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
|
Pankova O, Korzh O. Significance of plasma relaxin-2 levels in patients with primary hypertension and type 2 diabetes mellitus. Wien Med Wochenschr 2024; 174:161-172. [PMID: 38451351 DOI: 10.1007/s10354-024-01035-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 02/06/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND This study aimed to evaluate plasma relaxin‑2 (RLN-2) levels in patients with arterial hypertension (AH) and their relationships with clinical and laboratory parameters. METHODS The study involved 106 hypertensive patients, including 55 with type 2 diabetes mellitus (T2DM), and 30 control subjects. Plasma RLN-2 levels were measured using an enzyme-linked immunosorbent assay kit. RESULTS RLN-2 levels were reduced in patients with AH compared to healthy volunteers (p < 0.001), and hypertensive patients with T2DM had lower RLN-2 levels than those without impaired glucose metabolism (p < 0.001). RLN‑2 was negatively correlated with systolic blood pressure (SBP) (p < 0.001) and anthropometric parameters such as body mass index (BMI; p = 0.027), neck (p = 0.045) and waist (p = 0.003) circumferences, and waist-to-hip ratio (p = 0.011). RLN‑2 also had inverse associations with uric acid levels (p = 0.019) and lipid profile parameters, particularly triglycerides (p < 0.001) and non-HDL-C/HDL‑C (p < 0.001), and a positive relationship with HDL‑C (p < 0.001). RLN‑2 was negatively associated with glucose (p < 0.001), insulin (p = 0.043), HbA1c (p < 0.001), and HOMA-IR index (p < 0.001). Univariate binary logistic regression identified RLN‑2 as a significant predictor of impaired glucose metabolism (p < 0.001). CONCLUSIONS Decreased RLN-2 levels in patients with AH and T2DM and established relationships of RLN‑2 with SBP and parameters of glucose metabolism and lipid profile suggest a diagnostic role of RLN‑2 as a biomarker for AH with T2DM.
Collapse
Affiliation(s)
- Olena Pankova
- Department of General Practice-Family Medicine, Kharkiv National Medical University, Heroiv Kharkova Ave., 275, 61106, Kharkiv, Ukraine.
| | - Oleksii Korzh
- Department of General Practice-Family Medicine, Kharkiv National Medical University, Heroiv Kharkova Ave., 275, 61106, Kharkiv, Ukraine
| |
Collapse
|
3
|
Aragón-Herrera A, Couselo-Seijas M, Feijóo-Bandín S, Anido-Varela L, Moraña-Fernández S, Tarazón E, Roselló-Lletí E, Portolés M, Martínez-Sande JL, García-Seara J, Álvarez E, González-Juanatey JR, Rodríguez-Mañero M, Eiras S, Lago F. Relaxin-2 plasma levels in atrial fibrillation are linked to inflammation and oxidative stress markers. Sci Rep 2022; 12:22287. [PMID: 36566255 PMCID: PMC9789945 DOI: 10.1038/s41598-022-26836-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022] Open
Abstract
Relaxin-2 exerts many favourable cardiovascular effects in pathological circumstances such as atrial fibrillation (AF) and heart failure, but the mechanisms underlying its actions are not completely understood. Since inflammation and fibrosis are pivotal processes in the pathogenesis of AF, our aim was to study the relationship between relaxin-2 plasma levels in left atrium (LA) and peripheral vein with molecules implicated in fibrosis, inflammation and oxidative stress in AF patients, and to evaluate the anti-fibrotic ability of relaxin-2 in normal human atrial cardiac fibroblasts (NHCF-A). Peripheral vein relaxin-2 plasma levels were higher than LA relaxin-2 plasma levels in men while, in women, peripheral vein relaxin-2 levels were increased compared to men. AF patients with higher levels of relaxin-2 exhibited a reduction in H2O2 plasma levels and in mRNA levels of alpha-defensin 3 (DEFA3) and IL-6 in leucocytes from LA plasma. Relaxin-2-in-vitro treatment inhibited NHCF-A migration and decreased mRNA and protein levels of the pro-fibrotic molecule transforming growth factor-β1 (TGF-β1). Our results support an association between relaxin-2 and molecules involved in fibrosis, inflammation and oxidative stress in AF patients, and reinforce an anti-fibrotic protective role of this hormone in NHCF-A; strengthening the relevance of relaxin-2 in AF physiopathology, diagnosis and treatment.
Collapse
Affiliation(s)
- Alana Aragón-Herrera
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Marinela Couselo-Seijas
- Translational Cardiology Group, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Sandra Feijóo-Bandín
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Laura Anido-Varela
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Sandra Moraña-Fernández
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- Cardiology Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela and Health Research Institute, University Clinical Hospital of Santiago de Compostela, 15706, Santiago de Compostela, Spain
| | - Estefanía Tarazón
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
- Cardiocirculatory Unit, Health Institute La Fe University Hospital (IIS La Fe), Avda. de Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Esther Roselló-Lletí
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
- Cardiocirculatory Unit, Health Institute La Fe University Hospital (IIS La Fe), Avda. de Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Manuel Portolés
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
- Cardiocirculatory Unit, Health Institute La Fe University Hospital (IIS La Fe), Avda. de Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - José Luis Martínez-Sande
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
- Arrhytmia Unit, University Clinical Hospital of Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Javier García-Seara
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
- Arrhytmia Unit, University Clinical Hospital of Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Ezequiel Álvarez
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
- Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Moisés Rodríguez-Mañero
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain.
- Arrhytmia Unit, University Clinical Hospital of Santiago de Compostela, Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain.
| | - Sonia Eiras
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
- Translational Cardiology Group, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
| | - Francisca Lago
- Cellular and Molecular Cardiology Unit and Department of Cardiology, Institute of Biomedical Research of Santiago de Compostela (IDIS-SERGAS), Travesía da Choupana s/n, 15706, Santiago de Compostela, Spain
- CIBERCV, Institute of Health Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| |
Collapse
|
4
|
Relaxin-2 as a Potential Biomarker in Cardiovascular Diseases. J Pers Med 2022; 12:jpm12071021. [PMID: 35887517 PMCID: PMC9317583 DOI: 10.3390/jpm12071021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/17/2022] Open
Abstract
The pleiotropic hormone relaxin-2 plays a pivotal role in the physiology and pathology of the cardiovascular system. Relaxin-2 exerts relevant regulatory functions in cardiovascular tissues through the specific receptor relaxin family peptide receptor 1 (RXFP1) in the regulation of cardiac metabolism; the induction of vasodilatation; the reversion of fibrosis and hypertrophy; the reduction of inflammation, oxidative stress, and apoptosis; and the stimulation of angiogenesis, with inotropic and chronotropic effects as well. Recent preclinical and clinical outcomes have encouraged the potential use of relaxin-2 (or its recombinant form, known as serelaxin) as a therapeutic strategy during cardiac injury and/or in patients suffering from different cardiovascular disarrangements, especially heart failure. Furthermore, relaxin-2 has been proposed as a promising biomarker of cardiovascular health and disease. In this review, we emphasize the relevance of the endogenous hormone relaxin-2 as a useful diagnostic biomarker in different backgrounds of cardiovascular pathology, such as heart failure, atrial fibrillation, myocardial infarction, ischemic heart disease, aortic valve disease, hypertension, and atherosclerosis, which could be relevant in daily clinical practice and could contribute to comprehending the specific role of relaxin-2 in cardiovascular diseases.
Collapse
|
5
|
Li Y, Ricardo SD, Samuel CS. Enhancing the Therapeutic Potential of Mesenchymal Stromal Cell-Based Therapies with an Anti-Fibrotic Agent for the Treatment of Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms23116035. [PMID: 35682717 PMCID: PMC9181689 DOI: 10.3390/ijms23116035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 01/02/2023] Open
Abstract
Chronic kidney disease (CKD) affects 1 in 10 members of the general population, placing these patients at an increasingly high risk of kidney failure. Despite the significant burden of CKD on various healthcare systems, there are no effective cures that reverse or even halt its progression. In recent years, human bone-marrow-derived mesenchymal stromal cells (BM-MSCs) have been recognised as a novel therapy for CKDs, owing to their well-established immunomodulatory and tissue-reparative properties in preclinical settings, and their promising safety profile that has been demonstrated in patients with CKDs from several clinical trials. However, renal fibrosis (scarring), a hallmark of CKD, has been shown to impair the viability and functionality of BM-MSCs post-transplantation. This has suggested that BM-MSCs might require a pre-treatment or adjunct therapy that can enhance the viability and therapeutic efficacy of these stromal cells in chronic disease settings. To address this, recent studies that have combined BM-MSCs with the anti-fibrotic drug serelaxin (RLX), have demonstrated the enhanced therapeutic potential of this combination therapy in normotensive and hypertensive preclinical models of CKD. In this review, a critical appraisal of the preclinical data available on the anti-fibrotic and renoprotective actions of BM-MSCs or RLX alone and when combined, as a treatment option for normotensive vs. hypertensive CKD, is discussed.
