ; Qun Zhang,
| |
Collapse
|
5
|
Xu G, Gong Y, Lu F, Wang B, Yang Z, Chen L, Min J, Cheng C, Jiang T. Endothelin receptor B enhances liver injury and pro-inflammatory responses by increasing G-protein-coupled receptor kinase-2 expression in primary biliary cholangitis. Sci Rep 2022; 12:19772. [PMID: 36396948 PMCID: PMC9672122 DOI: 10.1038/s41598-022-21816-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/04/2022] [Indexed: 11/18/2022] Open
Abstract
Severe diseases like cirrhosis and liver failure can be developed from primary biliary cholangitis (PBC). Endothelin-2 (EDN2) and endothelin receptor B (EDNRB) are related to the pathogenesis of PBC. However, the roles of EDN2 and EDNRB in PBC-related liver injury and inflammation along with molecular mechanisms are poorly defined. In this study, histopathologic alterations of liver tissues were assessed through hematoxylin-eosin staining. Alanine transaminase (ALT), alkaline phosphatase (ALP), aspartate transaminase (AST), and γ-Glutamyltranspetidase (GGT) (4 liver function indexes) serum levels were detected with corresponding activity assay kits. Also, we determined the levels of M2 subtype anti-mitochondrial antibody (AMA-M2), interferon-gamma (IFN-γ), and tumor-necrosis factor alpha (TNFα) in serum with ELISA assay. Later, RT-qPCR assay was used to measure the expression of genes at mRNA levels, while western blotting and immunohistochemical techniques were used to detect protein levels of genes. Our results showed that the liver tissues of PBC patients and mice presented with severe hepatocyte injury and inflammatory cell infiltration as well as destruction of intrahepatic small bile ducts. ALP, AST, ALT, GGT, AMA-M2, IFN-γ, and TNF-α serum levels were higher in PBC patients and mice. Besides, EDN2 and EDNRB were highly expressed in serums and livers of PBC patients and mice. EDNRB potentiated PBC-related liver injury and pro-inflammatory responses, as evidenced by observation of serious liver pathologic injury and increased serum levels of ALP, AST, ALT, AMA-M2, IFN-γ, and TNF-α in PBC mice following EDNRB overexpression. EDNRB overexpression or activation via its agonist IRL-1620 TFA triggered liver injury and pro-inflammatory responses, increased GRK2 expression and induced NF-κB expression and activation in wild-type mice. EDNRB knockdown or inhibition by Bosentan alleviated liver damage and inflammation, reduced GRK2 expression, and inhibited NF-κB in PBC mice. These findings suggested EDNRB loss or inhibition weakened liver injury and pro-inflammatory responses by down-regulating GRK2 and inhibiting the NF-κB pathway in PBC mice.
Collapse
Affiliation(s)
- Guoxin Xu
- Department of Clinical Laboratory, The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600 China
| | - Yanping Gong
- Department of Clinical Immunology, Institution of Laboratory Medicine of Changshu, Changshu, 215500 China
| | - Fenying Lu
- grid.417303.20000 0000 9927 0537Department of Gastroenterology, The Affiliated Changshu Hospital of Xuzhou Medical University, Suzhou, 215501 China
| | - Bin Wang
- grid.417303.20000 0000 9927 0537Department of Gastroenterology, The Affiliated Changshu Hospital of Xuzhou Medical University, Suzhou, 215501 China
| | - Zaixing Yang
- grid.469601.cDepartment of Laboratory Medicine, Huangyan Hospital of Wenzhou Medical University, Taizhou First People’s Hospital, Taizhou, 318020 China
| | - Long Chen
- Department of Clinical Laboratory, The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, 215600 China
| | - Jingyu Min
- grid.417303.20000 0000 9927 0537Department of Gastroenterology, The Affiliated Changshu Hospital of Xuzhou Medical University, Suzhou, 215501 China
| | - Cuie Cheng
- grid.417303.20000 0000 9927 0537Department of Gastroenterology, The Affiliated Changshu Hospital of Xuzhou Medical University, Suzhou, 215501 China
| | - Tingwang Jiang
- grid.417303.20000 0000 9927 0537Department of Key Laboratory, The Affiliated Changshu Hospital of Xuzhou Medical University, Changshu, 215500 China
| |
Collapse
|
6
|
Relaxin does not prevent development of hypoxia-induced pulmonary edema in rats. Pflugers Arch 2022; 474:1053-1067. [PMID: 35778581 PMCID: PMC9492557 DOI: 10.1007/s00424-022-02720-9] [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: 02/17/2022] [Revised: 06/06/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022]
Abstract
Acute hypoxia impairs left ventricular (LV) inotropic function and induces development of pulmonary edema (PE). Enhanced and uneven hypoxic pulmonary vasoconstriction is an important pathogenic factor of hypoxic PE. We hypothesized that the potent vasodilator relaxin might reduce hypoxic pulmonary vasoconstriction and prevent PE formation. Furthermore, as relaxin has shown beneficial effects in acute heart failure, we expected that relaxin might also improve LV inotropic function in hypoxia. Forty-two rats were exposed over 24 h to normoxia or hypoxia (10% N2 in O2). They were infused with either 0.9% NaCl solution (normoxic/hypoxic controls) or relaxin at two doses (15 and 75 μg kg−1 day−1). After 24 h, hemodynamic measurements and bronchoalveolar lavage were performed. Lung tissue was obtained for histological and immunohistochemical analyses. Hypoxic control rats presented significant depression of LV systolic pressure by 19% and of left and right ventricular contractility by about 40%. Relaxin did not prevent the hypoxic decrease in LV inotropic function, but re-increased right ventricular contractility. Moreover, hypoxia induced moderate interstitial PE and inflammation in the lung. Contrasting to our hypothesis, relaxin did not prevent hypoxia-induced pulmonary edema and inflammation. In hypoxic control rats, PE was similarly distributed in the apical and basal lung lobes. In relaxin-treated rats, PE index was 35–40% higher in the apical than in the basal lobe, which is probably due to gravity effects. We suggest that relaxin induced exaggerated vasodilation, and hence pulmonary overperfusion. In conclusion, the results show that relaxin does not prevent but rather may aggravate PE formation.
Collapse
|
7
|
Chunduri P, Patel SA, Levick SP. Relaxin/serelaxin for cardiac dysfunction and heart failure in hypertension. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:183-211. [PMID: 35659372 DOI: 10.1016/bs.apha.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The pregnancy related hormone relaxin is produced throughout the reproductive system. However, relaxin also has important cardiovascular effects as part of the adaptation that the cardiovascular system undergoes in response to the extra demands of pregnancy. These effects are primarily mediated by the relaxin family peptide receptor 1, which is one of four known relaxin receptors. The effects of relaxin on the cardiovascular system during pregnancy, as well as its anti-fibrotic and anti-inflammatory properties, have led to extensive studies into the potential of relaxin therapy as an approach to treat heart failure. Cardiomyocytes, cardiac fibroblasts, and endothelial cells all possess relaxin family peptide receptor 1, allowing for direct effects of therapeutic relaxin on the heart. Many pre-clinical animal studies have demonstrated a beneficial effect of exogenous relaxin on adverse cardiac remodeling including inflammation, fibrosis, cardiomyocyte hypertrophy and apoptosis, as well as effects on cardiac contractile function. Despite this, clinical studies have yielded disappointing results for the synthetic seralaxin, even though seralaxin was well tolerated. This article will provide background on relaxin in the context of normal physiology, as well as the role of relaxin in pregnancy-related adaptations of the cardiovascular system. We will also present evidence from pre-clinical animal studies that demonstrate the potential benefits of relaxin therapy, as well as discussing the results from clinical trials. Finally, we will discuss possible reasons for the failure of these clinical trials as well as steps being taken to potentially improve relaxin therapy for heart failure.
Collapse
Affiliation(s)
- Prasad Chunduri
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Shrey A Patel
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Scott P Levick
- Physiology and Pharmacology, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
8
|
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: 3] [Impact Index Per Article: 1.0] [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
|
9
|
Abstract
Venous disease is a term that broadly covers both venous thromboembolic disease and chronic venous disease. The basic pathophysiology of venous thromboembolism and chronic venous disease differ as venous thromboembolism results from an imbalance of hemostasis and thrombosis while chronic venous disease occurs in the setting of tissue damage because of prolonged venous hypertension. Both diseases are common and account for significant mortality and morbidity, respectively, and collectively make up a large health care burden. Despite both diseases having well-characterized environmental components, it has been known for decades that family history is an important risk factor, implicating a genetic element to a patient's risk. Our understanding of the pathogenesis of these diseases has greatly benefited from an expansion of population genetic studies from pioneering familial studies to large genome-wide association studies; we now have multiple risk loci for each venous disease. In this review, we will highlight the current state of knowledge on the epidemiology and genetics of venous thromboembolism and chronic venous disease and directions for future research.
