1
|
Aubdool AA, Moyes AJ, Pérez-Ternero C, Baliga RS, Sanghera J, Syed MT, Jaigirdah K, Panesar AK, Tsui JC, Li Y, Vasquez HG, Shen YH, LeMaire SA, Raffort-Lareyre J, Mallat Z, Lu HS, Daugherty A, Hobbs AJ. Endothelium- and Fibroblast-Derived C-Type Natriuretic Peptide Prevents the Development and Progression of Aortic Aneurysms. Arterioscler Thromb Vasc Biol 2025. [PMID: 40177775 DOI: 10.1161/atvbaha.124.322350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Thoracic aortic aneurysm (AA) and abdominal AA are life-threatening diseases characterized by dilation, inflammation, and structural weakness; development of pharmacological therapies is desperately needed. CNP (C-type natriuretic peptide) plays a key role in vascular homeostasis, mediating vasodilator, anti-inflammatory, and antiatherogenic actions. Because such processes drive AA, we determined the role of endogenous CNP in offsetting pathogenesis. METHODS Tissue from patients with AA was analyzed to determine the consequences on CNP signaling. Ascending and suprarenal aortic diameters were assessed at baseline and following Ang II (angiotensin II; 1.44 mg/kg per day) infusion in wild-type, endothelium-restricted (ecCNP-/-), fibroblast-restricted (fbCNP-/-), global CNP (gbCNP-/-), or global NPR-C-/- mice infected with an adeno-associated virus expressing a proprotein convertase subtilisin/kexin type 9 gain-of-function mutation or backcrossed to an apoE-/- background. At 28 days, aortas were harvested for RT-qPCR and histological analyses. CNP (0.2 mg/kg per day) was infused to rescue any adverse phenotype. RESULTS Aneurysmal tissue from patients with thoracic AA and abdominal AA revealed that CNP and NPR-C (natriuretic peptide receptor-C) expression were overtly perturbed. ecCNP-/-, fbCNP-/-, and gbCNP-/- mice exhibited an aggravated phenotype compared with wild-type mice in both ascending and suprarenal aortas, exemplified by greater dilation, fibrosis, elastin degradation, and macrophage infiltration. CNP and NPR-C expression was also dysregulated in murine thoracic AA and abdominal AA, accompanied by increased accumulation of mRNA encoding markers of inflammation, extracellular matrix remodeling/calcification, fibrosis, and apoptosis. CNP also prevented activation of isolated macrophages and vascular smooth muscle cells. An essentially identical phenotype was observed in NPR-C-/- mice and while administration of CNP protected against disease severity in wild-type animals, this phenotypic rescue was not apparent in NPR-C-/- mice. CONCLUSIONS Endothelium- and fibroblast-derived CNP, via NPR-C activation, plays important roles in attenuating AA formation by preserving aortic structure and function. Therapeutic strategies aimed at mimicking CNP bioactivity hold potential to reduce the need for surgical intervention.
Collapse
Affiliation(s)
- Aisah A Aubdool
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Amie J Moyes
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Cristina Pérez-Ternero
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Reshma S Baliga
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Jasspinder Sanghera
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - M Taaha Syed
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Kareemah Jaigirdah
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Anmolpreet K Panesar
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| | - Janice C Tsui
- Division of Surgery and Interventional Science, University College London and Royal Free London NHS Foundation Trust, United Kingdom (J.C.T.)
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Hernan G Vasquez
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
| | - Scott A LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX (Y.L., H.G.V., Y.H.S., S.A.L.)
- Geisinger Research Institute and Heart & Vascular Institute (S.A.L.)
| | | | - Ziad Mallat
- Division of Cardiovascular Medicine, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge, United Kingdom (Z.M.)
| | - Hong S Lu
- Department of Physiology, Saha Cardiovascular Research Center, Saha Aortic Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Alan Daugherty
- Department of Physiology, Saha Cardiovascular Research Center, Saha Aortic Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Adrian J Hobbs
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Barts & The London Hospitals, Queen Mary University of London, United Kingdom (A.A.A., A.J.M., C.P.-T., R.S.B., J.S., M.T.S., K.J., A.K.P., A.J.H.)
| |
Collapse
|
2
|
Xiao F, Qi J, Ma S, Sun L, Sun Y. Research Progress on the Role and Mechanism in the Change of Cardiac Structure and Function of Cardiac Fibrosis in the Elderly. Cardiol Rev 2025:00045415-990000000-00457. [PMID: 40167333 DOI: 10.1097/crd.0000000000000911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Heart failure is closely related to aging. Elderly patients with heart failure are often able to retain normal systolic function, manifested by left ventricular hypertrophy with decreased diastolic function. Relevant studies have shown that age-related cardiac fibrosis plays an important role in the pathogenesis of cardiac diastolic heart failure. Activation of fibroblasts in the heart, the acquisition of a pro-fibrotic phenotype, and age-dependent accumulation of collagen can lead to progressive increases in myocardial stiffness and impaired diastolic function. The renin-angiotensin-aldosterone system, reactive oxygen species, and angiotensin II are closely related to fibrotic remodeling of the heart in the elderly, and their pro-fibrotic effects may be mainly mediated by transforming growth factor β. In this review, we summarize the research progress of the role and mechanism of cardiac fibrosis in the structural and functional changes of the elderly.