Collapse
Affiliation(s)
- Yifang Li
- Cardiovascular Disease Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia;
| | - Sharon D. Ricardo
- Development and Stem Cells Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
- Correspondence: (S.D.R.); (C.S.S.)
| | - Chrishan S. Samuel
- Cardiovascular Disease Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia;
- Development and Stem Cells Program, Department of Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Correspondence: (S.D.R.); (C.S.S.)
| |
Collapse
|
6
|
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
|
7
|
Samuel CS, Bennett RG. Relaxin as an anti-fibrotic treatment: Perspectives, challenges and future directions. Biochem Pharmacol 2021; 197:114884. [PMID: 34968489 DOI: 10.1016/j.bcp.2021.114884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 02/07/2023]
Abstract
Fibrosis refers to the scarring and hardening of tissues, which results from a failed immune system-coordinated wound healing response to chronic organ injury and which manifests from the aberrant accumulation of various extracellular matrix components (ECM), primarily collagen. Despite being a hallmark of prolonged tissue damage and related dysfunction, and commonly associated with high morbidity and mortality, there are currently no effective cures for its regression. An emerging therapy that meets several criteria of an effective anti-fibrotic treatment, is the recombinant drug-based form of the human hormone, relaxin (also referred to as serelaxin, which is bioactive in several other species). This review outlines the broad anti-fibrotic and related organ-protective roles of relaxin, mainly from studies conducted in preclinical models of ageing and fibrotic disease, including its ability to ameliorate several aspects of fibrosis progression and maturation, from immune cell infiltration, pro-inflammatory and pro-fibrotic cytokine secretion, oxidative stress, organ hypertrophy, cell apoptosis, myofibroblast differentiation and ECM production, to its ability to facilitate established ECM degradation. Studies that have compared and/or combined these therapeutic effects of relaxin with current standard of care medication have also been discussed, along with the main challenges that have hindered the translation of the anti-fibrotic efficacy of relaxin to the clinic. The review then outlines the future directions as to where scientists and several pharmaceutical companies that have recognized the therapeutic potential of relaxin are working towards, to progress its development as a treatment for human patients suffering from various fibrotic diseases.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria 3052, Australia.
| | - Robert G Bennett
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA; Department of Internal Medicine, Division of Diabetes, Endocrinology & Metabolism, University of Nebraska Medical Center, Omaha, NE 68198-4130, USA.
| |
Collapse
|
8
|
Mao A, Zhang P, Zhang K, Kan H, He D, Han X, Wang Z, Tang C, Ma X. Endothelial TRPV4-eNOS coupling as a vital therapy target for treatment of hypertension. Br J Pharmacol 2021; 179:2297-2312. [PMID: 34822720 DOI: 10.1111/bph.15755] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/25/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Reduced nitric oxide (NO) level and activity are signs of endothelial dysfunction, which is important in mediating blood pressure up-regulation. Previously, we demonstrated that transient receptor potential channel V4 (TRPV4) could form functional complex with other proteins to mediate vasodilation in the Endothelial cells (ECs). But how TRPV4 interacts with the NO pathway in larger arteries requires further exploration. EXPERIMENTAL APPROACH We used single-cell RNA-sequencing to find the CD106+ TRPV4high NOS3high ECs. The TRPV4-eNOS interaction was verified by co-immunoprecipitation and Immunofluorescence resonance energy transfer (FRET), and their binding site was found by site-directed mutagenesis. Endothelium-specific TRPV4 knockout (TRPV4EC -/- ) mice were used to study the effect of the TRPV4-eNOS interaction on blood pressure. A small molecule, JNc-463 was designed through molecular docking technology. KEY RESULTS We uncovered CD106+ TRPV4high NOS3high ECs in the mouse aorta, which they could regulate vasodilation via a TRPV4-eNOS interaction, and they were essential to regulate blood pressure. The TRPV4-eNOS interaction markedly decreased during the process of hypertension. We further attempted to identify the molecules re-join the TRPV4-eNOS interaction and develop a small-molecule drug, JNc-463, which could increase the TRPV4-eNOS interaction to enhance vasodilation, and exert antihypertensive effects in mice. CONCLUSION AND IMPLICATIONS This is the first study integrating single-cell RNA-Seq, single-cell functional study and drug screening in aorta. We identified a subpopulation of CD106+ TRPV4high NOS3high ECs, in which an impaired TRPV4-eNOS interaction was important in the progress of hypertension and we designed a small molecule, JNc-463 to improve the impaired TRPV4-eNOS interaction in hypertension.
Collapse
Affiliation(s)
- Aiqin Mao
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Peng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Ka Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Hao Kan
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongxu He
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xiping Han
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Zhiwei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
9
|
Kopaliani I, Jarzebska N, Billoff S, Kolouschek A, Martens-Lobenhoffer J, Bornstein SR, Bode-Böger SM, Ragavan VN, Weiss N, Mangoni AA, Deussen A, Rodionov RN. Overexpression of dimethylarginine dimethylaminohydrolase 1 protects from angiotensin II-induced cardiac hypertrophy and vascular remodeling. Am J Physiol Heart Circ Physiol 2021; 321:H825-H838. [PMID: 34533401 DOI: 10.1152/ajpheart.00064.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 09/01/2021] [Indexed: 11/22/2022]
Abstract
Cardiovascular complications are the leading cause of death, and elevated levels of asymmetric dimethyarginine (ADMA), an endogenous inhibitor of nitric oxide synthase, are implicated in their pathophysiology. We investigated the role of dimethylarginine dimethylaminohydrolase 1 (DDAH1), an enzyme hydrolyzing ADMA, in prevention of cardiovascular remodeling during hypertension. We hypothesized that the animals overexpressing DDAH1 will be protected from angiotensin II (ANG II)-induced end organ damage. Angiotensin II (ANG II) was infused in two doses: 0.75 and 1.5 mg/kg/day in DDAH1 transgenic mice (DDAH1 TG) and wild-type (WT) littermates for 2 or 4 wk. Echocardiography was performed in the first and fourth weeks of the infusion, systolic blood pressure (SBP) was measured weekly, and cardiac hypertrophy and vascular remodeling was assessed by histology. Increase in SBP after 1 wk of ANG II infusion was not different between the groups, whereas TG mice had lower SBP at later time points. TG mice were protected from cardiovascular remodeling after 2 wk of ANG II infusion in the high dose and after 4 wk in the moderate dose. TG mice had higher left ventricular lumen-to-wall ratio, lower cardiomyocyte cross-sectional area, and less interstitial fibrosis compared with WT controls. In aorta, TG mice had less adventitial fibrosis, lower medial thickness with preserved elastin content, lower counts of inflammatory cells, lower levels of active matrix metalloproteinase-2, and showed better endothelium-dependent relaxation. We demonstrated that overexpression of DDAH1 protects from ANG II-induced cardiovascular remodeling and progression of hypertension by preserving endothelial function and limiting inflammation.NEW & NOTEWORTHY We showed that overexpression of dimethylarginine dimethylaminohydrolase 1 (DDAH1) protects from angiotensin II-induced cardiovascular damage, progression of hypertension, and adverse vascular remodeling in vivo. This protective effect is associated with decreased levels of asymmetric dimethylarginine, preservation of endothelial function, inhibition of cardiovascular inflammation, and lower activity of matrix metalloproteinase-2. Our findings are highly clinically relevant, because they suggest that upregulation of DDAH1 might be a promising therapeutic approach against angiotensin II-induced end organ damage.
Collapse
Affiliation(s)
- Irakli Kopaliani
- Department of Physiology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Natalia Jarzebska
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- Department of Anesthesiology and Critical Care Medicine, University Hospital Dresden, Dresden University of Technology, Dresden, Germany
- Department of Internal Medicine III, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Silke Billoff
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
| | - Anne Kolouschek
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
| | | | - Stefan R Bornstein
- University Clinic and Polyclinic III, Dresden University of Technology, Dresden, Germany
| | - Stefanie M Bode-Böger
- Institute of Clinical Pharmacology, Otto-von-Guericke University, Magdeburg, Germany
| | - Vinitha N Ragavan
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Norbert Weiss
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- University Clinic and Polyclinic III, Dresden University of Technology, Dresden, Germany
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Andreas Deussen
- Department of Physiology, Medical Faculty, Dresden University of Technology, Dresden, Germany
| | - Roman N Rodionov
- University Center for Vascular Medicine, Dresden University of Technology, Dresden, Germany
- University Clinic and Polyclinic III, Dresden University of Technology, Dresden, Germany
| |
Collapse
|
10
|
Kohsaka T, Yoneda Y, Yoshida T, Minagawa I, Pitia AM, Iwasawa A, Ikegaya N. Relaxin exerts a protective effect during ischemia-reperfusion in the rat model. Andrology 2021; 10:179-189. [PMID: 34435470 DOI: 10.1111/andr.13096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND Testicular torsion, which causes ischemia-reperfusion (IR) injury, is a serious urological emergency that can lead to testicular dysfunction, including infertility, primarily among newborn and pubertal males; thus, effective drugs should be administered during or after ischemia. OBJECTIVES Using a rat model of testicular IR injury, the present study investigated the protective effects of relaxin (RLN) against oxidative stress, testicular dysfunction, inflammation, histological damage, arrested spermatogenesis, and germ cell apoptosis as well as explored the usefulness of RLN as a potential protective drug for IR injury combined with surgical treatment. MATERIALS AND METHODS Male Sprague-Dawley rats were subjected to left testicular ischemia for 2 h, followed by 24 h of reperfusion. They were subsequently divided into three groups: sham, IR, and IR + RLN groups. Porcine RLN (500 ng/h) or saline was infused using an implanted osmotic mini-pump 90 min after inducing ischemia. The RLN dose used herein was that which resulted in serum RLN levels comparable to those in mid-pregnant rats based on previous studies. RESULTS Testicular IR increased germ cell apoptosis and histological damage as well as promoted disorganized and arrested spermatogenesis, accompanied by a significant increase in oxidative stress and inflammation. However, RLN administration ameliorated the adverse consequences associated with IR injury by attenuating oxidative stress and mitigating apoptosis and inflammation. DISCUSSION AND CONCLUSION The study findings clearly demonstrated that RLN exerts a protective effect against IR-induced testicular injury by attenuating oxidative stress, apoptosis, and inflammation, suggesting that RLN together with surgical treatment is a potentially efficacious approach toward ameliorating testicular dysfunction following testicular torsion.
Collapse
Affiliation(s)
- Tetsuya Kohsaka
- Department of Applied Life Sciences, Shizuoka University, Shizuoka, Japan
| | | | - Takuya Yoshida
- Department of Clinical Nutrition, University of Shizuoka, Shizuoka, Japan
| | - Itaru Minagawa
- Department of Applied Life Sciences, Shizuoka University, Shizuoka, Japan
| | - Ali M Pitia
- Department of Applied Life Sciences, Shizuoka University, Shizuoka, Japan
| | - Atsushi Iwasawa
- Department of Agricultural and Environmental Science, Gifu University, Gifu, Japan
| | - Naoki Ikegaya
- Department of Medicine, Yaizu Municipal General Hospital, Shizuoka, Japan
| |
Collapse
|
11
|
Li Y, Shen M, Ferens D, Broughton BRS, Murthi P, Saini S, Widdop RE, Ricardo SD, Pinar AA, Samuel CS. Combining mesenchymal stem cells with serelaxin provides enhanced renoprotection against 1K/DOCA/salt-induced hypertension. Br J Pharmacol 2021; 178:1164-1181. [PMID: 33450051 DOI: 10.1111/bph.15361] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Fibrosis is a hallmark of chronic kidney disease (CKD) that significantly contributes to renal dysfunction, and impairs the efficacy of stem cell-based therapies. This study determined whether combining bone marrow-derived mesenchymal stem cells (BM-MSCs) with the renoprotective effects of recombinant human relaxin (serelaxin) could therapeutically reduce renal fibrosis in mice with one kidney/deoxycorticosterone acetate/salt (1K/DOCA/salt)-induced hypertension, compared with the effects of the ACE inhibitor, perindopril. EXPERIMENTAL APPROACH Adult male C57BL/6 mice were uni-nephrectomised and received deoxycorticosterone acetate and saline to drink (1K/DOCA/salt) for 21 days. Control mice were uni-nephrectomised but received water over the same time period. Sub-groups of 1K/DOCA/salt-injured mice (n = 5-8 per group) were treated with either serelaxin (0.5 mg·kg-1 ·day-1 ) or BM-MSCs (1 × 106 per mouse) alone; both treatments combined (with 0.5 × 106 or 1 × 106 BM-MSCs per mouse); or perindopril (2 mg·kg-1 ·day-1 ) from days 14-21. KEY RESULTS 1K/DOCA/salt-injured mice developed elevated BP and hypertension-induced renal damage, inflammation and fibrosis. BM-MSCs alone reduced the injury-induced fibrosis and attenuated BP to a similar extent as perindopril. Serelaxin alone modestly reduced renal fibrosis and effectively reduced tubular injury. Strikingly, the combined effects of BM-MSCs (at both doses) with serelaxin significantly inhibited renal fibrosis and proximal tubular epithelial injury while restoring renal architecture, to a greater extent than either therapy alone, and over the effects of perindopril. CONCLUSION AND IMPLICATIONS Combining BM-MSCs and serelaxin provided broader renoprotection over either therapy alone or perindopril and might represent a novel treatment for hypertensive CKD.