Collapse
Affiliation(s)
- Richard A. Baylis
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, CA
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle WA 98195, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle WA 98101, USA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle WA 98108, USA
| | - Derek Klarin
- Division of Vascular Surgery, University of Florida College of Medicine, Gainesville, FL
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eri Fukaya
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, CA
| |
Collapse
|
10
|
Martins RC, Pintalhão M, Leite-Moreira A, Castro-Chaves P. Relaxin and the Cardiovascular System: from Basic Science to Clinical Practice. Curr Mol Med 2021; 20:167-184. [PMID: 31642776 DOI: 10.2174/1566524019666191023121607] [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: 02/08/2018] [Revised: 08/07/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022]
Abstract
The peptide hormone relaxin was originally linked to reproductive physiology, where it is believed to mediate systemic and renal hemodynamic adjustments to pregnancy. Recently, its broad range of effects in the cardiovascular system has been the focus of intensive research regarding its implications under pathological conditions and potential therapeutic potential. An understanding of the multitude of cardioprotective actions prompted the study of serelaxin, recombinant human relaxin-2, for the treatment of acute heart failure. Despite early promising results from phase II studies, recently revealed RELAX-AHF-2 outcomes were rather disappointing and the treatment for acute heart failure remains an unmet medical need. This article reviews the physiologic actions of relaxin on the cardiovascular system and its relevance in the pathophysiology of cardiovascular disease. We summarize the most updated clinical data and discuss future directions of serelaxin for the treatment of acute heart failure. This should encourage additional work to determine how can relaxin's beneficial effects be exploited for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Rafael Clara Martins
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Internal Medicine Department, São João Hospital Centre, Porto, Portugal
| | - Mariana Pintalhão
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Internal Medicine Department, São João Hospital Centre, Porto, Portugal
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Cardiothoracic Surgery Department, São João Hospital Centre, Porto, Portugal
| | - Paulo Castro-Chaves
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.,Cardiovascular Research Centre, Porto, Portugal.,Internal Medicine Department, São João Hospital Centre, Porto, Portugal
| |
Collapse
|
11
|
Fournier SB, D'Errico JN, Stapleton PA. Uterine Vascular Control Preconception and During Pregnancy. Compr Physiol 2021; 11:1871-1893. [PMID: 34061977 DOI: 10.1002/cphy.c190015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Successful pregnancy and reproduction are dependent on adequate uterine blood flow, placental perfusion, and vascular responsivity to fetal demands. The ability to support pregnancy centers on systemic adaptation and endometrial preparation through decidualization, embryonic implantation, trophoblast invasion, arterial/arteriolar reactivity, and vascular remodeling. These adaptations occur through responsiveness to endocrine signaling and local uteroplacental mediators. The purpose of this article is to highlight the current knowledge associated with vascular remodeling and responsivity during uterine preparation for and during pregnancy. We focus on maternal cardiovascular systemic and uterine modifications, endometrial decidualization, implantation and invasion, uterine and spiral artery remodeling, local uterine regulatory mechanisms, placentation, and pathological consequences of vascular dysfunction during pregnancy. © 2021 American Physiological Society. Compr Physiol 11:1-23, 2021.
Collapse
Affiliation(s)
- Sara B Fournier
- Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey, USA
| | - Jeanine N D'Errico
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| | - Phoebe A Stapleton
- Environmental and Occupational Health Sciences Institute, Piscataway, New Jersey, USA.,Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
12
|
Ligand-activated RXFP1 gene therapy ameliorates pressure overload-induced cardiac dysfunction. Mol Ther 2021; 29:2499-2513. [PMID: 33839322 DOI: 10.1016/j.ymthe.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 02/10/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022] Open
Abstract
Recurrent episodes of decompensated heart failure (HF) represent an emerging cause of hospitalizations in developed countries with an urgent need for effective therapies. Recently, the pregnancy-related hormone relaxin (RLN) was found to mediate cardio-protective effects and act as a positive inotrope in the cardiovascular system. RLN binds to the RLN family peptide receptor 1 (RXFP1), which is predominantly expressed in atrial cardiomyocytes. We therefore hypothesized that ventricular RXFP1 expression might exert potential therapeutic effects in an in vivo model of cardiac dysfunction. Thus, mice were exposed to pressure overload by transverse aortic constriction and treated with AAV9 to ectopically express RXFP1. To activate RXFP1 signaling, RLN was supplemented subcutaneously. Ventricular RXFP1 expression was well tolerated. Additional RLN administration not only abrogated HF progression but restored left ventricular systolic function. In accordance, upregulation of fetal genes and pathological remodeling markers were significantly reduced. In vitro, RLN stimulation of RXFP1-expressing cardiomyocytes induced downstream signaling, resulting in protein kinase A (PKA)-specific phosphorylation of phospholamban (PLB), which was distinguishable from β-adrenergic activation. PLB phosphorylation corresponded to increased calcium amplitude and contractility. In conclusion, our results demonstrate that ligand-activated cardiac RXFP1 gene therapy represents a therapeutic approach to attenuate HF with the potential to adjust therapy by exogenous RLN supplementation.
Collapse
|
13
|
Sassoli C, Nistri S, Chellini F, Bani D. Human Recombinant Relaxin (Serelaxin) as Anti-fibrotic Agent: Pharmacology, Limitations and Actual Perspectives. Curr Mol Med 2021; 22:196-208. [PMID: 33687895 DOI: 10.2174/1566524021666210309113650] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 11/22/2022]
Abstract
Relaxin (recombinant human relaxin-2 hormone; RLX-2; serelaxin) had raised expectations as a new medication for fibrotic diseases. A plethora of in vitro and in vivo studies have offered convincing demonstrations that relaxin promotes remodelling of connective tissue extracellular matrix mediated by inhibition of multiple fibrogenic pathways, especially the downstream signalling of transforming growth factor (TGF)-β1, a major pro-fibrotic cytokine, and the recruitment and activation of myofibroblast, the main fibrosis-generating cells. However, all clinical trials with relaxin in patients with fibrotic diseases gave inconclusive results. In this review, we have summarized the molecular mechanisms of fibrosis, highlighting those which can be effectively targeted by relaxin. Then, we have performed a critical reappraisal of the clinical trials performed to-date with relaxin as anti-fibrotic drug, in order to highlight their key points of strength and weakness and to identify some future opportunities for the therapeutic use of relaxin, or its analogues, in fibrotic diseases and pathologic scarring which, in our opinion, deserve to be investigated.
Collapse
Affiliation(s)
- Chiara Sassoli
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology, Research Unit of Human Anatomy. Italy
| | - Silvia Nistri
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology, Research Unit of Histology & Embryology, University of Florence, Florence. Italy
| | - Flaminia Chellini
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology, Research Unit of Human Anatomy. Italy
| | - Daniele Bani
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology, Research Unit of Histology & Embryology, University of Florence, Florence. Italy
| |
Collapse
|
14
|
Searching for Preclinical Models of Acute Decompensated Heart Failure: a Concise Narrative Overview and a Novel Swine Model. Cardiovasc Drugs Ther 2020; 36:727-738. [PMID: 33098053 PMCID: PMC9270312 DOI: 10.1007/s10557-020-07096-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/09/2020] [Indexed: 11/25/2022]
Abstract
Purpose Available animal models of acute heart failure (AHF) and their limitations are discussed herein. A novel and preclinically relevant porcine model of decompensated AHF (ADHF) is then presented. Methods Myocardial infarction (MI) was induced by occlusion of left anterior descending coronary artery in 17 male pigs (34 ± 4 kg). Two weeks later, ADHF was induced in the survived animals (n = 15) by occlusion of the circumflex coronary artery, associated with acute volume overload and increases in arterial blood pressure by vasoconstrictor infusion. After onset of ADHF, animals received 48-h iv infusion of either serelaxin (n = 9) or placebo (n = 6). The pathophysiology and progression of ADHF were described by combining evaluation of hemodynamics, echocardiography, bioimpedance, blood gasses, circulating biomarkers, and histology. Results During ADHF, animals showed reduced left ventricle (LV) ejection fraction < 30%, increased thoracic fluid content > 35%, pulmonary edema, and high pulmonary capillary wedge pressure ~ 30 mmHg (p < 0.01 vs. baseline). Other ADHF-induced alterations in hemodynamics, i.e., increased central venous and pulmonary arterial pressures; respiratory gas exchanges, i.e., respiratory acidosis with low arterial PO2 and high PCO2; and LV dysfunction, i.e., increased LV end-diastolic/systolic volumes, were observed (p < 0.01 vs. baseline). Representative increases in circulating cardiac biomarkers, i.e., troponin T, natriuretic peptide, and bio-adrenomedullin, occurred (p < 0.01 vs. baseline). Finally, elevated renal and liver biomarkers were observed 48 h after onset of ADHF. Mortality was ~ 50%. Serelaxin showed beneficial effects on congestion, but none on mortality. Conclusion This new model, resulting from a combination of chronic and acute MI, and volume and pressure overload, was able to reproduce all the typical clinical signs occurring during ADHF in a consistent and reproducible manner. Electronic supplementary material The online version of this article (10.1007/s10557-020-07096-5) contains supplementary material, which is available to authorized users.
Collapse
|
15
|
Cabiati M, Botta L, Caselli C, Del Ry S. Transcriptional evaluation of relaxin and endothelin-1 axis in heart failure patients: First evidence of its involvement during left ventricular assist device support. Int J Cardiol 2020; 306:109-115. [PMID: 32143920 DOI: 10.1016/j.ijcard.2020.02.058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/18/2020] [Accepted: 02/23/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Left ventricular assist devices (LVAD) are implanted in patients with end-stage heart failure (ESHF) as a mechanical support for the failing myocardium, which is characterized by an activation of the neuro-hormonal system, with release of vasoactive mediators, such as endothelin (ET)-1 and relaxin (RLX)-2. The aim of this study was to evaluate whether LVAD is able to modulate the RLX-2 and ET-1 system expression in ESHF patients. METHODS Cardiac tissue was collected from ESHF patients before LVAD implantation (pre-LVAD group, n = 22), at the time of cardiac transplantation with concomitant LVAD removal (post-LVAD group, n = 6), and from stable HF patients on medical therapy at the time of cardiac transplantation (HTx group, n = 7). The expression of RLX-2, ET-1 system and inflammatory markers (IL-8, IL-6, TNF-α) were evaluated by Real-Time PCR. RESULTS RLX-2 mRNA resulted similar in pre-LVAD and HTx, but it was significantly increased in post-LVAD (p = 0.02/p = 0.01 respectively). A similar trend was observed for ET-1 and ET-converting enzyme (ECE)-1 while no significant difference was observed for ET-receptors. A positive correlation was found between ET-1 and ET-A (p = 0.031) and ECE-1 (p < 0.0001). The inflammatory markers resulted activated in all the three groups. A significant correlation between RLX-2 and ET-1 in pre-LVAD, as well as between RLX-2 and IL-8/IL-6, was found. CONCLUSIONS Our research investigates for the first time the involvement of RLX-2 and ET-1 system in ESHF patients supported by LVAD, demonstrating their potential ability to partially recover the failing myocardium, indicating their possible clinical role as biomarkers or pharmacological agents in LVAD patients. TRANSLATIONAL ASPECT The study of novel biomarkers in patients supported by continuous axial flow devices may be a starting point analysis applicable to patients with centrifugal flow devices.