Collapse
Affiliation(s)
- Fei Xiao
- From the Department of Cardiology, Zi-Bo Central Hospital, Zibo, China
| | - Jia Qi
- From the Department of Cardiology, Zi-Bo Central Hospital, Zibo, China
| | - Shanshan Ma
- Department of Geriatrics, Zi-Bo Central Hospital, Zibo, China
| | - Lei Sun
- Shinva Medical Instrument Co.Ltd, Zibo, China
| | - Yongchen Sun
- Department of Geriatrics, Zi-Bo Central Hospital, Zibo, China
| |
Collapse
|
3
|
Perez-Ternero C, Li W, Aubdool AA, Goldin RD, Loy J, Devalia K, Alazawi W, Hobbs AJ. Endogenous C-type natriuretic peptide offsets the pathogenesis of steatohepatitis, hepatic fibrosis, and portal hypertension. PNAS NEXUS 2025; 4:pgae579. [PMID: 39816244 PMCID: PMC11734523 DOI: 10.1093/pnasnexus/pgae579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), hepatic fibrosis, and portal hypertension constitute an increasing public health problem due to the growing prevalence of obesity and diabetes. C-type natriuretic peptide (CNP) is an endogenous regulator of cardiovascular homeostasis, immune cell reactivity, and fibrotic disease. Thus, we investigated a role for CNP in the pathogenesis of MASLD. Wild-type (WT), global CNP (gbCNP-/-), and natriuretic peptide receptor-C (NPR-C-/-) knockout mice were fed a choline-deficient defined amino acid diet or administered CCl4. Liver damage was assessed by histological and biochemical analyses, with steatosis and portal vein size determined by ultrasound. Portal vein pressure and reactivity were measured in vivo and ex vivo, respectively. Pharmacological CNP delivery was used to evaluate prospective therapeutic benefit, and plasma CNP concentration was compared in controls and patients with cirrhosis. Circulating CNP concentration was lower in patients with cirrhosis compared with controls. gbCNP-/- mice were more susceptible, versus WT, to advanced steatohepatitis and hepatic fibrosis, characterized by increased immune cell infiltration, fibrosis, ballooning, plasma alanine aminotransferase concentration, and up-regulation of markers driving these processes. gbCNP-/- mice had increased portal vein diameter and pressure, underpinned by CNP insensitivity. NPR-C-/- animals recapitulated, comparatively, the exaggerated pathogenic phenotype in gbCNP-/- mice, whereas CNP reduced hepatic stellate cell proliferation via NPR-B-dependent inhibition of extracellular signal-related kinase 1/2. Administration of CNP reversed many aspects of disease severity. These data define a new intrinsic role for CNP in offsetting the pathogenesis of MASLD, hepatic fibrosis, and portal hypertension and the potential for targeting CNP signaling for treating these disorders.
Collapse
Affiliation(s)
- Cristina Perez-Ternero
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Wenhao Li
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Aisah A Aubdool
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | - Robert D Goldin
- Centre for Pathology, St Mary’s Hospital, Imperial College, London W2 1NY, United Kingdom
| | - John Loy
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - Kalpana Devalia
- Bariatric Surgery Department, Homerton University Hospital, Homerton Row, London E9 6SR, United Kingdom
| | - William Alazawi
- Barts Liver Centre, Blizard Institute, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom
| | - Adrian J Hobbs
- Faculty of Medicine and Dentistry, William Harvey Research Institute, Barts and The London, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom
| |
Collapse
|
4
|
Zhazykbayeva S, Budde H, Kaçmaz M, Zemedie Y, Osman H, Hassoun R, Jaquet K, Akin I, El-Battrawy I, Herwig M, Hamdani N. Exploring PKG signaling as a therapeutic avenue for pressure overload, ischemia, and HFpEF. Expert Opin Ther Targets 2024; 28:857-873. [PMID: 39329430 DOI: 10.1080/14728222.2024.2400093] [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: 01/10/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION Heart failure (HF) is a complex and heterogeneous syndrome resulting from any diastolic or systolic dysfunction of the cardiac muscle. In addition to comorbid conditions, pressure overload, and myocardial ischemia are associated with cardiac remodeling which manifests as extracellular matrix (ECM) perturbations, impaired cellular responses, and subsequent ventricular dysfunction. AREAS COVERED The current review discusses the main aspects of the cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) pathway (cGMP-PKG) pathway modulators and highlights the promising outcomes of its novel pharmacological boosters. EXPERT OPINION Among several signaling pathways involved in the pathogenesis of pressure overload, ischemia and HF with preserved ejection fraction (HFpEF) is cGMP-PKG pathway. This pathway plays a pivotal role in the regulation of cardiac contractility, and modulation of cGMP-PKG signaling, contributing to the development of the diseases. Ventricular cardiomyocytes of HF patients and animal models are known to exhibit reduced cGMP levels and disturbed cGMP signaling including hypophosphorylation of PKG downstream targets. However, restoration of cGMP-PKG signaling improves cardiomyocyte function and promotes cardioprotective effects.
Collapse
Affiliation(s)
- S Zhazykbayeva
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - H Budde
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - M Kaçmaz
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Budapest, Hungary
| | - Y Zemedie
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - H Osman
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - R Hassoun
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - K Jaquet
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - I Akin
- Medical University Mannheim, Medical Faculty, Mannheim University, Heidelberg, Germany
| | - I El-Battrawy
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University, Bochum, Germany
| | - M Herwig
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - N Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, Budapest, Hungary
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University, Bochum, Germany
- Department of Physiology, Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
5
|
Fato BR, de Alwis N, Beard S, Binder NK, Pritchard N, Kaitu'u-Lino TJ, Bubb KJ, Hannan NJ. Exploring the Therapeutic Potential of C-Type Natriuretic Peptide for Preeclampsia. Hypertension 2024; 81:1883-1894. [PMID: 39016006 DOI: 10.1161/hypertensionaha.124.22820] [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: 01/29/2024] [Accepted: 06/21/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Preeclampsia is a serious condition of pregnancy, complicated by aberrant maternal vascular dysfunction. CNP (C-type natriuretic peptide) contributes to vascular homeostasis, acting through NPR-B (natriuretic peptide receptor-B) and NPR-C (natriuretic peptide receptor-C). CNP mitigates vascular dysfunction of arteries in nonpregnant cohorts; this study investigates whether CNP can dilate maternal arteries in ex vivo preeclampsia models. METHODS Human omental arteries were dissected from fat biopsies collected during cesarean section. CNP, NPR-B, and NPR-C mRNA expression was assessed in arteries collected from pregnancies complicated by preeclampsia (n=6) and normotensive controls (n=11). Using wire myography, we investigated the effects of CNP on dilation of arteries from normotensive pregnancies. Arteries were preconstricted with either serum from patients with preeclampsia (n=6) or recombinant ET-1 (endothelin-1; vasoconstrictor elevated in preeclampsia; n=6) to model vasoconstriction associated with preeclampsia. Preconstricted arteries were treated with recombinant CNP (0.001-100 µmol/L) or vehicle and vascular relaxation assessed. In further studies, arteries were preincubated with NPR-B (5 µmol/L) and NPR-C (10 µmol/L) antagonists before serum-induced constriction (n=4-5) to explore mechanistic signaling. RESULTS CNP, NPR-B, and NPR-C mRNAs were not differentially expressed in omental arteries from preeclamptic pregnancies. CNP potently stimulated maternal artery vasorelaxation in our model of preeclampsia (using preeclamptic serum). Its vasodilatory actions were driven through the activation of NPR-B predominantly; antagonism of this receptor alone dampened CNP vasorelaxation. Interestingly, CNP did not reduce ET-1-driven omental artery constriction. CONCLUSIONS Collectively, these data suggest that enhancing CNP signaling through NPR-B offers a potential therapeutic strategy to reduce systemic vascular constriction in preeclampsia.