Collapse
Affiliation(s)
- Yifang Li
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Matthew Shen
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Dorota Ferens
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Brad R S Broughton
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Padma Murthi
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Sheetal Saini
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Anita A Pinar
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash University, Clayton, Victoria, Australia.,Development and Stem Cells Program, Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
12
|
Donia T, Khamis A. Management of oxidative stress and inflammation in cardiovascular diseases: mechanisms and challenges. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:34121-34153. [PMID: 33963999 DOI: 10.1007/s11356-021-14109-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/21/2021] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases (CVDs) have diverse physiopathological mechanisms with interconnected oxidative stress and inflammation as one of the common etiologies which result in the onset and development of atherosclerotic plaques. In this review, we illustrate this strong crosstalk between oxidative stress, inflammation, and CVD. Also, mitochondrial functions underlying this crosstalk, and various approaches for the prevention of redox/inflammatory biological impacts will be illustrated. In part, we focus on the laboratory biomarkers and physiological tests for the evaluation of oxidative stress status and inflammatory processes. The impact of a healthy lifestyle on CVD onset and development is displayed as well. Furthermore, the differences in oxidative stress and inflammation are related to genetic susceptibility to cardiovascular diseases and the variability in the assessment of CVDs risk between individuals; Omics technologies for measuring oxidative stress and inflammation will be explored. Finally, we display the oxidative stress-related microRNA and the functions of the redox basis of epigenetic modifications.
Collapse
Affiliation(s)
- Thoria Donia
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Abeer Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
13
|
Wang C, Gaspari TA, Ferens D, Spizzo I, Kemp-Harper BK, Samuel CS. Simultaneous targeting of oxidative stress and fibrosis reverses cardiomyopathy-induced ventricular remodelling and dysfunction. Br J Pharmacol 2021; 178:2424-2442. [PMID: 33660265 DOI: 10.1111/bph.15428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 02/14/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Oxidative stress and fibrosis are hallmarks of cardiomyopathy-induced heart failure yet are not effectively targeted by current frontline therapies. Here, the therapeutic effects of the anti-oxidant, N-acetylcysteine (NAC), were compared and combined with an acute heart failure drug with established anti-fibrotic effects, serelaxin (RLX), in a murine model of cardiomyopathy. EXPERIMENTAL APPROACH Adult male 129sv mice were subjected to repeated isoprenaline (25 mg·kg-1 )-induced cardiac injury for five consecutive days and then left to undergo fibrotic healing until Day 14. Subgroups of isoprenaline-injured mice were treated with RLX (0.5 mg·kg-1 ·day-1 ), NAC (25 mg·kg-1 ·day-1 ) or both combined, given subcutaneously via osmotic minipumps from Day 7 to 14. Control mice received saline instead of isoprenaline. KEY RESULTS Isoprenaline-injured mice showed increased left ventricular (LV) inflammation (~5-fold), oxidative stress (~1-2.5-fold), cardiomyocyte hypertrophy (~25%), cardiac remodelling, fibrosis (~2-2.5-fold) and dysfunction by Day 14 after injury. NAC alone blocked the cardiomyopathy-induced increase in LV superoxide levels, to a greater extent than RLX. Additionally, either treatment alone only partly reduced several measures of LV inflammation, remodelling and fibrosis. In comparison, the combination of RLX and NAC prevented the cardiomyopathy-induced LV macrophage infiltration, remodelling, fibrosis and cardiomyocyte size, to a greater extent than either treatment alone after 7 days. The combination therapy also restored the isoprenaline-induced reduction in LV function, without affecting systolic BP. CONCLUSION AND IMPLICATIONS These findings demonstrated that the simultaneous targeting of oxidative stress and fibrosis is key to treating the pathophysiology and dysfunction induced by cardiomyopathy.
Collapse
Affiliation(s)
- Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Tracey A Gaspari
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Dorota Ferens
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Iresha Spizzo
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Barbara K Kemp-Harper
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | - Chrishan S Samuel
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
14
|
Oxymatrine Ameliorates Memory Impairment in Diabetic Rats by Regulating Oxidative Stress and Apoptosis: Involvement of NOX2/NOX4. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3912173. [PMID: 33273999 PMCID: PMC7683156 DOI: 10.1155/2020/3912173] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 08/11/2020] [Accepted: 10/28/2020] [Indexed: 01/09/2023]
Abstract
Oxymatrine (OMT) is the major quinolizidine alkaloid extracted from the root of Sophora flavescens Ait and has been shown to exhibit a diverse range of pharmacological properties. The aim of the present study was to investigate the role of OMT in diabetic brain injury in vivo and in vitro. Diabetic rats were induced by intraperitoneal injection of a single dose of 65 mg/kg streptozotocin (STZ) and fed a high-fat and high-cholesterol diet. Memory function was assessed using a Morris water maze test. A SH-SY5Y cell injury model was induced by incubation with glucose (30 mM/l) to simulate damage in vitro. The serum fasting blood glucose, insulin, serum S100B, malondialdehyde (MDA), and superoxide dismutase (SOD) levels were analyzed using commercial kits. Morphological changes were observed using Nissl staining and electron microscopy. Cell apoptosis was assessed using Hoechst staining and TUNEL staining. NADPH oxidase (NOX) and caspase-3 activities were determined. The effects of NOX2 and NOX4 knockdown were assessed using small interfering RNA. The expression levels of NOX1, NOX2, and NOX4 were detected using reverse transcription-quantitative PCR and western blotting, and the levels of caspase-3 were detected using western blotting. The diabetic rats exhibited significantly increased plasma glucose, insulin, reactive oxygen species (ROS), S-100B, and MDA levels and decreased SOD levels. Memory function was determined by assessing the percentage of time spent in the target quadrant, the number of times the platform was crossed, escape latency, and mean path length and was found to be significantly reduced in the diabetic rats. Hyperglycemia resulted in notable brain injury, including histological changes and apoptosis in the cortex and hippocampus. The expression levels of NOX2 and NOX4 were significantly upregulated at the protein and mRNA levels, and NOX1 expression was not altered in the diabetic rats. NOX and caspase-3 activities were increased, and caspase-3 expression was upregulated in the brain tissue of diabetic rats. OMT treatment dose-dependently reversed behavioral, biochemical, and molecular changes in the diabetic rats. In vitro, high glucose resulted in increases in reactive oxygen species (ROS), MDA levels, apoptosis, and the expressions of NOX2, NOX4, and caspase-3. siRNA-mediated knockdown of NOX2 and NOX4 decreased NOX2 and NOX4 expression levels, respectively, and reduced ROS levels and apoptosis. The results of the present study suggest that OMT alleviates diabetes-associated cognitive decline, oxidative stress, and apoptosis via NOX2 and NOX4 inhibition.
Collapse
|
15
|
Li X, Xuan W, Chen D, Gao H, Wang G, Guo Q, Wang Y, Song H, Cai B. Research Progress of Alzheimer's Disease Therapeutic Drugs: Based on Renin-Angiotensin System Axis. J Alzheimers Dis 2020; 78:1315-1338. [PMID: 33164932 DOI: 10.3233/jad-200770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It is widely recognized that Alzheimer's disease (AD) has a complicate link to renin-angiotensin system (RAS). It is known that cerebrovascular disease has some connections with AD, but most of the studies are still conducted in parallel or independently. Although previous research came up with large number of hypotheses about the pathogenesis of AD, it does not include the mechanism of RAS-related regulation of AD. It has been found that many components of RAS have been changed in AD. For example, the multifunctional and high-efficiency vasoconstrictor Ang II and Ang III with similar effects are changed under the action of other RAS signal peptides; these signal peptides are believed to help improve nerve injury and cognitive function. These changes may lead to neuropathological changes of AD, and progressive defects of cognitive function, which are association with some hypotheses of AD. The role of RAS in AD gradually attracts our attention, and RAS deserved to be considered carefully in the pathogenesis of AD. This review discusses the mechanisms of RAS participating in the three current hypotheses of AD: neuroinflammation, oxidative stress and amyloid-β protein (Aβ) hypothesis, as well as the drugs that regulate RAS systems already in clinical or in clinical trials. It further demonstrates the importance of RAS in the pathogenesis of AD, not only because of its multiple aspects of participation, which may be accidental, but also because of the availability of RAS drugs, which can be reused as therapies of AD.