Collapse
Affiliation(s)
| | - Luca Botta
- Department of Cardiac Surgery, Niguarda Ca' Granda Hospital, Milan, Italy; Department of Cardiac Surgery, AOU Bologna, S. Orsola Hospital, Italy
| | | | | |
Collapse
|
16
|
|
17
|
Valkovic AL, Bathgate RA, Samuel CS, Kocan M. Understanding relaxin signalling at the cellular level. Mol Cell Endocrinol 2019; 487:24-33. [PMID: 30592984 DOI: 10.1016/j.mce.2018.12.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/19/2018] [Accepted: 12/22/2018] [Indexed: 02/07/2023]
Abstract
The peptide hormone relaxin mediates many biological actions including anti-fibrotic, vasodilatory, angiogenic, anti-inflammatory, anti-apoptotic, and organ protective effects across a range of tissues. At the cellular level, relaxin binds to the G protein-coupled receptor relaxin family peptide receptor 1 (RXFP1) to activate a variety of downstream signal transduction pathways. This signalling cascade is complex and also varies in diverse cellular backgrounds. Moreover, RXFP1 signalling shows crosstalk with other receptors to mediate some of its physiological functions. This review summarises known signalling pathways induced by acute versus chronic treatment with relaxin across a range of cell types, it describes RXFP1 crosstalk with other receptors, signalling pathways activated by other ligands targeting RXFP1, and it also outlines physiological relevance of RXFP1 signalling outputs. Comprehensive understanding of the mechanism of relaxin actions in fibrosis, vasodilation, as well as organ protection, will further support relaxin's clinical potential.
Collapse
Affiliation(s)
- Adam L Valkovic
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Ross Ad Bathgate
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, 3052, Australia.
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, 3800, Australia
| | - Martina Kocan
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
18
|
Dschietzig TB. Relaxin-2 for heart failure with preserved ejection fraction (HFpEF): Rationale for future clinical trials. Mol Cell Endocrinol 2019; 487:54-58. [PMID: 30659842 DOI: 10.1016/j.mce.2019.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/11/2019] [Accepted: 01/14/2019] [Indexed: 01/06/2023]
Abstract
Heart Failure with preserved Ejection Fraction (HFpEF), a distinct sub-entity of chronic heart failure characterized by generalized inflammatory non-compliance of the cardio-vascular system, is associated with high mortality and still an unmet medical need. Many novel and promising therapeutic approaches have failed in large studies. This review focuses on basic research, pre-clinical and clinical findings that may account for the potential benefit of relaxin-2 in HFpEF. The peptide combines short-term hemodynamic advantages, such as moderate blood pressure decline and functional endothelin-1 antagonism, with a wealth of protective effects harboring long-term benefits, such as anti-inflammatory, anti-fibrotic, and anti-oxidative actions. These pleiotropic effects are exerted through a complex and intricate signaling cascade involving the relaxin-family peptide receptor-1, the glucocorticoid receptor, the nitric oxide system, and a cell type-dependent variety of down-stream mediators.
Collapse
Affiliation(s)
- Thomas Bernd Dschietzig
- Relaxera Pharmazeutische Gesellschaft mbH & Co. KG, Stubenwald-Allee 8a, 64625, Bensheim, Germany.
| |
Collapse
|
19
|
Martin B, Romero G, Salama G. Cardioprotective actions of relaxin. Mol Cell Endocrinol 2019; 487:45-53. [PMID: 30625345 DOI: 10.1016/j.mce.2018.12.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/20/2018] [Accepted: 12/22/2018] [Indexed: 01/19/2023]
Abstract
Relaxin is a hormone of pregnancy first discovered for its ability to induce ligament relaxation in nonpregnant guinea pig and is important for softening of the birth canal during parturition, decidualization, implantation, nipple development and increased maternal renal perfusion, glomerular filtration, and cardiac output. Subsequently, relaxin has been shown to exert multiple beneficial cardiovascular effects during pathological events such as hypertension, atrial fibrillation, heart failure and myocardial infarction, including suppression of arrhythmia and inflammation, and reversal of fibrosis. Despite extensive studies, the mechanisms underlying relaxin's effects are not well understood. Relaxin signals primarily through its G protein coupled receptor, the relaxin family peptide receptor-1, to activate multiple signaling pathways and this review summarizes our understanding of these pathways as they relate to the cardioprotective actions of relaxin, focusing on relaxin's anti-fibrotic, anti-arrhythmic and anti-inflammatory properties. Further, this review includes a brief overview of relaxin in clinical trials for heart failure and progress in the development of relaxin mimetics.
Collapse
Affiliation(s)
- Brian Martin
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Guillermo Romero
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Guy Salama
- Department of Medicine, Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
20
|
Meadows KL. Experimental models of focal and multifocal cerebral ischemia: a review. Rev Neurosci 2018; 29:661-674. [PMID: 29397392 DOI: 10.1515/revneuro-2017-0076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 11/26/2017] [Indexed: 02/07/2023]
Abstract
Rodent and rabbit stroke models have been instrumental in our current understanding of stroke pathophysiology; however, translational failure is a significant problem in preclinical ischemic stroke research today. There are a number of different focal cerebral ischemia models that vary in their utility, pathophysiology of causing disease, and their response to treatments. Unfortunately, despite active preclinical research using these models, treatment options for ischemic stroke have not significantly advanced since the food and drug administration approval of tissue plasminogen activator in 1996. This review aims to summarize current stroke therapies, the preclinical experimental models used to help develop stroke therapies, as well as their advantages and limitations. In addition, this review discusses the potential for naturally occurring canine ischemic stroke models to compliment current preclinical models and to help bridge the translational gap between small mammal models and human clinical trials.
Collapse
Affiliation(s)
- Kristy L Meadows
- Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Road, Grafton, MA 01536, USA
| |
Collapse
|
21
|
Lian X, Beer-Hammer S, König GM, Kostenis E, Nürnberg B, Gollasch M. RXFP1 Receptor Activation by Relaxin-2 Induces Vascular Relaxation in Mice via a Gα i2-Protein/PI3Kß/γ/Nitric Oxide-Coupled Pathway. Front Physiol 2018; 9:1234. [PMID: 30233409 PMCID: PMC6131674 DOI: 10.3389/fphys.2018.01234] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 08/15/2018] [Indexed: 01/10/2023] Open
Abstract
Background: Relaxins are small peptide hormones, which are novel candidate molecules that play important roles in cardiometablic syndrome. Relaxins are structurally related to the insulin hormone superfamily, which provide vasodilatory effects by activation of G-protein-coupled relaxin receptors (RXFPs) and stimulation of endogenous nitric oxide (NO) generation. Recently, relaxin could be demonstrated to activate Gi proteins and phosphoinositide 3-kinase (PI3K) pathways in cultured endothelial cells in vitro. However, the contribution of the Gi-PI3K pathway and their individual components in relaxin-dependent relaxation of intact arteries remains elusive. Methods: We used Gαi2- (Gnai2-/-) and Gαi3-deficient (Gnai3-/-) mice, pharmacological tools and wire myography to study G-protein-coupled signaling pathways involved in relaxation of mouse isolated mesenteric arteries by relaxins. Human relaxin-1, relaxin-2, and relaxin-3 were tested. Results: Relaxin-2 (∼50% relaxation at 10-11 M) was the most potent vasodilatory relaxin in mouse mesenteric arteries, compared to relaxin-1 and relaxin-3. The vasodilatory effects of relaxin-2 were inhibited by removal of the endothelium or treatment of the vessels with N (G)-nitro-L-arginine methyl ester (L-NAME, endothelial nitric oxide synthase (eNOS) inhibitor) or simazine (RXFP1 inhibitor). The vasodilatory effects of relaxin-2 were absent in arteries of mice treated with pertussis toxin (PTX). They were also absent in arteries isolated from Gnai2-/- mice, but not from Gnai3-/- mice. The effects were not affected by FR900359 (Gαq protein inhibitor) or PI-103 (PI3Kα inhibitor), but inhibited by TGX-221 (PI3Kβ inhibitor) or AS-252424 (PI3Kγ inhibitor). Simazine did not influence the anti-contractile effect of perivascular adipose tissue. Conclusion: Our data indicate that relaxin-2 produces endothelium- and NO-dependent relaxation of mouse mesenteric arteries by activation of RXFP1 coupled to Gi2-PI3K-eNOS pathway. Targeting vasodilatory Gi-protein-coupled RXFP1 pathways may provide promising opportunities for drug discovery in endothelial dysfunction and cardiometabolic disease.
Collapse
Affiliation(s)
- Xiaoming Lian
- Experimental and Clinical Research Center (ECRC), Charité - University Medicine Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Evi Kostenis
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), Charité - University Medicine Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Berlin, Germany
| |
Collapse
|
22
|
Tahmasebi F, Hurrell A, Ford A, Gupta M, Geindreau D, Pimenta D, O’Mahony C. Coronary artery dissection in the puerperium: A case report and literature review. Obstet Med 2018; 11:144-147. [PMID: 30214482 PMCID: PMC6134346 DOI: 10.1177/1753495x17740063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 09/27/2017] [Indexed: 11/16/2022] Open
Abstract
Spontaneous coronary artery dissection is a rare but important cause of acute coronary syndrome. Its relevance for women during the puerperium mandates awareness and understanding amongst obstetric healthcare professionals. The aetiology of the increased risk in pregnancy has not been fully elucidated, but include medial eosinophilic angitis, pregnancy-induced degeneration of collagen in conjunction with the stresses of parturition, and rupture of the vasa vasorum. The risk of mortality necessitates prompt diagnosis, usually by angiography. There is no one-size-fits-all treatment; management must be individualised according to haemodynamic status and affected vessel(s) and includes conservative management, percutaneous coronary intervention, or bypass grafting. Recovery complications include extension of the haematoma or false lumen, valvular pathology secondary to ischaemia, and sudden cardiac death. Close post-operative surveillance is mandatory. We present a 41-year-old lady with post-partum spontaneous coronary artery dissection, complicated by ischaemic papillary rupture and mitral regurgitation requiring valve replacement. Additionally, we present a literature review, including guidance on management and critical analysis of potential complications.