Collapse
Affiliation(s)
- Bianca R Fato
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natasha de Alwis
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Sally Beard
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natalie K Binder
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| | - Kristen J Bubb
- Department of Physiology, Biomedicine Discovery Institute (K.J.B.), Monash University, Clayton, Victoria, Australia
- Victorian Heart Institute, Faculty of Medicine, Nursing and Health Sciences (K.J.B.), Monash University, Clayton, Victoria, Australia
| | - Natalie J Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group (B.R.F., N.d.A., S.B., N.K.B., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
- Department of Obstetrics, Gynecology and Newborn Health, Mercy Hospital for Women (B.R.F., N.d.A., S.B., N.K.B., N.P., T.J.K.-L., N.J.H.), University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
6
|
Lu Z, Verginadis I, Kumazoe M, Castillo GM, Yao Y, Guerra RE, Bicher S, You M, McClung G, Qiu R, Xiao Z, Miao Z, George SS, Beiting DP, Nojiri T, Tanaka Y, Fujimura Y, Onda H, Hatakeyama Y, Nishimoto-Ashfield A, Bykova K, Guo W, Fan Y, Buynov NM, Diehl JA, Stanger BZ, Tachibana H, Gade TP, Puré E, Koumenis C, Bolotin EM, Fuchs SY. Modified C-type natriuretic peptide normalizes tumor vasculature, reinvigorates antitumor immunity, and improves solid tumor therapies. Sci Transl Med 2024; 16:eadn0904. [PMID: 39167664 PMCID: PMC11866103 DOI: 10.1126/scitranslmed.adn0904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/23/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
Deficit of oxygen and nutrients in the tumor microenvironment (TME) triggers abnormal angiogenesis that produces dysfunctional and leaky blood vessels, which fail to adequately perfuse tumor tissues. Resulting hypoxia, exacerbation of metabolic disturbances, and generation of an immunosuppressive TME undermine the efficacy of anticancer therapies. Use of carefully scheduled angiogenesis inhibitors has been suggested to overcome these problems and normalize the TME. Here, we propose an alternative agonist-based normalization approach using a derivative of the C-type natriuretic peptide (dCNP). Multiple gene expression signatures in tumor tissues were affected in mice treated with dCNP. In several mouse orthotopic and subcutaneous solid tumor models including colon and pancreatic adenocarcinomas, this well-tolerated agent stimulated formation of highly functional tumor blood vessels to reduce hypoxia. Administration of dCNP also inhibited stromagenesis and remodeling of the extracellular matrix and decreased tumor interstitial fluid pressure. In addition, treatment with dCNP reinvigorated the antitumor immune responses. Administration of dCNP decelerated growth of primary mouse tumors and suppressed their metastases. Moreover, inclusion of dCNP into the chemo-, radio-, or immune-therapeutic regimens increased their efficacy against solid tumors in immunocompetent mice. These results demonstrate the proof of principle for using vasculature normalizing agonists to improve therapies against solid tumors and characterize dCNP as the first in class among such agents.
Collapse
Affiliation(s)
- Zhen Lu
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ioannis Verginadis
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Motofumi Kumazoe
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | | | - Yao Yao
- PharmaIN Corp., Bothell, WA 98011, USA
| | | | - Sandra Bicher
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Menghao You
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - George McClung
- Dept. of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rong Qiu
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zebin Xiao
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhen Miao
- Dept. of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Subin S. George
- Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P. Beiting
- Dept. of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Takashi Nojiri
- Dept. of General Thoracic Surgery, Higashiosaka City Medical Center, Higashiosaka, 578-8588, Japan
| | - Yasutake Tanaka
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yoshinori Fujimura
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Hiroaki Onda
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yui Hatakeyama
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | | | | | - Wei Guo
- Dept. of Biology, School of Arts & Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yi Fan
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - J. Alan Diehl
- Dept. of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Ben Z. Stanger
- Dept. of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hirofumi Tachibana
- Div. of Applied Biological Chemistry, Dept. of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Terence P. Gade
- Dept. of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ellen Puré
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Constantinos Koumenis
- Dept. of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Serge Y. Fuchs
- Dept. of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Ma X, Peddibhotla S, Zheng Y, Pan S, Mehta A, Moroni DG, Chen QY, Ma X, Burnett JC, Malany S, Sangaralingham SJ. Discovery of small molecule guanylyl cyclase B receptor positive allosteric modulators. PNAS NEXUS 2024; 3:pgae225. [PMID: 38894878 PMCID: PMC11185183 DOI: 10.1093/pnasnexus/pgae225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
Myocardial fibrosis is a pathological hallmark of cardiovascular disease (CVD), and excessive fibrosis can lead to new-onset heart failure and increased mortality. Currently, pharmacological therapies for myocardial fibrosis are limited, highlighting the need for novel therapeutic approaches. The particulate guanylyl cyclase B (GC-B) receptor possesses beneficial antifibrotic actions through the binding of its natural ligand C-type natriuretic peptide (CNP) and the generation of the intracellular second messenger, cyclic guanosine 3',5'-monophosphate (cGMP). These actions include the suppression of fibroblast proliferation and reduction in collagen synthesis. With its abundant expression on fibroblasts, the GC-B receptor has emerged as a key molecular target for innovative CVD therapeutics. However, small molecules that can bind and potentiate the GC-B/cGMP pathway have yet to be discovered. From a cell-based high-throughput screening initiative of the NIH Molecular Libraries Small Molecule Repository and hit-to-lead evolution based on a series of structure-activity relationships, we report the successful discovery of MCUF-42, a GC-B-targeted small molecule that acts as a positive allosteric modulator (PAM). Studies herein support MCUF-42's ability to enhance the binding affinity between GC-B and CNP. Moreover, MCUF-42 potentiated cGMP levels induced by CNP in human cardiac fibroblasts (HCFs) and notably also enhanced the inhibitory effect of CNP on HCF proliferation. Together, our findings highlight that MCUF-42 is a small molecule that can modulate the GC-B/cGMP signaling pathway, potentially enhancing the antifibrotic actions of CNP. Thus, these data underscore the continued development of GC-B small molecule PAMs as a novel therapeutic strategy for targeting cardiac fibrosis and CVD.