Collapse
Affiliation(s)
- Xinquan Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Weiting Xuan
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Dabao Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Huawu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Guangyun Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Qiaoru Guo
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the Fifth Affiliated Hospital and School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.,Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China.,Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| |
Collapse
|
16
|
|
17
|
Kohsaka T, Minagawa I, Morimoto M, Yoshida T, Sasanami T, Yoneda Y, Ikegaya N, Sasada H. Efficacy of relaxin for cisplatin-induced testicular dysfunction and epididymal spermatotoxicity. Basic Clin Androl 2020; 30:3. [PMID: 32166037 PMCID: PMC7061478 DOI: 10.1186/s12610-020-0101-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 01/19/2020] [Indexed: 12/12/2022] Open
Abstract
Background Cisplatin (CP) is an extremely effective anticancer agent widely used to treat various cancer types, however, the potential side effects include testicular dysfunction. This study was to investigate, using a rat model of CP-induced testicular dysfunction, the protective effects of relaxin (RLN) against oxidative stress, testicular function, histological damage, spermatogenesis, germ-cell apoptosis, and sperm output, and to explore the usefulness of RLN as a potential protective drug for use with CP in chemotherapeutic treatments. Methods Sprague-Dawley male rats were used, which were divided into three groups: sham control, CP, and CP + RLN. Porcine RLN (500 ng/h) or saline was infused for 5 days using an implanted osmotic mini-pump following intraperitoneal injection of CP (6 mg/kg). RLN dose was chosen based on previous studies showing that it resulted in serum relaxin levels comparable to those in rats at the middle of pregnancy. At 5 days after CP administration, samples were collected and assessment of testicular histopathology, germ-cell apoptosis, oxidative stress, lipid peroxidation, and sperm quality was performed as main measures. Results The testicular CP model showed reduced testis weight and significantly decreased spermatogenesis scores. Additionally, CP administration induced a 4.6-fold increase in the apoptotic index associated with a significant increase in oxidative stress and upregulation of pro-apoptotic Casp3 and downregulation of anti-apoptotic Bcl2 levels, resulting in a marked reduction in sperm concentration. However, RLN administration caused a significant reduction in CP-mediated damage by attenuating oxidative stress and cell apoptosis. RLN administration efficiently scavenged ROS via the activation of SOD, CAT, and GPx and upregulation of GSH to prevent lipid peroxidation and decreased apoptosis by altering Bcl2 and Casp3 expression, thereby reducing histopathological damage and restoring spermatogenesis. Furthermore, RLN ameliorated attenuated sperm motility in the cauda epididymis resulting from CP treatment. Conclusions This study clearly indicates that RLN exerts a protective effect against CP-induced testicular damage through attenuation of oxidative stress and suppression of apoptosis. Our findings suggest RLN as a potentially efficacious drug for use with cisplatin chemotherapy in order to ameliorate CP-induced side effects and testicular injury adversely affecting spermatogenesis, sperm quality, and oxidative-stress parameters.
Collapse
Affiliation(s)
- Tetsuya Kohsaka
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Itaru Minagawa
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Masashi Morimoto
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Takuya Yoshida
- 2Department of Clinical Nutrition, School of Food and Nutritional Science, University of Shizuoka, Shizuoka, 422-8526 Japan
| | - Tomohiro Sasanami
- 1Department of Applied Life Sciences, Animal Reproduction & Physiology Faculty of Agriculture, Shizuoka University, 836 Ohya, Suruga-ku, Shizuoka, 422-8529 Japan
| | - Yoshitaka Yoneda
- Advanced Reproductive Medical Center, Shizuoka Ladies Clinic, Shizuoka, 420-0837 Japan
| | - Naoki Ikegaya
- Department of Medicine, Yaizu Municipal General Hospital, Shizuoka, 422-8505 Japan
| | - Hiroshi Sasada
- 5Division of Animal Science, Kitasato University School of Veterinary Medicine, Towada, 034-8628 Japan
| |
Collapse
|
18
|
Altered Cerebral Blood Flow and Potential Neuroprotective Effect of Human Relaxin-2 (Serelaxin) During Hypoxia or Severe Hypovolemia in a Sheep Model. Int J Mol Sci 2020; 21:ijms21051632. [PMID: 32120997 PMCID: PMC7084399 DOI: 10.3390/ijms21051632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 12/15/2022] Open
Abstract
Specific neuroprotective strategies to minimize cerebral damage caused by severe hypoxia or hypovolemia are lacking. Based on previous studies showing that relaxin-2/serelaxin increases cortical cerebral blood flow, we postulated that serelaxin might provide a neuroprotective effect. Therefore, we tested serelaxin in two emergency models: hypoxia was induced via inhalation of 5% oxygen and 95% nitrogen for 12 min; thereafter, the animals were reoxygenated. Hypovolemia was induced and maintained for 20 min by removal of 50% of the total blood volume; thereafter, the animals were retransfused. In each damage model, the serelaxin group received an intravenous injection of 30 µg/kg of serelaxin in saline, while control animals received saline only. Blood gases, shock index values, heart frequency, blood pressure, and renal blood flow showed almost no significant differences between control and treatment groups in both settings. However, serelaxin significantly blunted the increase of lactate during hypovolemia. Serelaxin treatment resulted in significantly elevated cortical cerebral blood flow (CBF) in both damage models, compared with the respective control groups. Measurements of the neuroproteins S100B and neuron-specific enolase in cerebrospinal fluid revealed a neuroprotective effect of serelaxin treatment in both hypoxic and hypovolemic animals, whereas in control animals, neuroproteins increased during the experiment. Western blotting showed the expression of relaxin receptors and indicated region-specific differences in relaxin receptor-mediated signaling in cortical and subcortical brain arterioles, respectively. Our findings support the hypothesis that serelaxin is a potential neuroprotectant during hypoxia and hypovolemia. Due to its preferential improvement of cortical CBF, serelaxin might reduce cognitive impairments associated with these emergencies.
Collapse
|
19
|
Chow BSM, Kocan M, Shen M, Wang Y, Han L, Chew JY, Wang C, Bosnyak S, Mirabito-Colafella KM, Barsha G, Wigg B, Johnstone EKM, Hossain MA, Pfleger KDG, Denton KM, Widdop RE, Summers RJ, Bathgate RAD, Hewitson TD, Samuel CS. AT1R-AT2R-RXFP1 Functional Crosstalk in Myofibroblasts: Impact on the Therapeutic Targeting of Renal and Cardiac Fibrosis. J Am Soc Nephrol 2019; 30:2191-2207. [PMID: 31511361 DOI: 10.1681/asn.2019060597] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/29/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Recombinant human relaxin-2 (serelaxin), which has organ-protective actions mediated via its cognate G protein-coupled receptor relaxin family peptide receptor 1 (RXFP1), has emerged as a potential agent to treat fibrosis. Studies have shown that serelaxin requires the angiotensin II (AngII) type 2 receptor (AT2R) to ameliorate renal fibrogenesis in vitro and in vivo. Whether its antifibrotic actions are affected by modulation of the AngII type 1 receptor (AT1R), which is expressed on myofibroblasts along with RXFP1 and AT2R, is unknown. METHODS We examined the signal transduction mechanisms of serelaxin when applied to primary rat renal and human cardiac myofibroblasts in vitro, and in three models of renal- or cardiomyopathy-induced fibrosis in vivo. RESULTS The AT1R blockers irbesartan and candesartan abrogated antifibrotic signal transduction of serelaxin via RXFP1 in vitro and in vivo. Candesartan also ameliorated serelaxin's antifibrotic actions in the left ventricle of mice with cardiomyopathy, indicating that candesartan's inhibitory effects were not confined to the kidney. We also demonstrated in a transfected cell system that serelaxin did not directly bind to AT1Rs but that constitutive AT1R-RXFP1 interactions could form. To potentially explain these findings, we also demonstrated that renal and cardiac myofibroblasts expressed all three receptors and that antagonists acting at each receptor directly or allosterically blocked the antifibrotic effects of either serelaxin or an AT2R agonist (compound 21). CONCLUSIONS These findings have significant implications for the concomitant use of RXFP1 or AT2R agonists with AT1R blockers, and suggest that functional interactions between the three receptors on myofibroblasts may represent new targets for controlling fibrosis progression.
Collapse
Affiliation(s)
- Bryna S M Chow
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Martina Kocan
- Florey Institute of Neuroscience and Mental Health.,Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Matthew Shen
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Yan Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Lei Han
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Jacqueline Y Chew
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Chao Wang
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Sanja Bosnyak
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Katrina M Mirabito-Colafella
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Giannie Barsha
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Belinda Wigg
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Elizabeth K M Johnstone
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | | | - Kevin D G Pfleger
- Harry Perkins Institute of Medical Research and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia.,Department of Pharmacology and Therapeutics, ARC Centre for Personalised Therapeutic Technologies, Melbourne, Australia; and.,Dimerix Limited, Nedlands, Western Australia, Australia
| | - Kate M Denton
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Roger J Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia.,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| | - Ross A D Bathgate
- Florey Institute of Neuroscience and Mental Health.,Department of Biochemistry and Molecular Biology, and
| | - Tim D Hewitson
- Department of Nephrology, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Chrishan S Samuel
- Department of Biochemistry and Molecular Biology, and .,Cardiovascular Disease Program, Monash Biomedicine Discovery Institute and Department of Pharmacology and
| |
Collapse
|
20
|
Wolf VL, Phillips TL, Taylor EB, Sasser JM, Ryan MJ. Human recombinant relaxin-2 does not attenuate hypertension or renal injury but exacerbates vascular dysfunction in a female mouse model of SLE. Am J Physiol Heart Circ Physiol 2019; 317:H234-H242. [PMID: 31125285 DOI: 10.1152/ajpheart.00174.2019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that disproportionately affects women of reproductive age and increases their risk for developing hypertension, vascular, and renal disease. Relaxin has potential beneficial therapeutic effects in cardiovascular disease through direct actions on the vasculature. The potential therapeutic benefit of relaxin on SLE-associated cardiovascular and renal risk factors like hypertension has not previously been tested. We hypothesized that relaxin would attenuate hypertension, renal injury, and vascular dysfunction in an established female mouse model of SLE (NZBWF1 mice). Serelaxin (human recombinant relaxin-2, 0.5 mg·kg-1·day-1) or vehicle was administered via osmotic mini-pump for 4 wk in female control (NZW) or SLE mice between 28 and 31 wk of age. Serelaxin treatment increased uterine weights in both groups, suggesting that the Serelaxin was bioactive. Mean arterial pressure, measured by carotid artery catheter, was significantly increased in vehicle-treated SLE mice compared with vehicle-treated controls, but was not changed by Serelaxin treatment. Albumin excretion rate, measured by ELISA, was similar between vehicle- and Serelaxin-treated SLE mice and between vehicle- and Serelaxin-treated control mice. Wire myography was performed using isolated carotid arteries to assess endothelial-independent and -dependent vasodilation, and data confirm that SLE mice have impaired endothelium-independent and -dependent relaxation compared with control mice. Serelaxin treatment did not affect endothelium-independent vasodilation, but exacerbated the endothelium-dependent dysfunction. These data suggest that, contrary to our hypothesis, Serelaxin infusion does not attenuate hypertension, renal injury, or vascular dysfunction in SLE, but worsens underlying vascular endothelial dysfunction in this experimental model of SLE. These data do not support the use of human recombinant relaxin-2 as an antihypertensive in the SLE patient population. NEW & NOTEWORTHY Relaxin is a peptide hormone commonly known for its role in pregnancy and for its use in recent clinical trials for the treatment of heart failure. Evidence suggests that relaxin has immunomodulatory effects; however, the potential therapeutic impact of relaxin in chronic immune mediated disease is unclear. This study tests whether recombinant human relaxin (Serelaxin) attenuates the progression of autoimmunity, and the associated cardiovascular consequences, in an experimental model of systemic lupus erythematosus.