Collapse
Affiliation(s)
- Farshad Tahmasebi
- Obstetrics and Gynaecology,
Whipps
Cross University Hospital, London, UK
| | - Alice Hurrell
- Obstetrics and Gynaecology,
Whipps
Cross University Hospital, London, UK
| | - Amie Ford
- Bart's and The London School of Medicine
and Dentistry, London, UK
| | - Manish Gupta
- Obstetrics and Gynaecology,
Whipps
Cross University Hospital, London, UK
| | | | | | | |
Collapse
|
23
|
Jelinic M, Marshall SA, Leo CH, Parry LJ, Tare M. From pregnancy to cardiovascular disease: Lessons from relaxin-deficient animals to understand relaxin actions in the vascular system. Microcirculation 2018; 26:e12464. [DOI: 10.1111/micc.12464] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/30/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Maria Jelinic
- School of BioSciences; University of Melbourne; Parkville VIC Australia
- Department of Physiology, Anatomy & Microbiology; La Trobe University; Bundoora VIC Australia
| | - Sarah A. Marshall
- School of BioSciences; University of Melbourne; Parkville VIC Australia
- Department of Obstetrics and Gynaecology; School of Clinical Sciences; Monash University; Clayton VIC Australia
| | - Chen H. Leo
- School of BioSciences; University of Melbourne; Parkville VIC Australia
- Science and Maths Cluster; Singapore University of Technology & Design; Singapore Singapore
| | - Laura J. Parry
- School of BioSciences; University of Melbourne; Parkville VIC Australia
| | - Marianne Tare
- Department of Physiology; Monash University; Melbourne VIC Australia
- Monash Rural Health; Monash University; Melbourne VIC Australia
| |
Collapse
|
24
|
Maruyama S, Wu CL, Yoshida S, Zhang D, Li PH, Wu F, Parker Duffen J, Yao R, Jardin B, Adham IM, Law R, Berger J, Di Marchi R, Walsh K. Relaxin Family Member Insulin-Like Peptide 6 Ameliorates Cardiac Fibrosis and Prevents Cardiac Remodeling in Murine Heart Failure Models. J Am Heart Assoc 2018; 7:e008441. [PMID: 29887522 PMCID: PMC6220528 DOI: 10.1161/jaha.117.008441] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/25/2018] [Indexed: 11/21/2022]
Abstract
BACKGROUND The insulin/insulin-like growth factor/relaxin family represents a group of structurally related but functionally diverse proteins. The family member relaxin-2 has been evaluated in clinical trials for its efficacy in the treatment of acute heart failure. In this study, we assessed the role of insulin-like peptide 6 (INSL6), another member of this protein family, in murine heart failure models using genetic loss-of-function and protein delivery methods. METHODS AND RESULTS Insl6-deficient and wild-type (C57BL/6N) mice were administered angiotensin II or isoproterenol via continuous infusion with an osmotic pump or via intraperitoneal injection once a day, respectively, for 2 weeks. In both models, Insl6-knockout mice exhibited greater cardiac systolic dysfunction and left ventricular dilatation. Cardiac dysfunction in the Insl6-knockout mice was associated with more extensive cardiac fibrosis and greater expression of fibrosis-associated genes. The continuous infusion of chemically synthesized INSL6 significantly attenuated left ventricular systolic dysfunction and cardiac fibrosis induced by isoproterenol infusion. Gene expression profiling suggests liver X receptor/retinoid X receptor signaling is activated in the isoproterenol-challenged hearts treated with INSL6 protein. CONCLUSIONS Endogenous Insl6 protein inhibits cardiac systolic dysfunction and cardiac fibrosis in angiotensin II- and isoproterenol-induced cardiac stress models. The administration of recombinant INSL6 protein could have utility for the treatment of heart failure and cardiac fibrosis.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Fibrosis
- Heart Failure/metabolism
- Heart Failure/pathology
- Heart Failure/physiopathology
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Intercellular Signaling Peptides and Proteins
- Intracellular Signaling Peptides and Proteins/deficiency
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Liver X Receptors/genetics
- Liver X Receptors/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/metabolism
- Myocardium/pathology
- Retinoid X Receptors/genetics
- Retinoid X Receptors/metabolism
- Signal Transduction
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Sonomi Maruyama
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Chia-Ling Wu
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Sumiko Yoshida
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Dongying Zhang
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Pei-Hsuan Li
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Fangzhou Wu
- Department of Chemistry, Indiana University, Bloomington, IN
| | - Jennifer Parker Duffen
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Rouan Yao
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Blake Jardin
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
| | - Ibrahim M Adham
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Ronald Law
- New Frontier Science, Takeda Pharmaceuticals International Co, Cambridge, MA
| | - Joel Berger
- New Frontier Science, Takeda Pharmaceuticals International Co, Cambridge, MA
| | | | - Kenneth Walsh
- Molecular Cardiology, Whitaker Cardiovascular Institute Boston University School of Medicine, Boston, MA
- Center for Hematovascular Biology, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA
| |
Collapse
|
25
|
Schiffner R, Nistor M, Bischoff SJ, Matziolis G, Schmidt M, Lehmann T. Effects of human relaxin-2 (serelaxin) on hypoxic pulmonary vasoconstriction during acute hypoxia in a sheep model. HYPOXIA (AUCKLAND, N.Z.) 2018; 6:11-22. [PMID: 29862306 PMCID: PMC5968803 DOI: 10.2147/hp.s165092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Purpose Hypoxia induces pulmonary vasoconstriction with a subsequent increase of pulmonary artery pressure (PAP), which can result in pulmonary hypertension. Serelaxin has shown an increase of pulmonary hemodynamic parameters after serelaxin injection. We therefore investigated the response of pulmonary hemodynamic parameters after serelaxin administration in a clinically relevant model. Methods Six controls and six sheep that received 30 μg/kg serelaxin underwent right heart catheterization during a 12-minute hypoxia period (inhalation of 5% oxygen and 95% nitrogen) and subsequent reoxygenation. Systolic, diastolic, and mean values of both PAP (respectively, PAPs, PAPd, and PAPm) and pulmonary capillary wedge pressure (respectively, PCWPs, PCWPd, and PCWPm), blood gases, heart rate (HR), and both peripheral and pulmonary arterial oxygen saturation were obtained. Cardiac output (CO), stroke volume (SV), pulmonary vascular resistance (PVR), pulmonary arterial compliance (PAcompl), and systemic vascular resistance (SVR) were calculated. Results The key findings of the current study are that serelaxin prevents the rise of PAPs (p≤0.001), PAPm, PCWPm, PCWPs (p≤0.03), and PAPd (p≤0.05) during hypoxia, while it simultaneously increases CO and SV (p≤0.001). Similar courses of decreases of PAPm, PAPd, PAPs, CO, SVR (p≤0.001), and PCWPd (p≤0.03) as compared to hypoxic values were observed during reoxygenation. In direct comparison, the experimental groups differed during hypoxia in regard to HR, PAPm, PVR, and SVR (p≤0.03), and during reoxygenation in regard to HR (p≤0.001), PAPm, PAPs, PAPd, PVR, SVR (p≤0.03), and PCWPd (p≤0.05). Conclusion The findings of this study suggest that serelaxin treatment improves pulmonary hemodynamic parameters during acute hypoxia.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas Lehmann
- Institute of Medical Statistics, Computer Sciences and Documentation Science, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
26
|
Neurohormonal targets in the treatment of pediatric heart failure. PROGRESS IN PEDIATRIC CARDIOLOGY 2018. [DOI: 10.1016/j.ppedcard.2017.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
27
|
Meadows KL. Ischemic stroke and select adipose-derived and sex hormones: a review. Hormones (Athens) 2018; 17:167-182. [PMID: 29876798 DOI: 10.1007/s42000-018-0034-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/27/2018] [Indexed: 02/03/2023]
Abstract
Ischemic stroke is the fifth leading cause of death in the USA and is the leading cause of serious, long-term disability worldwide. The principle sex hormones (estrogen, progesterone, and testosterone), both endogenous and exogenous, have profound effects on various stroke outcomes and have become the focus of a number of studies evaluating risk factors and treatment options for ischemic stroke. In addition, the expression of other hormones that may influence stroke outcome, including select adipose-derived hormones (adiponectin, leptin, and ghrelin), can be regulated by sex hormones and are also the focus of several ischemic stroke studies. This review aims to summarize some of the preclinical and clinical studies investigating the principle sex hormones, as well as select adipose-derived hormones, as risk factors or potential treatments for ischemic stroke. In addition, the potential for relaxin, a lesser studied sex hormone, as a novel treatment option for ischemic stroke is explored.
Collapse
Affiliation(s)
- Kristy L Meadows
- Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Rd., North Grafton, MA, 01536, USA.
| |
Collapse
|
28
|
Jelinic M, Leo CH, Marshall SA, Senadheera SN, Parry LJ, Tare M. Short-term (48 hours) intravenous serelaxin infusion has no effect on myogenic tone or vascular remodeling in rat mesenteric arteries. Microcirculation 2018; 24. [PMID: 28370794 DOI: 10.1111/micc.12371] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/24/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Short-term IV sRLX (recombinant human relaxin-2) infusion enhances endothelium-dependent relaxation in mesenteric arteries. This is initially underpinned by increased NO followed by a transition to prostacyclin. The effects of short-term IV sRLX treatment on pressure-induced myogenic tone and vascular remodeling in these arteries are unknown. Therefore, we investigated the effects of sRLX infusion on pressure-induced myogenic tone and passive mechanical wall properties in mesenteric arteries. METHODS Mesenteric artery myogenic tone and passive mechanics were examined after 48-hours and 10-days infusion of sRLX. Potential mechanisms of action were assessed by pressure myography, qPCR, and Western blot analysis. RESULTS Neither 48-hours nor 10-days sRLX treatment had significant effects on myogenic tone, passive arterial wall stiffness, volume compliance, or axial lengthening. However, in 48-hours sRLX -treated rats, incubation with the NO synthase blocker L-NAME significantly increased myogenic tone (P<.05 vs placebo), demonstrating an increased contribution of NO to the regulation of myogenic tone. eNOS dimerization, but not phosphorylation, was significantly upregulated in the arteries of sRLX -treated rats. CONCLUSION In mesenteric arteries, 48-hours sRLX treatment upregulates the role of NO in the regulation of myogenic tone by enhancing eNOS dimerization, without altering overall myogenic tone or vascular remodeling.