Collapse
Affiliation(s)
- Xiao Ma
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Ye Zheng
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Shuchong Pan
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Alka Mehta
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - Dante G Moroni
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Qi-Yin Chen
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL 32610, USA
| | - Xiaoyu Ma
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Siobhan Malany
- Department of Pharmacodynamics, University of Florida, Gainesville, FL 32610, USA
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Ensho T, Hino J, Ueda Y, Miyazato M, Iwakura H. Vascular endothelial cell-specific overexpression of CNP did not improve liver fibrosis in HFFCD-induced NASH, but did improve renal lesions. Peptides 2024; 172:171146. [PMID: 38157939 DOI: 10.1016/j.peptides.2023.171146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Mice with endothelial-cell-specific overexpression of C-type natriuretic peptide (E-CNP Tg mice) were shown to be protected against hepatic fibrosis and inflammation induced by high fat diet (HFD) feeding, with improved insulin sensitivity and attenuated weight gain. A recently developed high-fat, high-fructose, high-cholesterol diet (HFFCD) is considered to be a superior model to HFD, owing to the resemblance to human non-alcoholic steatohepatitis (NASH). In this study, we therefore aimed to reveal whether these previous findings with E-CNP Tg mice on HFD can be observed in a newly developed NASH model. Patients with NASH have been suggested to be at higher risk of developing chronic kidney disease, so we also assessed the kidney histology of these mice. After 8 months of HFFCD feeding, the livers of E-CNP Tg mice and controls showed progressive fibrosis, which resembled the features of human NASH. However, no significant differences were observed in NAFLD activity scores between E-CNP Tg mice and controls, although there was a tendency for improvement in E-CNP Tg mice. The reduced levels of GCB, a receptor for CNP, may have weakened the action of CNP in the current model. In the kidneys, HFFCD showed glomerular hypertrophy and tubular atrophy in the cortical region, which were suppressed in E-CNP Tg mice. The present study did not prove the therapeutic effect of CNP on NASH in the HFFCD model, but provided evidence of its potential beneficial effects on NASH-associated renal damage.
Collapse
Affiliation(s)
- Takuya Ensho
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, Wakayama, Japan
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Yoko Ueda
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, Wakayama, Japan
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Hiroshi Iwakura
- Department of Pharmacotherapeutics, School of Pharmaceutical Science, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
9
|
Meng L, Lu Y, Wang X, Cheng C, Xue F, Xie L, Zhang Y, Sui W, Zhang M, Zhang Y, Zhang C. NPRC deletion attenuates cardiac fibrosis in diabetic mice by activating PKA/PKG and inhibiting TGF-β1/Smad pathways. SCIENCE ADVANCES 2023; 9:eadd4222. [PMID: 37531438 PMCID: PMC10396312 DOI: 10.1126/sciadv.add4222] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 06/29/2023] [Indexed: 08/04/2023]
Abstract
Cardiac fibrosis plays a key role in the progression of diabetic cardiomyopathy (DCM). Previous studies demonstrated the cardioprotective effects of natriuretic peptides. However, the effects of natriuretic peptide receptor C (NPRC) on cardiac fibrosis in DCM remains unknown. Here, we observed that myocardial NPRC expression was increased in mice and patients with DCM. NPRC-/- diabetic mice showed alleviated cardiac fibrosis, as well as improved cardiac function and remodeling. NPRC knockdown in both cardiac fibroblasts and cardiomyocytes decreased collagen synthesis and proliferation of cardiac fibroblasts. RNA sequencing identified that NPRC deletion up-regulated the expression of TGF-β-induced factor homeobox 1 (TGIF1), which inhibited the phosphorylation of Smad2/3. Furthermore, TGIF1 up-regulation was mediated by the activation of cAMP/PKA and cGMP/PKG signaling induced by NPRC deletion. These findings suggest that NPRC deletion attenuated cardiac fibrosis and improved cardiac remodeling and function in diabetic mice, providing a promising approach to the treatment of diabetic cardiac fibrosis.
Collapse
Affiliation(s)
- Linlin Meng
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yue Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinlu Wang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Cheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Fei Xue
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Lin Xie
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaoyuan Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wenhai Sui
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | | | - Yun Zhang
- Corresponding author. (Y.Z.); (C.Z.)
| | | |
Collapse
|
10
|
Zheng H, Patel TA, Liu X, Patel KP. C-type natriuretic peptide (CNP) in the paraventricular nucleus-mediated renal sympatho-inhibition. Front Physiol 2023; 14:1162699. [PMID: 37082246 PMCID: PMC10110992 DOI: 10.3389/fphys.2023.1162699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
Volume reflex produces sympatho-inhibition that is mediated by the hypothalamic paraventricular nucleus (PVN). However, the mechanisms for the sympatho-inhibitory role of the PVN and the neurochemical factors involved remain to be identified. In this study, we proposed C-type natriuretic peptide (CNP) as a potential mediator of this sympatho-inhibition within the PVN. Microinjection of CNP (1.0 μg) into the PVN significantly decreased renal sympathetic nerve activity (RSNA) (-25.8% ± 1.8% vs. -3.6% ± 1.5%), mean arterial pressure (-15.0 ± 1.9 vs. -0.1 ± 0.9 mmHg) and heart rate (-23.6 ± 3.5 vs. -0.3 ± 0.9 beats/min) compared with microinjection of vehicle. Picoinjection of CNP significantly decreased the basal discharge of extracellular single-unit recordings in 5/6 (83%) rostral ventrolateral medulla (RVLM)-projecting PVN neurons and in 6/13 (46%) of the neurons that were not antidromically activated from the RVLM. We also observed that natriuretic peptide receptor type C (NPR-C) was present on the RVLM projecting PVN neurons detected by dual-labeling with retrograde tracer. Prior NPR-C siRNA microinjection into the PVN significantly blunted the decrease in RSNA to CNP microinjections into the PVN. Volume expansion-mediated reduction in RSNA was significantly blunted by prior administration of NPR-C siRNA into the PVN. These results suggest a potential role for CNP within the PVN in regulating RSNA, specifically under physiological conditions of alterations in fluid balance.