Collapse
Affiliation(s)
- Victoria L Wolf
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Taylor L Phillips
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Erin B Taylor
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Jennifer M Sasser
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,GV (Sonny) Montgomery Veterans Affairs Medical Center , Jackson, Mississippi
| |
Collapse
|
21
|
Angiotensin II-induced hypertension increases the mutant frequency in rat kidney. Arch Toxicol 2019; 93:2045-2055. [PMID: 31098697 DOI: 10.1007/s00204-019-02477-8] [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: 03/29/2019] [Accepted: 05/08/2019] [Indexed: 01/05/2023]
Abstract
Epidemiological studies revealed an increased risk for kidney cancer in hypertensive patients. In many of these patients, the blood pressure regulating renin-angiotensin-aldosterone system (RAAS) is activated. A stimulated RAAS leads to oxidative stress and increases markers of DNA damage, both in vitro and in animal models of hypertension. However, the mutagenic potential of RAAS activation has not been investigated yet. To quantify hypertension-induced mutations, BigBlue®+/- rats, which carry a transgenic lacI gene for mutation analysis, were treated for 20 weeks with a mean dose of 400 µg angiotensin II/kg × day. Angiotensin II-treated animals showed significantly increased blood pressure and impaired kidney function. Urinary excretion of oxidized nucleobases was raised. Additionally, in the renal cortex, oxidative stress, oxidatively generated DNA lesions and DNA strandbreaks were significantly increased. Further, a significant elevation of the mutant frequency in kidney DNA was detected. Sequencing revealed the presence of GC → T:A transversions in the mutated lacI genes of the angiotensin II-treated animals as a result of unrepaired oxidatively modified DNA bases, while no such transversions were found in the mutated lacI genes from control animals. The results demonstrate that the oxidative stress and DNA damage previously observed in kidney cells in vitro and in vivo after angiotensin II treatment indeed is associated with the accumulation of mutations in rat kidneys, providing further evidence for a cancer-initiating potential of elevated angiotensin II concentrations.
Collapse
|
22
|
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: 1.7] [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
|
23
|
Pouwels S, Van Genderen ME, Kreeftenberg HG, Ribeiro R, Parmar C, Topal B, Celik A, Ugale S. Utility of the cold pressor test to predict future cardiovascular events. Expert Rev Cardiovasc Ther 2019; 17:305-318. [PMID: 30916592 DOI: 10.1080/14779072.2019.1598262] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The cold pressor test (CPT) is a common and extensively validated test, which induces systemic stress involving immersion of an individual's hand in ice water (normally temperature between 0 and 5 degrees Celsius) for a period of time. CPT has been used in various fields, like examining effects of stress on memory, decision-making, pain and cardiovascular health. Areas covered: In terms of cardiovascular health, current research is mainly interested in predicting the occurrence of cardiovascular (CV) events. The objective of this review is to give an overview of the history and methodology of the CPT, and clinical utility in possibly predicting CV events in CAD and other atherosclerotic diseases. Secondly, we will discuss possible future applications of the CPT in clinical care. Expert opinion: An important issue to address is the fact that the physiology of the CPT is not fully understood at this moment. As pointed out multiple mechanisms might be responsible for contributing to either coronary vasodilatation or coronary vasoconstriction. Regarding the physiological mechanism of the CPT and its effect on the measurements of the carotid artery reactivity even less is known.
Collapse
Affiliation(s)
- Sjaak Pouwels
- a Department of Surgery , Franciscus Gasthuis & Vlietland , Rotterdam/Schiedam , The Netherlands
| | - Michel E Van Genderen
- b Department of Internal Medicine , Franciscus Gasthuis & Vlietland , Rotterdam/Schiedam , The Netherlands
| | - Herman G Kreeftenberg
- c Department of Internal Medicine , Catharina Hospital , Eindhoven , The Netherlands.,d Department of Intensive Care Medicine , Catharina Hospital , Eindhoven , The Netherlands
| | - Rui Ribeiro
- e Metabolic Patient Multidisciplinary Centre , Clínica de Santo António , Lisbon , Portugal
| | - Chetan Parmar
- f Department of Surgery , Whittington Hospital , London , UK
| | - Besir Topal
- g Department of Cardiothoracic Surgery , OLVG , Amsterdam , The Netherlands
| | - Alper Celik
- h Department of metabolic surgery , Metabolic Surgery Clinic , Istanbul , Turkey
| | - Surendra Ugale
- i Department of Surgery , Virinchi Hospitals , Hyderbad , India
| |
Collapse
|
24
|
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of Cerebrovascular Endothelial Cells: Prelude to Vascular Dementia. Front Aging Neurosci 2018; 10:376. [PMID: 30505270 PMCID: PMC6250852 DOI: 10.3389/fnagi.2018.00376] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia after Alzheimer's disease (AD), characterized by progressive cognitive impairment, memory loss, and thinking or speech problems. VaD is usually caused by cerebrovascular disease, during which, cerebrovascular endothelial cells (CECs) are vulnerable. CEC dysfunction occurs before the onset of VaD and can eventually lead to dysregulation of cerebral blood flow and blood-brain barrier damage, followed by the activation of glia and inflammatory environment in the brain. White matter, neuronal axons, and synapses are compromised in this process, leading to cognitive impairment. The present review summarizes the mechanisms underlying CEC impairment during hypoperfusion and pathological role of CECs in VaD. Through the comprehensive examination and summarization, endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway, Ras homolog gene family member A (RhoA) signaling pathway, and CEC-derived caveolin-1 (CAV-1) are proposed to serve as targets of new drugs for the treatment of VaD.
Collapse
Affiliation(s)
- Feixue Wang
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wolf Dieter Rausch
- Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
25
|
Giam B, Chu PY, Kuruppu S, Smith AI, Horlock D, Murali A, Kiriazis H, Du XJ, Kaye DM, Rajapakse NW. Serelaxin attenuates renal inflammation and fibrosis in a mouse model of dilated cardiomyopathy. Exp Physiol 2018; 103:1593-1602. [DOI: 10.1113/ep087189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/11/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Beverly Giam
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
- Central Clinical School; Monash University; Melbourne Victoria Australia
| | - Po-Yin Chu
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
| | - Sanjaya Kuruppu
- Biomedicine Discovery Institute; Department of Biochemistry & Molecular Biology; Monash University; Melbourne Victoria Australia
| | - A. Ian Smith
- Biomedicine Discovery Institute; Department of Biochemistry & Molecular Biology; Monash University; Melbourne Victoria Australia
| | - Duncan Horlock
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
| | - Aishwarya Murali
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
| | - David M. Kaye
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
- Department of Medicine; Monash University; Melbourne Victoria Australia
| | - Niwanthi W. Rajapakse
- Baker Heart and Diabetes Institute; Melbourne Victoria Australia
- School of Biomedical Sciences; University of Queensland; Brisbane Queensland Australia
| |
Collapse
|
26
|
Nafisa A, Gray SG, Cao Y, Wang T, Xu S, Wattoo FH, Barras M, Cohen N, Kamato D, Little PJ. Endothelial function and dysfunction: Impact of metformin. Pharmacol Ther 2018; 192:150-162. [PMID: 30056057 DOI: 10.1016/j.pharmthera.2018.07.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular and metabolic diseases remain the leading cause of morbidity and mortality worldwide. Endothelial dysfunction is a key player in the initiation and progression of cardiovascular and metabolic diseases. Current evidence suggests that the anti-diabetic drug metformin improves insulin resistance and protects against endothelial dysfunction in the vasculature. Hereby, we provide a timely review on the protective effects and molecular mechanisms of metformin in preventing endothelial dysfunction and cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Asma Nafisa
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Susan G Gray
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Yingnan Cao
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China
| | - Tinghuai Wang
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Feroza H Wattoo
- Department of Biochemistry, PMAS Arid Agriculture University, Shamasabad, Muree Road, Rawalpindi 4600, Pakistan..
| | - Michael Barras
- Dept. of Pharmacy, Princess Alexandra Hospital, 199 Ipswich Rd, Woolloongabba, QLD 4102, Australia.
| | - Neale Cohen
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia.
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| |
Collapse
|
27
|
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: 5] [Impact Index Per Article: 0.7] [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
|
28
|
Jelinic M, Marshall SA, Stewart D, Unemori E, Parry LJ, Leo CH. Peptide hormone relaxin: from bench to bedside. Am J Physiol Regul Integr Comp Physiol 2018; 314:R753-R760. [DOI: 10.1152/ajpregu.00276.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The peptide hormone relaxin has numerous roles both within and independent of pregnancy and is often thought of as a “pleiotropic hormone.” Relaxin targets several tissues throughout the body, and has many functions associated with extracellular matrix remodeling and the vasculature. This review considers the potential therapeutic applications of relaxin in cervical ripening, in vitro fertilization, preeclampsia, acute heart failure, ischemia-reperfusion, and cirrhosis. We first outline the animal models used in preclinical studies to progress relaxin into clinical trials and then discuss the findings from these studies. In many cases, the positive outcomes from preclinical animal studies were not replicated in human clinical trials. Therefore, the focus of this review is to evaluate the various animal models used to develop relaxin as a potential therapeutic and consider the limitations that must be addressed in future studies. These include the use of human relaxin in animals, duration of relaxin treatment, and the appropriateness of the clinical conditions being considered for relaxin therapy.