Collapse
Affiliation(s)
- Maria Jelinic
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Chen Huei Leo
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Sarah A Marshall
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Laura J Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Marianne Tare
- Department of Physiology, Monash University, Clayton, Victoria, Australia.,Monash Rural Health, Monash University, Churchill, Victoria, Australia
| |
Collapse
|
29
|
Ng HH, Leo CH, Parry LJ, Ritchie RH. Relaxin as a Therapeutic Target for the Cardiovascular Complications of Diabetes. Front Pharmacol 2018; 9:501. [PMID: 29867503 PMCID: PMC5962677 DOI: 10.3389/fphar.2018.00501] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular complications are the major cause of mortality in patients with diabetes. This is closely associated with both macrovascular and microvascular complications of diabetes, which lead to organ injuries in diabetic patients. Previous studies have consistently demonstrated the beneficial effects of relaxin treatment for protection of the vasculature, with evidence of antioxidant and anti-remodeling actions. Relaxin enhances nitric oxide, prostacyclin and endothelium-derived hyperpolarization (EDH)-type-mediated relaxation in various vascular beds. These effects of relaxin on the systemic vasculature, coupled with its cardiac actions, reduce pulmonary capillary wedge pressure and pulmonary artery pressure. This results in an overall decrease in systemic and pulmonary vascular resistance in heart failure patients. The anti-fibrotic actions of relaxin are well established, a desirable property in the context of diabetes. Further, relaxin ameliorates diabetic wound healing, with accelerated angiogenesis and vasculogenesis. Relaxin-mediated stimulation of vascular endothelial growth factor (VEGF) and stromal cell-derived factor 1-α, as well as regulation of metalloproteinase expression, ameliorates cardiovascular fibrosis in diabetic mice. In the heart, relaxin is a cardioprotective molecule in several experimental animal models, exerting anti-fibrotic, anti-hypertrophy and anti-apoptotic effects in diabetic pathologies. Collectively, these studies provide a foundation to propose the therapeutic potential for relaxin as an adjunctive agent in the prevention or treatment of diabetes-induced cardiovascular complications. This review provides a comprehensive overview of the beneficial effects of relaxin, and identifies its therapeutic possibilities for alleviating diabetes-related cardiovascular injury.
Collapse
Affiliation(s)
- Hooi Hooi Ng
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Human and Molecular Genetics, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Chen Huei Leo
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
- Science and Math Cluster, Singapore University of Technology and Design, Singapore, Singapore
| | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Rebecca H. Ritchie
- Heart Failure Pharmacology, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Pharmacology & Therapeutics, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW In spite of advances in our understanding of acute heart failure (AHF) and its different phenotypic expressions, AHF management is still centered on volume removal with intravenous diuretics. This narrative review describes the pathophysiology underlying hypertensive AHF and appraises therapies targeting these mechanisms. RECENT FINDINGS Vascular redistribution rather than volume overload may be the primary determinant of elevated cardiac filling pressures and subsequent pulmonary congestion in patients with hypertensive AHF; in these patients, vasodilators should be the predominant treatment. Additional therapy with diuretics in hypertensive AHF should be relegated to the treatment of overt volume overload or persistent congestion in spite of optimized hemodynamics. Intravenous nitroglycerin at high doses can rapidly achieve pulmonary decongestion and reduce downstream critical care needs in these patients. The therapeutic role for synthetic peptides with vasodilator properties has yet to be defined. Evidence supporting both old and new vasodilator therapies is limited by a paucity of well-designed studies and failure to demonstrate improvement in long-term outcomes. Targeted study of this phenotype of AHF is needed before vasodilator therapies become incorporated into treatment guidelines.
Collapse
|
31
|
Preeclampsia and the brain: neural control of cardiovascular changes during pregnancy and neurological outcomes of preeclampsia. Clin Sci (Lond) 2017; 130:1417-34. [PMID: 27389588 DOI: 10.1042/cs20160108] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 05/13/2016] [Indexed: 02/07/2023]
Abstract
Preeclampsia (PE) is a form of gestational hypertension that complicates ∼5% of pregnancies worldwide. Over 70% of the fatal cases of PE are attributed to cerebral oedema, intracranial haemorrhage and eclampsia. The aetiology of PE originates from abnormal remodelling of the maternal spiral arteries, creating an ischaemic placenta that releases factors that drive the pathophysiology. An initial neurological outcome of PE is the absence of the autonomically regulated cardiovascular adaptations to pregnancy. PE patients exhibit sympathetic overactivation, in comparison with both normotensive pregnant and hypertensive non-pregnant females. Moreover, PE diminishes baroreceptor reflex sensitivity (BRS) beyond that observed in healthy pregnancy. The absence of the cardiovascular adaptations to pregnancy, combined with sympathovagal imbalance and a blunted BRS leads to life-threatening neurological outcomes. Behaviourally, the increased incidences of maternal depression, anxiety and post-traumatic stress disorder (PTSD) in PE are correlated to low fetal birth weight, intrauterine growth restriction (IUGR) and premature birth. This review addresses these neurological consequences of PE that present in the gravid female both during and after the index pregnancy.
Collapse
|
32
|
Schiffner R, Reiche J, Schmidt M, Jung C, Walther S, Irintchev A, Bischoff SJ. Pulmonary arterial compliance and pulmonary hemodynamic effects of Serelaxin in a sheep model. Clin Hemorheol Microcirc 2017; 66:219-229. [DOI: 10.3233/ch-170269] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- René Schiffner
- Department of Orthopaedic, Jena University Hospital, Friedrich Schiller University, Jena, Germany
- Department of Neurology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Juliane Reiche
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology and Vascular Medicine, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sebastian Walther
- Department of Orthopaedic, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Andrey Irintchev
- Department of Otorhinolaryngology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Sabine J. Bischoff
- Institute for Laboratory Animal Science and Welfare, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
33
|
Herrero-Puente P, Prieto-García B, García-García M, Llorens P, Martín-Sánchez FJ, Jacob J, Mebazaa A, Mueller C, Maisel AS, Gil V, García-Hernandez P, Miró Ò. The relationship of circulating relaxin-2 concentrations with short-term prognosis in patients with acute heart failure: the RELAHF study. Eur J Heart Fail 2017; 19:1205-1209. [PMID: 28580623 DOI: 10.1002/ejhf.875] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/09/2017] [Accepted: 04/03/2017] [Indexed: 12/28/2022] Open
Affiliation(s)
- Pablo Herrero-Puente
- Emergency Department, Central University Hospital of Asturias, University of Oviedo, Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Belén Prieto-García
- Clinical Biochemistry Service, Laboratory of Medicine, Central University Hospital of Asturias, Oviedo, Spain.,Biochemistry and Molecular Biology, University of Oviedo, Spain
| | - María García-García
- Clinical Biochemistry Service, Laboratory of Medicine, Central University Hospital of Asturias, Oviedo, Spain
| | - Pere Llorens
- Emergency Service, Short Stay Unit and Home Care Unit, General University Hospital of Alicante, Alicante, Spain
| | | | - Javier Jacob
- Emergency Service, Bellvitge University Hospital, L'Hospitalet de Llobregat, Spain
| | - Alexandre Mebazaa
- Department of Anaesthesiology and Critical Care Medicine, Hospital Lariboisière, Paris Diderot University, Paris, France.,GREAT (Global REsearch on Acute Conditions Team), Network, Rome, Italy
| | - Christian Mueller
- GREAT (Global REsearch on Acute Conditions Team), Network, Rome, Italy.,Department of Cardiology and Cardiovascular Research Institute Basel, University Hospital Basel, Basel, Switzerland
| | - Alan S Maisel
- GREAT (Global REsearch on Acute Conditions Team), Network, Rome, Italy.,Coronary Care Unit and Heart Failure Program, Veterans Affairs (VA) San Diego, San Diego, CA, USA
| | - Víctor Gil
- Group for Investigation of Emergency Processes and Pathologies (Grupo de Investigación Urgencias: Procesos y Patologías), August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Pablo García-Hernandez
- Clinical Biochemistry Service, Laboratory of Medicine, Central University Hospital of Asturias, Oviedo, Spain
| | - Òscar Miró
- GREAT (Global REsearch on Acute Conditions Team), Network, Rome, Italy.,Group for Investigation of Emergency Processes and Pathologies (Grupo de Investigación Urgencias: Procesos y Patologías), August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.,Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Unemori E. Serelaxin in clinical development: past, present and future. Br J Pharmacol 2017; 174:921-932. [PMID: 28009437 DOI: 10.1111/bph.13695] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/18/2016] [Accepted: 12/05/2016] [Indexed: 12/15/2022] Open
Abstract
The availability of highly purified recombinant human relaxin, serelaxin, has allowed clinical trials to be conducted in several indications and the elucidation of its pharmacology in human subjects. These studies have demonstrated that serelaxin has unique haemodynamic properties that are likely to contribute to organ protection and long-term outcome benefits in acute heart failure. Clinical observations support its consideration for therapeutic use in other patient populations, including those with chronic heart failure, coronary artery disease, portal hypertension and acute renal failure. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.