Collapse
Affiliation(s)
- Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, United States
| | - Tapan A. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Xuefei Liu
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD, United States
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
11
|
Mavragani A, Pearson JF, Troughton RW, Kennedy MA, Espiner EA. The Predictive Value of A, B, and C-Type Natriuretic Peptides in People at Risk of Heart Disease: Protocol for a Longitudinal Observational Study. JMIR Res Protoc 2023; 12:e37011. [PMID: 36630163 PMCID: PMC9878369 DOI: 10.2196/37011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Heart disease and stroke are major and often unheralded causes of serious morbidity and premature death in middle age. Early detection of those most at risk is an urgent unmet need for instituting preventative measures. In an earlier community study (Canterbury Health, Ageing and Life Course [CHALICE]) of healthy people aged 50 years, contrary to previous reports, low levels of the heart hormone B-type natriuretic peptide (BNP) were associated with reduced measures of heart function and higher markers of vascular risk. A specific gene variant (rs198358) was found to be an independent contributor to higher BNP levels. A closely related vascular hormone (C-type natriuretic peptide [CNP]) showed opposite associations-higher levels were correlated with higher vascular risk and reduced cardiac function. To determine whether these novel findings predict serious heart or vascular disease in later life, this proposal re-examines the same CHALICE participants 15 years later. OBJECTIVE The primary objective is to determine the predictive value of (1) low plasma concentrations of the circulating cardiac hormones (atrial natriuretic peptide [ANP] and BNP) and (2) high levels of the vascular hormone CNP at age 50 years in detecting impaired cardiac and vascular function 15 years later. Secondary objectives are to determine specific associations of individual analytes (ANP, BNP, CNP, cyclic guanosine monophosphate [cGMP]) with echo-derived changes in cardiac performance at ages 50 years and 65 years. METHODS All of the 348 participants (205/348, 58.9% female; 53/348, 15.2% Māori or Pacifica ethnicity) participating in the original CHALICE study-free of history of heart or renal disease at age 50 years and who consented to further study-will be contacted, recruited, and restudied as previously described. Data will include intervening health history, physical examination, heart function (speckle-tracking echocardiography), vascular status (carotid intimal thickness), and genetic status (genome-wide genotyping). Laboratory measures will include fasting blood sampling and routine biochemistry, ANP, BNP, CNP, their downstream effector (cGMP), and their bio-inactive products. Humoral metabolic-cardiovascular risk factors will be measured after an overnight fast. Primary outcomes will be analyzed using multiple linear regression. RESULTS The study will commence in 2022 and be completed in 2024. CONCLUSIONS Proving our hypothesis-that low BNP and high CNP at any age in healthy people predict premature aging of heart and blood vessels, respectively-opens the way to early detection and improved outcomes for those most at risk. Confirmation of our hypotheses would improve current methods of screening and, in appropriate cases, enable interventions aimed at increasing natriuretic hormones and reducing risk of serious cardiovascular complications using drugs already available. Such advances in detection, and from interventional corrections, have the potential to not only improve health in the community but also reduce the high costs inevitably associated with heart failure. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/37011.
Collapse
Affiliation(s)
| | - John F Pearson
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.,Biostatistics and Computational Biology Unit, University of Otago, Christchurch, New Zealand
| | - Richard W Troughton
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christcurch, New Zealand
| | - Martin A Kennedy
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Eric A Espiner
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christcurch, New Zealand
| |
Collapse
|
12
|
Tokudome T, Otani K. Molecular Mechanism of Blood Pressure Regulation through the Atrial Natriuretic Peptide. BIOLOGY 2022; 11:biology11091351. [PMID: 36138830 PMCID: PMC9495342 DOI: 10.3390/biology11091351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Atrial natriuretic peptide (ANP) is a cardiac peptide hormone that was identified by Kangawa and Matsuo in 1984. In Japan, ANP has been used as an intravenous drug for the treatment of acute heart failure since 1995. Because ANP has a hypotensive effect, it is important to avoid excessive lowering of blood pressure when ANP is used. Recently, a compound that inhibits neutral endopeptidase, the enzyme that degrades ANP, has been developed (angiotensin receptor-neprilysin inhibitor (ARNI)). ARNI has been approved worldwide for the treatment of chronic heart failure and has been authorized in Japan as an antihypertensive drug. However, it is not understood exactly how ANP exerts its hypotensive effect. In this review, we discuss the molecular mechanism of the blood pressure-regulating effects of ANP, focusing on our recent findings. Abstract Natriuretic peptides, including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP), have cardioprotective effects and regulate blood pressure in mammals. ANP and BNP are hormones secreted from the heart into the bloodstream in response to increased preload and afterload. Both hormones act through natriuretic peptide receptor 1 (NPR1). In contrast, CNP acts through natriuretic peptide receptor 2 (NPR2) and was found to be produced by the vascular endothelium, chondrocytes, and cardiac fibroblasts. Based on its relatively low plasma concentration compared with ANP and BNP, CNP is thought to function as both an autocrine and a paracrine factor in the vasculature, bone, and heart. The cytoplasmic domains of both NPR1 and NPR2 display a guanylate cyclase activity that catalyzes the formation of cyclic GMP. NPR3 lacks this guanylate cyclase activity and is reportedly coupled to Gi-dependent signaling. Recently, we reported that the continuous infusion of the peptide osteocrin, an endogenous ligand of NPR3 secreted by bone and muscle cells, lowered blood pressure in wild-type mice, suggesting that endogenous natriuretic peptides play major roles in the regulation of blood pressure. Neprilysin is a neutral endopeptidase that degrades several vasoactive peptides, including natriuretic peptides. The increased worldwide clinical use of the angiotensin receptor-neprilysin inhibitor for the treatment of chronic heart failure has brought renewed attention to the physiological effects of natriuretic peptides. In this review, we provide an overview of the discovery of ANP and its translational research. We also highlight our recent findings on the blood pressure regulatory effects of ANP, focusing on its molecular mechanisms.