Collapse
Affiliation(s)
- Maria Jelinic
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Sarah A. Marshall
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Dennis Stewart
- Molecular Medicine Research Institute, Sunnyvale, California
| | | | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Chen Huei Leo
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
- Science and Maths Cluster, Singapore University of Technology and Design, Singapore
| |
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: 31] [Impact Index Per Article: 4.4] [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
|
Schiffner R, Lehmann T, Bischoff SJ, Zippelius T, Nistor M, Schmidt M. Pulmonary hemodynamic effects and pulmonary arterial compliance during hypovolemic shock and reinfusion with human relaxin-2 (serelaxin) treatment in a sheep model. Clin Hemorheol Microcirc 2018; 70:311-325. [PMID: 29710689 DOI: 10.3233/ch-180382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Previous studies on the recombinant form of human relaxin-2 (serelaxin) have shown a decrease of pulmonary hemodynamics after serelaxin injection. Currently, the effect of serelaxin treatment during hypovolemia in a large animal model remains mostly unknown. METHODS 12 sheep were randomly assigned to a sham or serelaxin (30μg/kg serelaxin) group and underwent right heart catheterization. 50% of the estimated total blood volume were removed to induce hypovolemia, and subsequently retransfused 20 min later (reinfusion). Blood gases, heart rate, peripheral and pulmonary arterial oxygen saturation, systolic, diastolic and mean values of both pulmonary artery pressure (PAP) and pulmonary capillary wedge pressure (PCW) were measured. Cardiac output (CO), pulmonary vascular resistance (PVR), pulmonary arterial compliance (PAcompl) and systemic vascular resistance (SVR) were calculated. RESULTS Hypovolemia and shock led to a similar decrease of PAP and PCW in both groups (p≤0.001). CO, SV and PAcompl decreased only in the control group (p≤0.05) and remained higher in the serelaxin-treated group. The results of this study suggest that serelaxin treatment did not negatively influence hemodynamic parameters during hypovolemic shock. CONCLUSION The main conclusion of this study is that cardiopulmonary adaption mechanisms are not critically altered by serelaxin administration during severe hypovolemia and retransfusion.
Collapse
Affiliation(s)
- René Schiffner
- Department of Orthopaedics, Jena University Hospital, Friedrich Schiller University, Jena, Germany.,Department of Neurology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Thomas Lehmann
- Institute of Medical Statistics, Computer Sciences and Documentation Science, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Sabine J Bischoff
- Central Animal Facility, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Timo Zippelius
- Department of Orthopaedics, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Marius Nistor
- Department of Neurology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
31
|
Deng A, Conrad K, Baylis C. Relaxin-mediated renal vasodilation in the rat is associated with falls in glomerular blood pressure. Am J Physiol Regul Integr Comp Physiol 2018; 314:R147-R152. [PMID: 29046312 PMCID: PMC5867670 DOI: 10.1152/ajpregu.00148.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/25/2017] [Accepted: 10/06/2017] [Indexed: 11/22/2022]
Abstract
Relaxin (RLX) is a pleiotropic peptide hormone with marked renal vasodilatory actions that are physiologically important during pregnancy. RLX also has potent antifibrotic actions and is being tested therapeutically in various fibrotic diseases, including chronic kidney disease (CKD). Since renal vasodilation may expose the glomerulus to increased blood pressure [glomerular capillary pressure (PGC)], which exacerbates progression of CKD, we assessed the glomerular hemodynamic actions of acute (0.89 µg·100 g body wt-1·h-1 iv over 75 min) and chronic (1.5 µg·100 g body wt-1·h-1 sc) administration of RLX. Both acute and chronic RLX produced marked renal vasodilation and increased renal plasma flow (RPF) in euvolemic, anesthetized male rats. Glomerular filtration rate also increased with RLX, but the magnitude of the rise was much less than the increase in RPF due to concomitant decreases in filtration fraction. The fall in filtration fraction was the result of significant decreases in PGC, despite a slight increase in mean arterial blood pressure (MAP) with acute RLX and no net change in MAP with chronic RLX. This fall in PGC occurred because of the "in-series" arrangement of the afferent and efferent arteriolar resistance vessels, which can regulate PGC independently of MAP. With both acute and chronic RLX, efferent arteriolar resistance vessels relaxed to a greater extent than afferent arteriolar resistance vessels, thus producing falls in PGC. Based on this finding, RLX has a beneficial hemodynamic impact on the kidney, which, together with the antifibrotic actions of RLX, suggests a strong therapeutic potential for use in CKD.
Collapse
Affiliation(s)
- Aihua Deng
- Departments of Physiology and Functional Genomics, University of Florida , Gainesville, Florida
| | - Kirk Conrad
- Departments of Obstetrics and Gynecology, University of Florida , Gainesville, Florida
- Department of Medicine, University of Florida , Gainesville, Florida
| | - Chris Baylis
- Departments of Obstetrics and Gynecology, University of Florida , Gainesville, Florida
| |
Collapse
|
32
|
Shao W, Rosales CB, Gonzalez C, Prieto MC, Navar LG. Effects of serelaxin on renal microcirculation in rats under control and high-angiotensin environments. Am J Physiol Renal Physiol 2018; 314:F70-F80. [PMID: 28978531 DOI: 10.1152/ajprenal.00201.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Serelaxin is a novel recombinant human relaxin-2 that has been investigated for the treatment of acute heart failure. However, its effects on renal function, especially on the renal microcirculation, remain incompletely characterized. Our immunoexpression studies localized RXFP1 receptors on vascular smooth muscle cells and endothelial cells of afferent arterioles and on principal cells of collecting ducts. Clearance experiments were performed in male and female normotensive rats and Ang II-infused male rats. Serelaxin increased mean arterial pressure slightly and significantly increased renal blood flow, urine flow, and sodium excretion rate. Group analysis of all serelaxin infusion experiments showed significant increases in GFR. During infusion with subthreshold levels of Ang II, serelaxin did not alter mean arterial pressure, renal blood flow, GFR, urine flow, or sodium excretion rate. Heart rates were elevated during serelaxin infusion alone (37 ± 5%) and in Ang II-infused rats (14 ± 2%). In studies using the in vitro isolated juxtamedullary nephron preparation, superfusion with serelaxin alone (40 ng/ml) significantly dilated afferent arterioles (10.8 ± 1.2 vs. 13.5 ± 1.1 µm) and efferent arterioles (9.9 ± 0.9 vs. 11.9 ± 1.0 µm). During Ang II superfusion, serelaxin did not alter afferent or efferent arteriolar diameters. During NO synthase inhibition (l-NNA), afferent arterioles also did not show any vasodilation during serelaxin infusion. In conclusion, serelaxin increased overall renal blood flow, urine flow, GFR, and sodium excretion and dilated the afferent and efferent arterioles in control conditions, but these effects were attenuated or prevented in the presence of exogenous Ang II and NO synthase inhibitors.
Collapse
Affiliation(s)
- Weijian Shao
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine , New Orleans, Louisiana
| | - Carla B Rosales
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine , New Orleans, Louisiana
| | - Camila Gonzalez
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine , New Orleans, Louisiana
| | - Minolfa C Prieto
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine , New Orleans, Louisiana
| | - L Gabriel Navar
- Department of Physiology, Tulane Hypertension and Renal Center of Excellence, Tulane University School of Medicine , New Orleans, Louisiana
| |
Collapse
|
33
|
The Role of Genetics in Peripartum Cardiomyopathy. J Cardiovasc Transl Res 2017; 10:437-445. [DOI: 10.1007/s12265-017-9764-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/25/2017] [Indexed: 10/19/2022]
|
34
|
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: 0.8] [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
|
35
|
Nonhoff J, Ricke-Hoch M, Mueller M, Stapel B, Pfeffer T, Kasten M, Scherr M, von Kaisenberg C, Bauersachs J, Haghikia A, Hilfiker-Kleiner D. Serelaxin treatment promotes adaptive hypertrophy but does not prevent heart failure in experimental peripartum cardiomyopathy. Cardiovasc Res 2017; 113:598-608. [PMID: 28453725 PMCID: PMC5412020 DOI: 10.1093/cvr/cvw245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 10/03/2016] [Accepted: 12/01/2016] [Indexed: 01/20/2023] Open
Abstract
AIMS Peripartum cardiomyopathy (PPCM) is a systolic left ventricular dysfunction developing in the peripartum phase in previously healthy women. Relaxin-2 is a pregnancy hormone with potential beneficial effects in heart failure patients. We evaluated Relaxin-2 as a potential diagnostic marker and/or a therapeutic agent in PPCM. METHODS AND RESULTS In healthy peripartum women, serum Relaxin-2 levels (measured by ELISA in the second half of pregnancy) were elevated showing a decreasing trend in the first postpartum week and returned to non-pregnant levels thereafter. In PPCM patients diagnosed in the first postpartum week, serum Relaxin-2 levels were lower compared to healthy postpartum stage-matched controls. In PPCM patients diagnosed later (0.5-10 months postpartum) Relaxin-2 levels were in the range of non-pregnant controls and not different from healthy postpartum stage-matched controls. In mice, serum Relaxin-1 (functional equivalent of human Relaxin-2) was increased late in pregnancy and rapidly cleared in the first postpartum week. In mice with PPCM due to a cardiomyocyte-specific knockout of STAT3 (CKO) neither low nor high dose of recombinant Relaxin-2 (serelaxin, sRlx-LD: 30 µg/kg/day; sRlx-HD: 300 µg/kg/day) affected cardiac fibrosis, inflammation and heart failure but sRlx-HD increased capillary/cardiomyocyte ratio. sRlx-HD significantly increased heart/body weight ratio and cardiomyocyte cross-sectional area in postpartum CKO and wild-type mice without changing the foetal gene expression program (ANP or β-MHC). sRlx-HD augmented plasma Prolactin levels in both genotypes, which induced cardiac activation of STAT5. In vitro analyses showed that Prolactin induces cardiomyocyte hypertrophy via activation of STAT5. CONCLUSION Although Relaxin-2 levels seemed lower in PPCM patients diagnosed early postpartum, we observed a high pregnancy-related variance of serum Relaxin-2 levels peripartum making it unsuitable as a biomarker for this condition. Supplementation with sRlx may contribute to angiogenesis and compensatory hypertrophy in the diseased heart, but the effects are not sufficient to prevent heart failure in an experimental PPCM model.