Collapse
|
35
|
Chen YT, Wang J, Tong KS, Wong LL, Liew OW, Richards AM. The association of heart failure-related microRNAs with neurohormonal signaling. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2031-2040. [PMID: 28065846 DOI: 10.1016/j.bbadis.2016.12.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 01/05/2023]
Abstract
Heart failure (HF) is a widely prevalent syndrome imposing a significant burden of morbidity and mortality world-wide. Differential circulating microRNA profiles observed in HF cohorts suggest the diagnostic utility of microRNAs as biomarkers. Given their function in fine tuning gene expression, alternations in microRNA landscape could reflecting the underlying mechanisms of disease and present potential therapeutic targets. Using multiple computational target predicting algorithms together with the luciferase-based reporting platform, the interactions between HF-related microRNAs and the 3' untranslated regions (3'UTRs) of neurohormone associated genes were examined and compared. Our results indicate that although in silico prediction provides an overview of possible microRNA-mRNA target pairs, less than half of the predicted interactions were experimentally confirmed by reporter assays in HeLa cells. Thus, the establishment of microRNA/3'UTR reporters is essential to systemically evaluate the roles of microRNAs for signaling cascades of interest, including cardiovascular neurohormonal signaling. The physiological relevance of HF-related microRNAs on the expression of putative gene targets was further established by using gain-of-function assays in two human cardiac-derived cells. Our findings, for the first time, provide direct evidence of the regulatory effects of HF-related microRNAs on the neurohormonal signaling in cardiac cells. More importantly, our study presents a rational approach to further exploring microRNA profiling data in deciphering the role of microRNA in complex syndromes such as HF. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Yei-Tsung Chen
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, #08-01, MD6 Centre for Translational Medicine, 14 Medical Drive, 117599, Singapore.
| | - Juan Wang
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, #08-01, MD6 Centre for Translational Medicine, 14 Medical Drive, 117599, Singapore.
| | - Kai Sing Tong
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, #08-01, MD6 Centre for Translational Medicine, 14 Medical Drive, 117599, Singapore.
| | - Lee Lee Wong
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, #08-01, MD6 Centre for Translational Medicine, 14 Medical Drive, 117599, Singapore.
| | - Oi Wah Liew
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, #08-01, MD6 Centre for Translational Medicine, 14 Medical Drive, 117599, Singapore.
| | - Arthur Mark Richards
- Cardiovascular Research Institute, Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, #08-01, MD6 Centre for Translational Medicine, 14 Medical Drive, 117599, Singapore; Cardiac Department, National University Health System, Tower Block Level 9, 1E Kent Ridge Road, 119228, Singapore; Christchurch Heart Institute, Department of Medicine, University of Otago, PO Box 4345, Christchurch 8014, New Zealand.
| |
Collapse
|
36
|
Ghosh RK, Banerjee K, Tummala R, Ball S, Ravakhah K, Gupta A. Serelaxin in acute heart failure: Most recent update on clinical and preclinical evidence. Cardiovasc Ther 2016; 35:55-63. [DOI: 10.1111/1755-5922.12231] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Raktim Kumar Ghosh
- Department of Cardiovascular Medicine; St. Vincent Charity Medical Center; A Teaching Hospital affiliated to Case Western Reserve University; Cleveland OH USA
| | | | - Ramyashree Tummala
- Department of Internal Medicine; St. Vincent Charity Medical Center; A Teaching Hospital affiliated to Case Western Reserve University; Cleveland OH USA
| | - Somedeb Ball
- Department of Internal Medicine; St Francis Hospital and Medical Center; Hartford CT USA
| | - Keyvan Ravakhah
- Department of Internal Medicine; St. Vincent Charity Medical Center; A Teaching Hospital affiliated to Case Western Reserve University; Cleveland OH USA
| | - Anjan Gupta
- Department of Cardiovascular Medicine; St. Vincent Charity Medical Center; A Teaching Hospital affiliated to Case Western Reserve University; Cleveland OH USA
| |
Collapse
|
37
|
Leo CH, Jelinic M, Ng HH, Marshall SA, Novak J, Tare M, Conrad KP, Parry LJ. Vascular actions of relaxin: nitric oxide and beyond. Br J Pharmacol 2016; 174:1002-1014. [PMID: 27590257 DOI: 10.1111/bph.13614] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/24/2016] [Accepted: 08/29/2016] [Indexed: 12/28/2022] Open
Abstract
The peptide hormone relaxin regulates the essential maternal haemodynamic adaptations in early pregnancy through direct actions on the renal and systemic vasculature. These vascular actions of relaxin occur mainly through endothelium-derived NO-mediated vasodilator pathways and improvements in arterial compliance in small resistance-size arteries. This work catalysed a plethora of studies which revealed quite heterogeneous responses across the different regions of the vasculature, and also uncovered NO-independent mechanisms of relaxin action. In this review, we first describe the role of endogenous relaxin in maintaining normal vascular function, largely referring to work in pregnant and male relaxin-deficient animals. We then discuss the diversity of mechanisms mediating relaxin action in different vascular beds, including the involvement of prostanoids, VEGF, endothelium-derived hyperpolarisation and antioxidant activity in addition to the classic NO-mediated vasodilatory pathway. We conclude the review with current perspectives on the vascular remodelling capabilities of relaxin. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.
Collapse
Affiliation(s)
- C H Leo
- School of BioSciences, The University of Melbourne, Parkville, VIC, Australia
| | - M Jelinic
- School of BioSciences, The University of Melbourne, Parkville, VIC, Australia
| | - H H Ng
- School of BioSciences, The University of Melbourne, Parkville, VIC, Australia
| | - S A Marshall
- School of BioSciences, The University of Melbourne, Parkville, VIC, Australia
| | - J Novak
- Division of Mathematics and Science, Walsh University, North Canton, OH, USA
| | - M Tare
- Department of Physiology, Monash University, Clayton, VIC, Australia.,School of Rural Health, Monash University, Clayton, VIC, Australia
| | - K P Conrad
- Department of Physiology and Functional Genomics, Department of Obstetrics and Gynaecology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - L J Parry
- School of BioSciences, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
38
|
Sarwar M, Du XJ, Dschietzig TB, Summers RJ. The actions of relaxin on the human cardiovascular system. Br J Pharmacol 2016; 174:933-949. [PMID: 27239943 DOI: 10.1111/bph.13523] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/01/2016] [Accepted: 04/26/2016] [Indexed: 12/14/2022] Open
Abstract
The insulin-like peptide relaxin, originally identified as a hormone of pregnancy, is now known to exert a range of pleiotropic effects including vasodilatory, anti-fibrotic, angiogenic, anti-apoptotic and anti-inflammatory effects in both males and females. Relaxin produces these effects by binding to a cognate receptor RXFP1 and activating a variety of signalling pathways including cAMP, cGMP and MAPKs as well as by altering gene expression of TGF-β, MMPs, angiogenic growth factors and endothelin receptors. The peptide has been shown to be effective in halting or reversing many of the adverse effects including fibrosis in animal models of cardiovascular disease including ischaemia/reperfusion injury, myocardial infarction, hypertensive heart disease and cardiomyopathy. Relaxin given to humans is safe and produces favourable haemodynamic changes. Serelaxin, the recombinant form of relaxin, is now in extended phase III clinical trials for the treatment of acute heart failure. Previous clinical studies indicated that a 48 h infusion of relaxin improved 180 day mortality, yet the mechanism underlying this effect is not clear. This article provides an overview of the cellular mechanism of effects of relaxin and summarizes its beneficial actions in animal models and in the clinic. We also hypothesize potential mechanisms for the clinical efficacy of relaxin, identify current knowledge gaps and suggest new ways in which relaxin could be useful therapeutically. LINKED ARTICLES This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.
Collapse
Affiliation(s)
- Mohsin Sarwar
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Thomas B Dschietzig
- Immundiagnostik AG, Bensheim, Germany.,Campus Mitte, Medical Clinic for Cardiology and Angiology, Charité-University Medicine Berlin, Berlin, Germany.,Relaxera Pharmazeutische Gesellschaft mbH & Co. KG, Bensheim, Germany
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Australia
| |
Collapse
|
39
|
Conrad KP. G-Protein-coupled receptors as potential drug candidates in preeclampsia: targeting the relaxin/insulin-like family peptide receptor 1 for treatment and prevention. Hum Reprod Update 2016; 22:647-64. [PMID: 27385360 DOI: 10.1093/humupd/dmw021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/16/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Important roles for G-protein-coupled receptors (GPCRs) have been identified in the maternal physiological adaptations to pregnancy and in the pathogenesis of preeclampsia. On this basis, GPCRs are potential therapeutic targets for preeclampsia. OBJECTIVES AND RATIONALE In this review, vasopressin and apelin are initially considered in this context before the focus on the hormone relaxin and its cognate receptor, the relaxin/insulin-like family peptide receptor 1 (RXFP1). Based on both compelling scientific rationale and a promising safety profile, the relaxin ligand-receptor system is comprehensively evaluated as a potential therapeutic endpoint in preeclampsia. SEARCH METHODS The published literature relating to the topic was searched through January 2016 using PubMed. OUTCOMES Relaxin is a peptide hormone secreted by the corpus luteum; it circulates in the luteal phase and during pregnancy. Activation of RXFP1 is vasodilatory; thus, relaxin supplementation is expected to at least partly restore the fundamental vasodilatory changes of normal pregnancy, thereby alleviating maternal organ hypoperfusion, which is a major pathogenic manifestation of severe preeclampsia. Specifically, by exploiting its pleiotropic hemodynamic attributes in preeclampsia, relaxin administration is predicted to (i) reverse robust arterial myogenic constriction; (ii) blunt systemic and renal vasoconstriction in response to activation of the angiotensin II receptor, type 1; (iii) mollify the action of endogenous vasoconstrictors on uterine spiral arteries with failed remodeling and retained smooth muscle; (iv) increase arterial compliance; (v) enhance insulin-mediated glucose disposal by promoting skeletal muscle vasodilation and (vi) mobilize and activate bone marrow-derived angiogenic progenitor cells, thereby repairing injured endothelium and improving maternal vascularity in organs such as breast, uterus, pancreas, skin and fat. By exploiting its pleiotropic molecular attributes in preeclampsia, relaxin supplementation is expected to (i) enhance endothelial nitric oxide synthesis and bioactivity, as well as directly reduce vascular smooth muscle cytosolic calcium, thus promoting vasodilation; (ii) improve the local angiogenic balance by augmenting arterial vascular endothelial and placental growth factor (VEGF and PLGF) activities; (iii) ameliorate vascular inflammation; (iv) enhance placental peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (PCG1α) expression, and hence, peroxisome proliferator-activated receptor gamma (PPAR-γ) activity and (v) confer cytotrophoblast and endothelial cytoprotection. Insofar as impaired endometrial maturation (decidualization) predisposes to the development of preeclampsia, relaxin administration in the late secretory phase and during early pregnancy would be anticipated to improve decidualization, and hence trophoblast invasion and spiral artery remodeling, thereby reducing the risk of preeclampsia. Relaxin has a favorable safety profile both in the non-pregnant condition and during pregnancy. WIDER IMPLICATIONS There is a strong scientific rationale for RXFP1 activation in severe preeclampsia by administration of relaxin, relaxin analogs or small molecule mimetics, in order to mollify the disease pathogenesis for safe prolongation of pregnancy, thus allowing time for more complete fetal maturation, which is a primary therapeutic endpoint in treating the disease. In light of recent data implicating deficient or defective decidualization as a potential etiological factor in preeclampsia and the capacity of relaxin to promote endometrial maturation, the prophylactic application of relaxin to reduce the risk of preeclampsia is a plausible therapeutic approach to consider. Finally, given its pleiotropic and beneficial attributes particularly in the cardiovascular system, relaxin, although traditionally considered as a 'pregnancy' hormone, is likely to prove salutary for several disease indications in the non-pregnant population.