Collapse
Affiliation(s)
- Takeshi Tokudome
- Department of Pathophysiology of Heart Failure and Therapeutics, National Cerebral and Cardiovascular Center Research Institute, Suita 564-8565, Japan
- Correspondence: ; Tel.: +81-6-6170-1069
| | - Kentaro Otani
- Center for Regenerative Medicine, National Cerebral and Cardiovascular Center Research Institute, Suita 564-8565, Japan
| |
Collapse
|
13
|
CNP, the Third Natriuretic Peptide: Its Biology and Significance to the Cardiovascular System. BIOLOGY 2022; 11:biology11070986. [PMID: 36101368 PMCID: PMC9312265 DOI: 10.3390/biology11070986] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/19/2022]
Abstract
Simple Summary CNP is the third natriuretic peptide to be isolated and is widely expressed in the central nervous system, osteochondral system, and vascular system. The receptor that is mainly targeted by CNP is GC-B, which differs from GC-A, the receptor targeted by the other two natriuretic peptides, ANP and BNP. Consequently, the actions of CNP differ somewhat from those of ANP and BNP. Research into the actions of CNP has shown that CNP attenuates cardiac remodeling in animal models of cardiac hypertrophy, myocardial infarction, and myocarditis. Studies examining CNP/GC-B signaling showed that it contributes to the prevention of cardiac stiffness. Endogenous CNP, perhaps acting in part through CNP/NPR-C signaling, contributes to the regulation of vascular function and blood pressure. CNP regulates vascular remodeling and angiogenesis via CNP/GC-B/CGK signaling. CNP attenuates interstitial fibrosis and fibrosis-related gene expression in pressure overload and myocardial infarction models. The clinical application of CNP as a therapeutic agent for cardiovascular diseases is anticipated. Abstract The natriuretic peptide family consists of three biologically active peptides: ANP, BNP, and CNP. CNP is more widely expressed than the other two peptides, with significant levels in the central nervous system, osteochondral system, and vascular system. The receptor that is mainly targeted by CNP is GC-B, which differs from GC-A, the receptor targeted by ANP and BNP. Consequently, the actions of CNP differ somewhat from those of ANP and BNP. CNP knockout leads to severe dwarfism, and there has been important research into the role of CNP in the osteochondral system. As a result, a CNP analog is now available for clinical use in patients with achondroplasia. In the cardiovascular system, CNP and its downstream signaling are involved in the regulatory mechanisms underlying myocardial remodeling, cardiac function, vascular tone, angiogenesis, and fibrosis, among others. This review focuses on the roles of CNP in the cardiovascular system and considers its potential for clinical application in the treatment of cardiovascular diseases.
Collapse
|
14
|
Physiological and Pathophysiological Effects of C-Type Natriuretic Peptide on the Heart. BIOLOGY 2022; 11:biology11060911. [PMID: 35741432 PMCID: PMC9219612 DOI: 10.3390/biology11060911] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 01/06/2023]
Abstract
Simple Summary C-type natriuretic peptide (CNP) is the third member of the natriuretic peptide family. Unlike atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), CNP was not previously regarded as an important cardiac modulator. However, recent studies have revealed the physiological and pathophysiological importance of CNP in the heart; in concert with its cognate natriuretic peptide receptor-B (NPR-B), CNP has come to be regarded as the major heart-protective natriuretic peptide in the failed heart. In this review, I introduce the history of research on CNP in the cardiac field. Abstract C-type natriuretic peptide (CNP) is the third member of the natriuretic peptide family. Unlike other members, i.e., atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), which are cardiac hormones secreted from the atrium and ventricle of the heart, respectively, CNP is regarded as an autocrine/paracrine regulator with broad expression in the body. Because of its low expression levels compared to ANP and BNP, early studies failed to show its existence and role in the heart. However, recent studies have revealed the physiological and pathophysiological importance of CNP in the heart; in concert with the distribution of its specific natriuretic peptide receptor-B (NPR-B), CNP has come to be regarded as the major heart-protective natriuretic peptide in the failed heart. NPR-B generates intracellular cyclic guanosine 3′,5′-monophosphate (cGMP) upon CNP binding, followed by various molecular effects including the activation of cGMP-dependent protein kinases, which generates diverse cytoprotective actions in cardiomyocytes, as well as in cardiac fibroblasts. CNP exerts negative inotropic and positive lusitropic responses in both normal and failing heart models. Furthermore, osteocrin, the intrinsic and specific ligand for the clearance receptor for natriuretic peptides, can augment the effects of CNP and may supply a novel therapeutic strategy for cardiac protection.