Collapse
Affiliation(s)
- Justus Nonhoff
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Mirco Mueller
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Britta Stapel
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Tobias Pfeffer
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Martina Kasten
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Michaela Scherr
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Constantin von Kaisenberg
- Department of Obstetrics and Gynecology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Arash Haghikia
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, Carl-Neuberg Str. 1, 30625 Hannover, Germany
| |
Collapse
|
36
|
Leo CH, Fernando DT, Tran L, Ng HH, Marshall SA, Parry LJ. Serelaxin Treatment Reduces Oxidative Stress and Increases Aldehyde Dehydrogenase-2 to Attenuate Nitrate Tolerance. Front Pharmacol 2017; 8:141. [PMID: 28377719 PMCID: PMC5359255 DOI: 10.3389/fphar.2017.00141] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/07/2017] [Indexed: 02/01/2023] Open
Abstract
Background: Glyceryl trinitrate (GTN) is a commonly prescribed treatment for acute heart failure patients. However, prolonged GTN treatment induces tolerance, largely due to increased oxidative stress and reduced aldehyde dehydrogenase-2 (ALDH-2) expression. Serelaxin has several vasoprotective properties, which include reducing oxidative stress and augmenting endothelial function. We therefore tested the hypothesis in rodents that serelaxin treatment could attenuate low-dose GTN-induced tolerance. Methods and Results: Co-incubation of mouse aortic rings ex vivo with GTN (10 μM) and serelaxin (10 nM) for 1 h, restored GTN responses, suggesting that serelaxin prevented the development of GTN tolerance. Male Wistar rats were subcutaneously infused with ethanol (control), low-dose GTN+placebo or low-dose GTN+serelaxin via osmotic minipumps for 3 days. Aortic vascular function and superoxide levels were assessed using wire myography and lucigenin-enhanced chemiluminescence assay respectively. Changes in aortic ALDH-2 expression were measured by qPCR and Western blot respectively. GTN+placebo infusion significantly increased superoxide levels, decreased ALDH-2 and attenuated GTN-mediated vascular relaxation. Serelaxin co-treatment with GTN significantly enhanced GTN-mediated vascular relaxation, reduced superoxide levels and increased ALDH-2 expression compared to GTN+placebo-treated rats. Conclusion: Our data demonstrate that a combination of serelaxin treatment with low dose GTN attenuates the development of GTN-induced tolerance by reducing superoxide production and increasing ALDH-2 expression in the rat aorta. We suggest that serelaxin may improve nitrate efficacy in a clinical setting.
Collapse
Affiliation(s)
- Chen Huei Leo
- School of BioSciences, The University of Melbourne, Parkville VIC, Australia
| | | | - Lillie Tran
- School of BioSciences, The University of Melbourne, Parkville VIC, Australia
| | - Hooi Hooi Ng
- School of BioSciences, The University of Melbourne, Parkville VIC, Australia
| | - Sarah A Marshall
- School of BioSciences, The University of Melbourne, Parkville VIC, Australia
| | - Laura J Parry
- School of BioSciences, The University of Melbourne, Parkville VIC, Australia
| |
Collapse
|
37
|
Toxic Dimethylarginines: Asymmetric Dimethylarginine (ADMA) and Symmetric Dimethylarginine (SDMA). Toxins (Basel) 2017; 9:toxins9030092. [PMID: 28272322 PMCID: PMC5371847 DOI: 10.3390/toxins9030092] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/04/2017] [Indexed: 02/07/2023] Open
Abstract
Asymmetric and symmetric dimethylarginine (ADMA and SDMA, respectively) are toxic, non-proteinogenic amino acids formed by post-translational modification and are uremic toxins that inhibit nitric oxide (NO) production and play multifunctional roles in many human diseases. Both ADMA and SDMA have emerged as strong predictors of cardiovascular events and death in a range of illnesses. Major progress has been made in research on ADMA-lowering therapies in animal studies; however, further studies are required to fill the translational gap between animal models and clinical trials in order to treat human diseases related to elevated ADMA/SDMA levels. Here, we review the reported impacts of ADMA and SDMA on human health and disease, focusing on the synthesis and metabolism of ADMA and SDMA; the pathophysiological roles of these dimethylarginines; clinical conditions and animal models associated with elevated ADMA and SDMA levels; and potential therapies against ADMA and SDMA. There is currently no specific pharmacological therapy for lowering the levels and counteracting the deleterious effects of ADMA and SDMA. A better understanding of the mechanisms underlying the impact of ADMA and SDMA on a wide range of human diseases is essential to the development of specific therapies against diseases related to ADMA and SDMA.
Collapse
|
38
|
Liu B, Ren KD, Peng JJ, Li T, Luo XJ, Fan C, Yang JF, Peng J. Suppression of NADPH oxidase attenuates hypoxia-induced dysfunctions of endothelial progenitor cells. Biochem Biophys Res Commun 2017; 482:1080-1087. [PMID: 27913300 DOI: 10.1016/j.bbrc.2016.11.161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 11/29/2016] [Indexed: 12/20/2022]
Abstract
NADPH oxidases (NOX) - derived reactive oxygen species (ROS) contribute to oxidative injury in hypoxia-induced pulmonary arterial hypertension. This study aims to evaluate the status of NOX in endothelial progenitor cells (EPCs) under hypoxic condition and to determine whether NOX inhibitors could attenuate hypoxia-induced dysfunctions of EPCs. EPCs were isolated from peripheral blood of SD rats and subjected to hypoxia (O2/N2/CO2, 1/94/5) for 24 h. The cells were collected for β-galactosidase or Hoechst staining, or for functional analysis (migration, adhesion and tube formation). The NOX expression, activity and H2O2 content in EPCs were measured. The results showed that hypoxia treatment promoted EPC senescence and apoptosis, accompanied by the deteriorated functions of EPCs (the reduced abilities in adhesion, migration and tube formation), as well as an increase in NOX2 and NOX4 expression, NOX activity and H2O2 production, these phenomena were attenuated by NOX inhibitors. Furthermore, administration of catalase could also improve the functions of hypoxia-treated EPCs. Based on these observations, we conclude that NOX-derived ROS contributes to the dysfunctions of EPCs under hypoxic condition. Thus, suppression of NOX may provide a novel strategy to improve endothelial functions in hypoxia-relevant diseases.
Collapse
Affiliation(s)
- Bin Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kai-Di Ren
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jing-Jie Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Tao Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, 410011, Changsha, China
| | - Jin-Fu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, 410011, Changsha, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
39
|
Serelaxin treatment reverses vascular dysfunction and left ventricular hypertrophy in a mouse model of Type 1 diabetes. Sci Rep 2017; 7:39604. [PMID: 28067255 PMCID: PMC5220363 DOI: 10.1038/srep39604] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/24/2016] [Indexed: 12/20/2022] Open
Abstract
Serelaxin prevents endothelial dysfunction in the mouse aorta ex vivo and inhibits apoptosis in cardiomyocytes under acute hyperglycaemia. Less is known about the effects of serelaxin in an in vivo mouse model of diabetes. Therefore, we tested the hypothesis in streptozotocin (STZ)-treated mice that serelaxin is able to reverse diabetes-induced vascular dysfunction and cardiac remodelling. Mice were divided into citrate buffer + placebo, STZ + placebo and STZ + serelaxin (0.5 mg/kg/d, 2 weeks) groups. After 12 weeks of diabetes, sensitivity to the endothelium-dependent agonist acetylcholine (ACh) was reduced in the mesenteric artery. This was accompanied by an enhanced vasoconstrictor prostanoid contribution and a decrease in endothelium-derived hyperpolarisation (EDH)-mediated relaxation. Serelaxin restored endothelial function by increasing nitric oxide (NO)-mediated relaxation but not EDH. It also normalised the contribution of vasoconstrictor prostanoids to endothelial dysfunction and suppressed diabetes-induced hyper-responsiveness of the mesenteric artery to angiotensin II. Similarly, diabetes reduced ACh-evoked NO-mediated relaxation in the aorta which was reversed by serelaxin. In the left ventricle, diabetes promoted apoptosis, hypertrophy and fibrosis; serelaxin treatment reversed this ventricular apoptosis and hypertrophy, but had no effect on fibrosis. In summary, serelaxin reversed diabetes-induced endothelial dysfunction by enhancing NO-mediated relaxation in the mouse vasculature and attenuating left ventricular hypertrophy and apoptosis.
Collapse
|
40
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 629] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
41
|
Li TB, Zhang JJ, Liu B, Luo XJ, Ma QL, Peng J. Dysfunction of endothelial progenitor cells in hyperlipidemic rats involves the increase of NADPH oxidase derived reactive oxygen species production. Can J Physiol Pharmacol 2016; 95:474-480. [PMID: 28177697 DOI: 10.1139/cjpp-2016-0142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
NADPH oxidase (NOX) is a major source of reactive oxygen species (ROS) in the body and it plays a key role in mediation of oxidative injury in the cardiovascular system. The purposes of this study are to evaluate the status of NOX in endothelial progenitor cells (EPCs) of hyperlipidemic rats and to determine whether NOX-derived ROS promotes the dysfunction of EPCs. The rats were fed on a high-fat diet for 8 weeks to establish a hyperlipidemic rat model, which showed the increased plasma lipids and the impaired functions of circulating EPCs (including the reduced abilities in migration and adhesion) accompanied by an increase in NOX activity and ROS production. Next, EPCs were isolated from normal rats and they were treated with oxidized low-density lipoprotein (ox-LDL) (100 μg/mL) for 24 h to induce a dysfunctional model in vitro. In agreement with our findings in vivo, ox-LDL treatment increased the dysfunctions of EPCs concomitant with an increase in NOX activity and ROS production; these phenomena were reversed by the NOX inhibitor. Based on these observations, we conclude that NOX-derived ROS involved in the dysfunctions of circulating EPCs in hyperlipidemic rats and inhibition of NOX might provide a novel strategy to improve EPC functions in hyperlipidemia.