Collapse
Affiliation(s)
- Kirk P Conrad
- Department of Physiology and Functional Genomics and Department of Obstetrics and Gynecology, D.H. Barron Reproductive and Perinatal Biology Research Program, University of Florida, 1600 SW Archer Road, PO Box 100274 M522, Gainesville, FL 32610, USA
| |
Collapse
|
40
|
|
41
|
Anti-atherosclerotic effects of serelaxin in apolipoprotein E-deficient mice. Atherosclerosis 2016; 251:430-437. [PMID: 27341752 DOI: 10.1016/j.atherosclerosis.2016.06.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Serelaxin (SLX) is a recombinant form of human relaxin-2, a naturally occurring peptide that regulates maternal cardiovascular adaptations to pregnancy. It is unclear whether SLX has a therapeutic effect on atherosclerosis. Therefore, we investigated direct vascular effects of SLX in a mouse model of atherosclerosis. METHODS 6-8 week-old female apolipoprotein E-deficient mice were fed a high-fat, cholesterol-rich diet for 6 weeks and additionally received a continuous treatment with vehicle or SLX (0.05 or 0.1 μg/h), during the last 4 weeks, via subcutaneously implanted osmotic mini-pumps. Vascular oxidative stress, vasorelaxation and atherosclerotic plaque development were assessed. RESULTS Vascular oxidative stress was reduced in SLX-treated mice (vehicle: 322.67 RLU/s, SLX 0.05 μg/h: 119.76 RLU/s (p < 0.001 vs. vehicle), SLX 0.1 μg/h: 109.33 RLU/s (p < 0.001 vs. vehicle; p = 0.967 vs. 0.05 μg/h SLX)). Further SLX improved endothelium-dependent vasodilatation without influencing endothelium-independent vasorelaxation. Atherosclerotic plaque development was significantly reduced by SLX (vehicle: 0.38 ± 0.02 mm(2), 0.05 μg/h SLX: 0.32 ± 0.02 mm(2) (p = 0.047 vs. vehicle), 0.1 μg/h SLX: 0.29 ± 0.02 mm(2) (p = 0.002 vs. vehicle; p = 0.490 vs. 0.05 μg/h SLX)). Neither vascular macrophage, T-cell or neutrophil infiltration, nor collagen/vascular smooth muscle cell content differed between the groups. We observed a significant down-regulation of the angiotensin II type 1a receptor and a decrease in IL-6 and an increase in IL-10 plasma concentrations. CONCLUSIONS Our data demonstrates novel pleiotropic effects of SLX on vascular oxidative stress, endothelial dysfunction and atherosclerotic plaque burden. Therefore, SLX could serve as a new drug for the treatment of atherosclerosis-related diseases.
Collapse
|
42
|
Antifibrotic Actions of Serelaxin – New Roles for an Old Player. Trends Pharmacol Sci 2016; 37:485-497. [DOI: 10.1016/j.tips.2016.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/16/2016] [Accepted: 02/19/2016] [Indexed: 12/25/2022]
|
43
|
Shuai XX, Meng YD, Lu YX, Su GH, Tao XF, Han J, Xu SD, Luo P. Relaxin-2 improves diastolic function of pressure-overloaded rats via phospholamban by activating Akt. Int J Cardiol 2016; 218:305-311. [PMID: 27240156 DOI: 10.1016/j.ijcard.2016.05.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/18/2016] [Accepted: 05/12/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Relaxin is a peptide hormone which has been demonstrated to be safe and has a therapeutic effect on acute heart failure in clinic trials. However, its effect on diastolic function is still unknown. The aims of the study were to determine whether relaxin could improve the diastolic function in pressure-overloaded rat model and to analyze potential mechanisms. METHODS AND RESULTS In the present study, a pressure-overloaded rat model induced by transaortic constriction (TAC) was established. Four weeks after TAC, echocardiography was performed and then all the rat models were randomly divided into 3 groups: models without intramyocardial injection (TAC), with intramyocardial injection of empty adenoviral vector (TAC+GFP) and adenoviral vector overexpression relaxin-2 gene (TAC+RLN2). A sham group was also included. Twelve days after intramyocardial injection, echocardiography and hemodynamics were carried out to evaluate diastolic function in sham, TAC, TAC+GFP and TAC+RLN2 groups. Then hearts were harvested for subsequent examinations. The results indicated that relaxin-2 had ameliorated diastolic function in the pressure-overloaded rats. Compared with the TAC and TAC+GFP groups, the relaxin-2 gene transfer increased phosphorylation of Akt at both the Ser473 and Thr308 sites. Meanwhile, it increased the Ser16 and Thr17- phosphorylation levels of phospholamban (PLB). Furthermore, SERCA2 activity was enhanced in the TAC+RLN2 group more than in the TAC and TAC+GFP groups. CONCLUSIONS These results demonstrated that relaxin-2 gene therapy improved diastolic function in pressure-overloaded rats. The potential mechanism may be that relaxin-2 gene transfer enhances SERCA2 activity in hearts by increasing phospholamban phosphorylation through nuclear-targeted Akt phosphorylation.
Collapse
Affiliation(s)
- Xin-Xin Shuai
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yi-di Meng
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yong-Xin Lu
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Guan-Hua Su
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiao-Fang Tao
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jun Han
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Su-Dan Xu
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ping Luo
- Department of Cardiology, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
44
|
Pini A, Boccalini G, Baccari MC, Becatti M, Garella R, Fiorillo C, Calosi L, Bani D, Nistri S. Protection from cigarette smoke-induced vascular injury by recombinant human relaxin-2 (serelaxin). J Cell Mol Med 2016; 20:891-902. [PMID: 26915460 PMCID: PMC4831370 DOI: 10.1111/jcmm.12802] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/02/2016] [Indexed: 12/14/2022] Open
Abstract
Smoking is regarded as a major risk factor for the development of cardiovascular diseases (CVD). This study investigates whether serelaxin (RLX, recombinant human relaxin-2) endowed with promising therapeutic properties in CVD, can be credited of a protective effect against cigarette smoke (CS)-induced vascular damage and dysfunction. Guinea pigs exposed daily to CS for 8 weeks were treated with vehicle or RLX, delivered by osmotic pumps at daily doses of 1 or 10 μg. Controls were non-smoking animals. Other studies were performed on primary guinea pig aortic endothelial (GPAE) cells, challenged with CS extracts (CSE) in the absence and presence of 100 ng/ml (17 nmol/l) RLX. In aortic specimens from CS-exposed guinea pigs, both the contractile and the relaxant responses to phenylephrine and acetylcholine, respectively, were significantly reduced in amplitude and delayed, in keeping with the observed adverse remodelling of the aortic wall, endothelial injury and endothelial nitric oxide synthase (eNOS) down-regulation. RLX at both doses maintained the aortic contractile and relaxant responses to a control-like pattern and counteracted aortic wall remodelling and endothelial derangement. The experiments with GPAE cells showed that CSE significantly decreased cell viability and eNOS expression and promoted apoptosis by sparkling oxygen free radical-related cytotoxicity, while RLX counterbalanced the adverse effects of CSE. These findings demonstrate that RLX is capable of counteracting CS-mediated vascular damage and dysfunction by reducing oxidative stress, thus adding a tile to the growing mosaic of the beneficial effects of RLX in CVD.
Collapse
Affiliation(s)
- Alessandro Pini
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology & Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | - Giulia Boccalini
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology & Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | | | - Matteo Becatti
- Department of Experimental & Clinical Biomedical Sciences 'Mario Serio', Section of Biochemistry, University of Florence, Florence, Italy
| | - Rachele Garella
- Section of Physiology, University of Florence, Florence, Italy
| | - Claudia Fiorillo
- Department of Experimental & Clinical Biomedical Sciences 'Mario Serio', Section of Biochemistry, University of Florence, Florence, Italy
| | - Laura Calosi
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology & Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | - Daniele Bani
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology & Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| | - Silvia Nistri
- Department of Experimental & Clinical Medicine, Section of Anatomy & Histology & Research Unit of Histology & Embryology, University of Florence, Florence, Italy
| |
Collapse
|
45
|
Serelaxin: A Novel Therapeutic for Vascular Diseases. Trends Pharmacol Sci 2016; 37:498-507. [PMID: 27130518 DOI: 10.1016/j.tips.2016.04.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/19/2022]
Abstract
Vascular dysfunction is an important hallmark of cardiovascular disease. It is characterized by increased sensitivity to vasoconstrictors, decreases in the endothelium-derived vasodilators nitric oxide (NO) and prostacyclin (PGI2), and endothelium-derived hyperpolarization (EDH). Serelaxin (recombinant human relaxin) has gained considerable attention as a new vasoactive drug, largely through its beneficial therapeutic effects in acute heart failure. In this review we first describe the contribution of endogenous relaxin to vascular homeostasis. We then provide a comprehensive overview of the novel mechanisms of serelaxin action in blood vessels that differentiate it from other vasodilator drugs and explain how this peptide could be used more widely as a therapeutic to alleviate vascular dysfunction in several cardiovascular diseases.
Collapse
|
46
|
Abstract
Outcomes for patients with acute heart failure remain suboptimal and treatments principally target improvement of symptoms. To date there has been no therapy approved for acute heart failure shown to improve mortality or readmission risk post-discharge. Serelaxin, a recombinant form of the naturally occurring polypeptide hormone relaxin, has demonstrated promise in preclinical and early clinical trials as a potentially novel therapy for acute heart failure. It is postulated through its anti-fibrotic and vasodilatory effects that this agent can improve outcomes in both the short and long term in these patients. Randomized clinical data has suggested that the medication is safe and well tolerated. However, definitive outcomes data is currently being assessed in a large multi-center trial.