Collapse
|
15
|
Hu P, Chen H, Wang LH, Jiang JB, Li JM, Tang MY, Guo YC, Zhu QF, Pu ZX, Lin XP, Ng S, Liu XB, Wang JA. Elevated N-terminal pro C-type natriuretic peptide is associated with mortality in patients undergoing transcatheter aortic valve replacement. BMC Cardiovasc Disord 2022; 22:164. [PMID: 35413789 PMCID: PMC9004019 DOI: 10.1186/s12872-022-02615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
Background Unlike N-terminal pro-B-type natriuretic peptide (NT-proBNP), which have been extensively studied, little is known about the role of N-terminal pro-C-type natriuretic peptide (NT-proCNP) for predicting survival post transcatheter aortic valve replacement (TAVR). Methods A total of 309 patients were included in the analysis. Patients were grouped into quartiles (Q1–4) according to the baseline NT-proCNP value. Blood for NT-proCNP analysis was obtained prior to TAVR procedure. The primary endpoint was mortality after a median follow-up of 32 months. Multivariable Cox proportional hazards regression models analyzed prognostic factors. The predictive capability was compared between NT-proBNP and NT-proCNP using receiver operator curve (ROC) analysis. Results A total of 309 subjects with the mean age of 76.8 ± 6.3 years, among whom 58.6% were male, were included in the analysis. A total of 58 (18.8%) patients died during follow-up. Cox multivariable analyses indicated society of thoracic surgeons (STS)-score was a strong independent predictor for mortality (hazard ratio (HR) 1.08, 95% confidential interval (CI) 1.05–1.12, P < 0.001). Elevated NT-proCNP was associated with a higher risk of cardiovascular mortality (HR 1.02, 95% CI 1.00–1.03, P = 0.025) and All-cause mortality (HR 1.01, 95% CI 1.00–1.03, P = 0.027), whereas NT-proBNP showed a small effect size on mortality. ROC analysis indicated that NT-proCNP was superior to NT-proBNP for TAVR risk evaluation in patients with left ventricular ejection fraction (LVEF) < 50% [(Area under the curve (AUC)-values of 0.79 (0.69; 0.87) vs. 0.59 (0.48; 0.69), P = 0.0453]. Conclusions NT-proCNP and STS-Score were the independent prognostic factors of mortality among TAVR patients. Furthermore, NT-proCNP was superior to NT-proBNP for TAVR risk evaluation in patients with LVEF < 50%. Trial registration NCT02803294, 16/06/2016. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-022-02615-8.
Collapse
Affiliation(s)
- Po Hu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Han Chen
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Li-Han Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Ju-Bo Jiang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Jia-Min Li
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China
| | - Meng-Yao Tang
- Renal Division, Brigham and Women's Hospital, Boston, MA, USA.,Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yu-Chao Guo
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Qi-Feng Zhu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Zhao-Xia Pu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Department of Echocardiography, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Xin-Ping Lin
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.,Department of Echocardiography, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Stella Ng
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China
| | - Xian-Bao Liu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China.
| | - Jian-An Wang
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China. .,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
16
|
Prognostic Value of Urinary and Plasma C-Type Natriuretic Peptide in Acute Decompensated Heart Failure. JACC-HEART FAILURE 2021; 9:613-623. [PMID: 34246604 DOI: 10.1016/j.jchf.2021.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVES This study sought to characterize urinary and plasma C-type natriuretic peptide (CNP) in acute decompensated heart failure (ADHF) to define their relationship with clinical variables and to determine whether urinary and plasma CNP together add prognostic value. BACKGROUND CNP is a protective hormone that is synthesized in the kidney and endothelium and possesses antiremodeling properties. Urinary and plasma CNP levels are elevated in pathophysiological conditions; however, their regulation and prognostic value in heart failure (HF) is unclear. METHODS Urinary and plasma CNP were measured in 109 healthy subjects and 208 patients with ADHF; the 95th percentile of CNP values from healthy subjects established the normal contemporary cutoffs. Patients with ADHF were stratified based on urinary and plasma CNP levels for clinical characterization and the assessment of risk for adverse outcomes. RESULTS There was no significant correlation between urinary and plasma CNP in both cohorts. Urinary and plasma CNP were significantly elevated in patients with ADHF, and both increased with disease severity and were positively correlated with plasma N-terminal pro-B-type natriuretic peptide (NT-proBNP). Of the patients with ADHF, 23% had elevations in both urinary and plasma CNP, whereas 24% had normal CNP levels. During a median follow-up of 3 years, patients with elevated urinary and plasma CNP had a significantly higher risk of rehospitalization and/or death (HR: 1.79; P = 0.03) and rehospitalization (HR: 2.16; P = 0.01) after adjusting for age, sex, left ventricular ejection fraction, renal function, and plasma NT-proBNP. The C-statistic and integrated discrimination analyses further supported that the addition of urinary and plasma CNP to established risk models improved the prediction of adverse outcomes in patients with ADHF. CONCLUSIONS Urinary and plasma CNP are differentially regulated in ADHF, and elevations in both provided independent prognostic value for predicting adverse outcomes.
Collapse
|
17
|
Cardiac morphological and functional changes induced by C-type natriuretic peptide are different in normotensive and spontaneously hypertensive rats. J Hypertens 2021; 38:2305-2317. [PMID: 32649642 DOI: 10.1097/hjh.0000000000002570] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Inflammation and fibrosis are key mechanisms in cardiovascular remodeling. C-type natriuretic peptide (CNP) is an endothelium-derived factor with a cardiovascular protective role, although its in-vivo effect on cardiac remodeling linked to hypertension has not been investigated. The aim of this study was to determine the effects of chronic administration of CNP on inflammatory and fibrotic cardiac mechanisms in normotensive Wistar rats and spontaneously hypertensive rats (SHR). METHODS Twelve-week-old male SHR and normotensive rats were infused with CNP (0.75 μg/h/100 g) or isotonic saline (NaCl 0.9%) for 14 days (subcutaneous micro-osmotic pumps). Echocardiograms and electrocardiograms were performed, and SBP was measured. After treatment, transforming growth factor-beta 1, Smad proteins, tumor necrosis factor-alpha, interleukin-1 and interleukin-6, nitric oxide (NO) system and 2-thiobarbituric acid-reactive substances were evaluated in left ventricle. Histological studies were also performed. RESULTS SHR showed lower cardiac output with signs of fibrosis and hypertrophy in left ventricle, higher NO-system activity and more oxidative damage, as well as higher pro-inflammatory and pro-fibrotic markers than normotensive rats. Chronic CNP treatment-attenuated hypertension and ventricular hypertrophy in SHR, with no changes in normotensive rats. In left ventricle, CNP induced an anti-inflammatory and antifibrotic response, decreasing both pro-fibrotic and pro-inflammatory cytokines in SHR. In addition, CNP reduced oxidative damage as well as collagen content, and upregulated the NO system in both groups. CONCLUSION Chronic CNP treatment appears to attenuate hypertension and associated end-organ damage in the heart by reducing inflammation and fibrosis.