Collapse
Affiliation(s)
- Ting-Bo Li
- a Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Jie-Jie Zhang
- a Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Bin Liu
- a Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiu-Ju Luo
- b Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Qi-Lin Ma
- c Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jun Peng
- a Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.,d Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
42
|
Bischoff SJ, Schmidt M, Lehmann T, Irintchev A, Schubert H, Jung C, Schwab M, Huber O, Matziolis G, Schiffner R. Increase of cortical cerebral blood flow and further cerebral microcirculatory effects of Serelaxin in a sheep model. Am J Physiol Heart Circ Physiol 2016; 311:H613-20. [PMID: 27402664 DOI: 10.1152/ajpheart.00118.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 07/02/2016] [Indexed: 12/17/2022]
Abstract
Serelaxin, recombinant human relaxin-2, modulates endothelial vasodilatory functionality and is under evaluation for treatment of acute heart failure. Little is known about acute effects on cerebral perfusion. We tested the hypothesis that Serelaxin might also have effects on the cerebral microcirculation in a sheep model, which resembles human brain structure quite well. We used laser Doppler flowmetry and sidestream dark-field (SDF) imaging techniques, which are reliable tools to continuously assess dynamic changes in cerebral perfusion. Laser Doppler flowmetry shows that bolus injection of 30 μg Serelaxin/kg body wt induces an increase (P = 0.006) to roughly 150% of cortical cerebral blood flow (CBF), whereas subcortical CBF remains unchanged (P = 0.688). The effects on area-dependent CBF were significantly different after the bolus injection (P = 0.042). Effects on cortical CBF were further confirmed by SDF imaging. The bolus injection of Serelaxin increased total vessel density to 127% (P = 0.00046), perfused vessel density to 145% (P = 0.024), and perfused capillary density to 153% (P = 0.024). Western blotting confirmed the expression of relaxin receptors RXFP1 and truncated RXFP2-variants in the respective brain regions, suggesting a possible contribution of RXFP1 on the effects of Serelaxin. In conclusion, the injection of a high dose of Serelaxin exerts quick effects on the cerebral microcirculation. Therefore, Serelaxin might be suitable to improve cortical microcirculation and exert neuroprotective effects in clinically relevant scenarios that involve cortical hypoperfusion. These findings need to be confirmed in relevant experimental settings involving cerebral cortical hypoperfusion and can possibly be translated into clinical practice.
Collapse
Affiliation(s)
- Sabine J Bischoff
- Institute for Laboratory Animal Science and Welfare, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Thomas Lehmann
- Institute of Medical Statistics, Computer Sciences and Documentation Science, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Andrey Irintchev
- Department of Otorhinolaryngology, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Harald Schubert
- Institute for Laboratory Animal Science and Welfare, 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
| | - Matthias Schwab
- Department of Neurology, Jena University Hospital, Friedrich Schiller University, Jena, Germany; and
| | - Otmar Huber
- Institute for Biochemistry II, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Georg Matziolis
- Orthopaedic Department, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - René Schiffner
- Department of Neurology, Jena University Hospital, Friedrich Schiller University, Jena, Germany; and Orthopaedic Department, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
43
|
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: 3.2] [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
|
44
|
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: 1.8] [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
|
45
|
Marshall SA, Leo CH, Senadheera SN, Girling JE, Tare M, Parry LJ. Relaxin deficiency attenuates pregnancy-induced adaptation of the mesenteric artery to angiotensin II in mice. Am J Physiol Regul Integr Comp Physiol 2016; 310:R847-57. [DOI: 10.1152/ajpregu.00506.2015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/22/2016] [Indexed: 12/11/2022]
Abstract
Pregnancy is associated with reduced peripheral vascular resistance, underpinned by changes in endothelial and smooth muscle function. Failure of the maternal vasculature to adapt correctly leads to serious pregnancy complications, such as preeclampsia. The peptide hormone relaxin regulates the maternal renal vasculature during pregnancy; however, little is known about its effects in other vascular beds. This study tested the hypothesis that functional adaptation of the mesenteric and uterine arteries during pregnancy will be compromised in relaxin-deficient ( Rln−/−) mice. Smooth muscle and endothelial reactivity were examined in small mesenteric and uterine arteries of nonpregnant (estrus) and late-pregnant ( day 17.5) wild-type ( Rln+/+) and Rln−/− mice using wire myography. Pregnancy per se was associated with significant reductions in contraction to phenylephrine, endothelin-1, and ANG II in small mesenteric arteries, while sensitivity to endothelin-1 was reduced in uterine arteries of Rln+/+ mice. The normal pregnancy-associated attenuation of ANG II-mediated vasoconstriction in mesenteric arteries did not occur in Rln−/− mice. This adaptive failure was endothelium-independent and did not result from altered expression of ANG II receptors or regulator of G protein signaling 5 ( Rgs5) or increases in reactive oxygen species generation. Inhibition of nitric oxide synthase with l-NAME enhanced ANG II-mediated contraction in mesenteric arteries of both genotypes, whereas blockade of prostanoid production with indomethacin only increased ANG II-induced contraction in arteries of pregnant Rln+/+ mice. In conclusion, relaxin deficiency prevents the normal pregnancy-induced attenuation of ANG II-mediated vasoconstriction in small mesenteric arteries. This is associated with reduced smooth muscle-derived vasodilator prostanoids.
Collapse
Affiliation(s)
- Sarah A. Marshall
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Chen Huei Leo
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Jane E. Girling
- Gynaecology Research Centre, Department of Obstetrics and Gynaecology, The University of Melbourne and Royal Women's Hospital, Parkville, Victoria, Australia; and
| | - Marianne Tare
- Department of Physiology, Monash University, Victoria, Australia; and
- School of Rural Health, Monash University, Victoria, Australia
| | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
46
|
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: 3.0] [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
|
47
|
Díez J, Ruilope LM. Serelaxin for the treatment of acute heart failure: a review with a focus on end-organ protection. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2015; 2:119-30. [PMID: 27418970 PMCID: PMC4853824 DOI: 10.1093/ehjcvp/pvv046] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/26/2015] [Indexed: 12/15/2022]
Abstract
Acute heart failure (AHF) is a complex clinical syndrome characterized by fluid overload and haemodynamic abnormalities (short-term clinical consequences) and the development of end-organ damage (long-term consequences). Current therapies for the treatment of AHF, such as loop diuretics and vasodilators, help to relieve haemodynamic imbalance and congestion, but have not been shown to prevent (and may even contribute to) end-organ damage, or to provide long-term clinical benefit. Serelaxin is the recombinant form of human relaxin-2, a naturally occurring hormone involved in mediating haemodynamic changes during pregnancy. Preclinical and clinical studies have investigated the effects mediated by serelaxin and the suitability of this agent for the treatment of patients with AHF. Data suggest that serelaxin acts via multiple pathways to improve haemodynamics at the vascular, cardiac, and renal level and provide effective congestion relief. In addition, this novel agent may protect the heart, kidneys, and liver from damage by inhibiting inflammation, oxidative stress, cell death, and tissue fibrosis, and stimulating angiogenesis. Serelaxin may therefore improve both short- and long-term outcomes in patients with AHF. In this review, we examine the unique mechanisms underlying the potential benefits of serelaxin for the treatment of AHF, in particular, those involved in mediating end-organ protection.
Collapse
Affiliation(s)
- Javier Díez
- Program of Cardiovascular Diseases, Centre for Applied Medical Research and Department of Cardiology and Cardiac Surgery, University of Navarra Clinic, University of Navarra, Av. Pío XII 55, Pamplona 31008, Spain
| | - Luis M Ruilope
- Research Institute, Hypertension Unit, Hospital 12 de Octubre and Department of Public Health and Preventive Medicine, University Autónoma, Madrid, Spain
| |
Collapse
|
48
|
Tietjens J, Teerlink JR. Serelaxin and acute heart failure. Heart 2015; 102:95-9. [DOI: 10.1136/heartjnl-2014-306786] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/02/2015] [Indexed: 01/11/2023] Open
|
49
|
Ng HH, Jelinic M, Parry LJ, Leo CH. Increased superoxide production and altered nitric oxide-mediated relaxation in the aorta of young but not old male relaxin-deficient mice. Am J Physiol Heart Circ Physiol 2015; 309:H285-96. [DOI: 10.1152/ajpheart.00786.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 05/06/2015] [Indexed: 11/22/2022]
Abstract
The vascular effects of exogenous relaxin (Rln) treatment are well established and include decreased myogenic reactivity and enhanced relaxation responses to vasodilators in small resistance arteries. These vascular responses are reduced in older animals, suggesting that Rln is less effective in mediating arterial function with aging. The present study investigated the role of endogenous Rln in the aorta and the possibility that vascular dysfunction occurs more rapidly with aging in Rln-deficient ( Rln−/−) mice. We compared vascular function and underlying vasodilatory pathways in the aorta of male wild-type ( Rln+/+) and Rln−/− mice at 4 and 16 mo of age using wire myography. Superoxide production, but not nitrotyrosine or NADPH oxidase expression, was significantly increased in the aorta of young Rln−/− mice, whereas endothelial nitric oxide (NO) synthase and basal NO availability were both significantly decreased compared with Rln+/+ mice. In the presence of the cyclooxygenase inhibitor indomethacin, sensitivity to ACh was significantly decreased in young Rln−/− mice, demonstrating altered NO-mediated relaxation that was normalized in the presence of a membrane-permeable SOD or ROS scavenger. These vascular phenotypes were not exacerbated in old Rln−/− mice and, in most cases, did not differ significantly from old Rln+/+ mice. Despite the vascular phenotypes in Rln−/− mice, endothelium-dependent and -independent vasodilation were not adversely affected. Our data show a role for endogenous Rln in reducing superoxide production and maintaining NO availability in the aorta but also demonstrate that Rln deficiency does not compromise vascular function in this artery or exacerbate endothelial dysfunction associated with aging.
Collapse
Affiliation(s)
- Hooi H. Ng
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Maria Jelinic
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura J. Parry
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Chen-Huei Leo
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
50
|
Metabonomic analysis of potential biomarkers and drug targets involved in diabetic nephropathy mice. Sci Rep 2015; 5:11998. [PMID: 26149603 PMCID: PMC4493693 DOI: 10.1038/srep11998] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/11/2015] [Indexed: 11/08/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the lethal manifestations of diabetic systemic microvascular disease. Elucidation of characteristic metabolic alterations during diabetic progression is critical to understand its pathogenesis and identify potential biomarkers and drug targets involved in the disease. In this study, (1)H nuclear magnetic resonance ((1)H NMR)-based metabonomics with correlative analysis was performed to study the characteristic metabolites, as well as the related pathways in urine and kidney samples of db/db diabetic mice, compared with age-matched wildtype mice. The time trajectory plot of db/db mice revealed alterations, in an age-dependent manner, in urinary metabolic profiles along with progression of renal damage and dysfunction. Age-dependent and correlated metabolite analysis identified that cis-aconitate and allantoin could serve as biomarkers for the diagnosis of DN. Further correlative analysis revealed that the enzymes dimethylarginine dimethylaminohydrolase (DDAH), guanosine triphosphate cyclohydrolase I (GTPCH I), and 3-hydroxy-3-methylglutaryl-CoA lyase (HMG-CoA lyase) were involved in dimethylamine metabolism, ketogenesis and GTP metabolism pathways, respectively, and could be potential therapeutic targets for DN. Our results highlight that metabonomic analysis can be used as a tool to identify potential biomarkers and novel therapeutic targets to gain a better understanding of the mechanisms underlying the initiation and progression of diseases.
Collapse
|