Collapse
Affiliation(s)
- Danyaal S Moin
- a Division of Cardiology , Stony Brook University School of Medicine , Stony Brook , NY , USA
| | - Michelle W Bloom
- a Division of Cardiology , Stony Brook University School of Medicine , Stony Brook , NY , USA
| | - Lampros Papadimitriou
- a Division of Cardiology , Stony Brook University School of Medicine , Stony Brook , NY , USA
| | - Javed Butler
- a Division of Cardiology , Stony Brook University School of Medicine , Stony Brook , NY , USA
| |
Collapse
|
47
|
Castrini AI, Carubelli V, Lazzarini V, Bonadei I, Lombardi C, Metra M. Serelaxin a novel treatment for acute heart failure. Expert Rev Clin Pharmacol 2016; 8:549-57. [PMID: 26294074 DOI: 10.1586/17512433.2015.1073587] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Acute heart failure (AHF) represents a major healthcare burden with a high risk of in-hospital and post-discharge mortality, which remained almost unchanged in the last few decades, underscoring the need of new treatments. Relaxin is a naturally occurring human peptide initially identified as a reproductive hormone and has been shown to play a key role in the maternal hemodynamic and renal adjustments that accommodate pregnancy. Recently, the new molecule serelaxin, a recombinant form of the naturally occurring hormone relaxin has been studied in patients hospitalized for AHF. In addition to vasodilation, serelaxin has anti-oxidative, anti-inflammatory and connective tissue regulating properties. In preclinical studies, it reduced both systemic and renal vascular resistance and, in the clinical trials Pre-RELAX-AHF and RELAX-AHF, it improved dyspnea and signs of congestion. In addition, serelaxin was associated with a reduction of 180-day mortality. The aim of this review is to summarize the pharmacological properties of serelaxin and the results of the preclinical and clinical studies.
Collapse
Affiliation(s)
- Anna Isotta Castrini
- a Department of Medical and Surgical Specialties, Cardiology, Radiological Sciences, and Public Health, University and Civil Hospital of Brescia, Brescia, Italy
| | | | | | | | | | | |
Collapse
|
48
|
Dschietzig TB, Krause-Relle K, Hennequin M, von Websky K, Rahnenführer J, Ruppert J, Grön HJ, Armbruster FP, Bathgate RAD, Aschenbach JR, Forssmann WG, Hocher B. Relaxin-2 does not ameliorate nephropathy in an experimental model of type-1 diabetes. Kidney Blood Press Res 2016; 40:77-88. [PMID: 25791819 DOI: 10.1159/000368484] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS In diabetic nephropathy (DN), the current angiotensin-II-blocking pharmacotherapy is frequently failing. For diabetic cardiomyopathy (DC), there is no specific remedy available. Relaxin-2 (Rlx) - an anti-fibrotic, anti-inflammatory, and vasoprotecting peptide – is a candidate drug for both. METHODS Low-dose (32 μg/kg/day) and high-dose (320 μg/kg/day) Rlx were tested against vehicle (n = 20 each) and non-diabetic controls (n = 14) for 12 weeks in a model of type-1 diabetes induced in endothelial nitric oxide synthase knock-out (eNOS-KO) mice by intraperitoneal injection of streptozotocin. RESULTS Diabetic animals showed normal plasma creatinine, markedly increased albuminuria and urinary malonyldialdehyde, elevated relative kidney weight, glomerulosclerosis, and increased glomerular size, but no relevant interstitial fibrosis. Neither dose of Rlx affected these changes although the drug was active and targeted plasma levels were achieved. Of note, we found no activation of the renal TGF-β pathway in this model. In the hearts of diabetic animals, no fibrotic alterations indicative of DC could be determined which precluded testing of the initial hypothesis. CONCLUSIONS We investigated a model showing early DN without overt tubulointerstitial fibrosis and activation of the TGF-β-Smad-2/3 pathway. In this model, Rlx proved ineffective; however, the same may not apply to other models and types of diabetes.
Collapse
|
49
|
Sarwar M, Samuel CS, Bathgate RA, Stewart DR, Summers RJ. Enhanced serelaxin signalling in co-cultures of human primary endothelial and smooth muscle cells. Br J Pharmacol 2016; 173:484-96. [PMID: 26493539 DOI: 10.1111/bph.13371] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 10/06/2015] [Accepted: 10/10/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE In the phase III clinical trial, RELAX-AHF, serelaxin caused rapid and long-lasting haemodynamic changes. However, the cellular mechanisms involved are unclear in humans. EXPERIMENTAL APPROACH This study examined the effects of serelaxin in co-cultures of human primary endothelial cells (ECs) and smooth muscle cells (SMCs) on cAMP and cGMP signalling. KEY RESULTS Stimulation of HUVECs or human coronary artery endothelial cells (HCAECs) with serelaxin, concentration-dependently increased cGMP accumulation in co-cultured SMCs to a greater extent than in monocultures of either cell type. This was not observed in human umbilical artery endothelial cells (HUAECs) that do not express the relaxin receptor, RXFP1. Treatment of ECs with l-N(G) -nitro arginine (NOARG; 30 μM, 30 min) inhibited serelaxin-mediated (30 nM) cGMP accumulation in HUVECs, HCAECs and co-cultured SMCs. In HCAECs, but not HUVECs, pre-incubation with indomethacin (30 μM, 30 min) also inhibited cGMP accumulation in SMCs. Pre-incubation of SMCs with the guanylate cyclase inhibitor ODQ (1 μM, 30 min) had no effect on serelaxin-mediated (30 nM) cGMP accumulation in HUVECs and HCAECs but inhibited cGMP accumulation in SMCs. Serelaxin stimulation of HCAECs, but not HUVECs, increased cAMP accumulation concentration-dependently in SMCs. Pre-incubation of HCAECs with indomethacin, but not l-NOARG, abolished cAMP accumulation in co-cultured SMCs, suggesting involvement of prostanoids. CONCLUSIONS AND IMPLICATIONS In co-cultures, treatment of ECs with serelaxin caused marked cGMP accumulation in SMCs and with HCAEC also cAMP accumulation. Responses involved EC-derived NO and with HCAEC prostanoid production. Thus, serelaxin differentially modulates vascular tone in different vascular beds.
Collapse
Affiliation(s)
- M Sarwar
- Drug Discovery Biology, Monash Institute of Pharmacology, Monash University, Australia
| | - C S Samuel
- Department of Pharmacology, Monash University, Australia
| | - R A Bathgate
- The Florey Institute of Neuroscience and Mental Health and the Department of Biochemistry and Molecular Biology, University of Melbourne, Australia
| | | | - R J Summers
- Drug Discovery Biology, Monash Institute of Pharmacology, Monash University, Australia
| |
Collapse
|
50
|
Perez AL, Grodin JL, Wu Y, Hernandez AF, Butler J, Metra M, Felker GM, Voors AA, McMurray JJ, Armstrong PW, Starling RC, O'Connor CM, Tang WHW. Increased mortality with elevated plasma endothelin-1 in acute heart failure: an ASCEND-HF biomarker substudy. Eur J Heart Fail 2015; 18:290-7. [PMID: 26663359 DOI: 10.1002/ejhf.456] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 10/09/2015] [Accepted: 10/24/2015] [Indexed: 12/16/2022] Open
Abstract
AIMS Endothelin-1 (ET-1) is an endogenous vasoconstrictor implicated in pulmonary and systemic hypertension, as well as ventricular dysfunction, through effects on vascular smooth muscle, the kidneys, and cardiomyocytes. We aimed to determine the association between serial ET-1 levels and acute heart failure patient outcomes. METHODS AND RESULTS We measured plasma ET-1 at baseline, 48-72 h, and 30 days in a cohort of 872 patients hospitalized with acute heart failure from the ASCEND-HF trial (randomized to nesiritide vs. placebo), and its association with 30-day mortality, 180-day mortality, in-hospital death or worsening heart failure, and 30-day mortality or rehospitalization. Median ET-1 was 7.6 [interquartile range (IQR) 5.9-10] pg/mL at baseline, 6.3 (IQR 4.9-8.1) pg/mL at 48-72 h, and 5.9 (IQR 4.7-7.9) pg/mL at 30 days (P < 0.001). Baseline and 48-72 h ET-1 were found to be independently associated with 180-day mortality in a multivariable analysis [hazard ratio (HR) 1.6, 95% confidence interval (CI) 1.3-2.0, P < 0.001 and HR 1.5, 95% CI 1.2-1.9, P = 0.001, respectively, log-transformed]. ET-1 that was measured at 48-72 h was also independently associated with death or worsening heart failure prior to discharge [odds ratio (OR) 1.6, 95% CI 1.03-2.4, P = 0.03]. These independent associations remained significant after including NT-proBNP in the multivariable analysis. CONCLUSIONS We observed an independent association between elevated ET-1 and short-term in-hospital clinical outcomes and 180-day mortality in hospitalized patients with acute heart failure ET-1 provided additional prognostic information which was incremental to that yielded by NT-proBNP.
Collapse
Affiliation(s)
- Antonio L Perez
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Justin L Grodin
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuping Wu
- Cleveland State University, Department of Mathematics, Cleveland, OH, USA
| | - Adrian F Hernandez
- Duke University Medical Center, Duke Clinical Research Institute, Durham, NC, USA
| | - Javed Butler
- Cardiovascular Division, Stony Brook University, Stony Brook, NY, USA
| | - Marco Metra
- Institute of Cardiology, University of Brescia, Brescia, Italy
| | - G Michael Felker
- Duke University Medical Center, Duke Clinical Research Institute, Durham, NC, USA
| | - Adriaan A Voors
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John J McMurray
- Department of Cardiology, University of Glasgow, Glasgow, UK
| | - Paul W Armstrong
- Department of Cardiology, University of Alberta, Edmonton, Canada
| | | | | | - W H Wilson Tang
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|