Collapse
|
18
|
Egom EEA. Natriuretic Peptide Clearance Receptor (NPR-C) Pathway as a Novel Therapeutic Target in Obesity-Related Heart Failure With Preserved Ejection Fraction (HFpEF). Front Physiol 2021; 12:674254. [PMID: 34093235 PMCID: PMC8176210 DOI: 10.3389/fphys.2021.674254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/12/2021] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a major public health problem with cases projected to double over the next two decades. There are currently no US Food and Drug Administration–approved therapies for the health-related outcomes of HFpEF. However, considering the high prevalence of this heterogeneous syndrome, a directed therapy for HFpEF is one the greatest unmet needs in cardiovascular medicine. Additionally, there is currently a lack of mechanistic understanding about the pathobiology of HFpEF. The phenotyping of HFpEF patients into pathobiological homogenous groups may not only be the first step in understanding the molecular mechanism but may also enable the development of novel targeted therapies. As obesity is one of the most common comorbidities found in HFpEF patients and is associated with many cardiovascular effects, it is a viable candidate for phenotyping. Large outcome trials and registries reveal that being obese is one of the strongest independent risk factors for developing HFpEF and that this excess risk may not be explained by traditional cardiovascular risk factors. Recently, there has been increased interest in the intertissue communication between adipose tissue and the heart. Evidence suggests that the natriuretic peptide clearance receptor (NPR-C) pathway may play a role in the development and pathobiology of obesity-related HFpEF. Therefore, therapeutic manipulations of the NPR-C pathway may represent a new pharmacological strategy in the context of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Emmanuel Eroume A Egom
- Institut du Savoir Montfort, Hôpital Montfort, University of Ottawa, Ottawa, ON, Canada.,Laboratory of Endocrinology and Radioisotopes, Institute of Medical Research and Medicinal Plants Studies, Yaoundé, Cameroon
| |
Collapse
|
19
|
Murphy SP, Prescott MF, Maisel AS, Butler J, Piña IL, Felker GM, Ward JH, Williamson KM, Camacho A, Kandanelly RR, Solomon SD, Januzzi JL. Association Between Angiotensin Receptor-Neprilysin Inhibition, Cardiovascular Biomarkers, and Cardiac Remodeling in Heart Failure With Reduced Ejection Fraction. Circ Heart Fail 2021; 14:e008410. [PMID: 33998243 DOI: 10.1161/circheartfailure.120.008410] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Sacubitril/valsartan (S/V) treatment is associated with reverse cardiac remodeling and reductions in biomarkers reflecting ventricular wall stress and myocardial injury, such as NT-proBNP (N-terminal pro-B-type natriuretic peptide), hs-cTnT (high-sensitivity cardiac troponin T), and soluble suppressor of tumorigenicity 2 (sST2). How longitudinal changes in these biomarkers analyzed collectively are associated with cardiac remodeling in patients with heart failure with reduced ejection fraction treated with S/V is uncertain. METHODS In a prospective study of S/V in patients with heart failure with reduced ejection fraction, this prespecified exploratory analysis included patients with serially collected biomarkers and echocardiographic measures of cardiac remodeling through 12 months of treatment. A multivariate latent growth curve model assessed associations between simultaneous changes in biomarkers and left ventricular ejection fraction and left atrial volume index. RESULTS Seven hundred fifteen out of 794 total study participants were included (mean age 65 years, 73% male). Mean baseline left ventricular ejection fraction and left atrial volume index were 29% and 40 mL/m2, respectively. Adjusted geometric mean baseline concentrations for biomarkers included NT-proBNP of 649 pg/mL, hs-cTnT of 15.9 ng/L, and sST2 of 24.7 ng/mL. Following initiation of S/V, circulating concentrations of NT-proBNP, hs-cTnT, and sST2 significantly decreased within 30 days and remained significantly different than baseline at all subsequent timepoints. From baseline to month 12, decreases in adjusted biomarker concentrations averaged -27.9% (95% CI, -35.1% to -20.7%; P<0.001) for NT-proBNP; -6.7% (95% CI, -8.8% to -4.7%; P<0.001) for hs-cTnT; and -1.6% (95% CI, -2.9% to -0.4%; P<0.001) for sST2. NT-proBNP concentrations were predictive of later changes in hs-cTnT. The magnitude of reductions in NT-proBNP and hs-cTnT concentrations associated with improvements in left ventricular ejection fraction and left atrial volume index. There was no association between changes in sST2 and changes in other measures. CONCLUSIONS Following initiation of S/V, NT-proBNP, hs-cTnT, and sST2 concentrations decreased significantly. Longitudinal changes in NT-proBNP and hs-cTnT together associated with left atrial and left ventricular reverse remodeling. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02887183.
Collapse
Affiliation(s)
- Sean P Murphy
- Massachusetts General Hospital, Boston (S.P.M., A.C., R.R.K., J.L.J.)
| | | | - Alan S Maisel
- University of California, San Diego School of Medicine (A.S.M.)
| | - Javed Butler
- University of Mississippi Medical Center, Jackson (J.B.)
| | | | - G Michael Felker
- Duke University Medical Center and Duke Clinical Research Institute, Durham, NC (G.M.F.)
| | - Jonathan H Ward
- Novartis Pharmaceuticals, East Hanover, NJ (M.F.P., J.H.W., K.M.W.)
| | | | - Alexander Camacho
- Massachusetts General Hospital, Boston (S.P.M., A.C., R.R.K., J.L.J.)
| | | | - Scott D Solomon
- Brigham and Women's Hospital, Boston, MA (S.D.S.).,Harvard Medical School, Boston, MA (S.D.S., J.L.J.)
| | - James L Januzzi
- Massachusetts General Hospital, Boston (S.P.M., A.C., R.R.K., J.L.J.).,Harvard Medical School, Boston, MA (S.D.S., J.L.J.).,Baim Institute for Clinical Research, Boston, MA (J.L.J.)
| |
Collapse
|
20
|
Abstract
The myocardium consists of different cell types, of which endothelial cells, cardiomyocytes, and fibroblasts are the most abundant. Communication between these different cell types, also called paracrine signaling, is essential for normal cardiac function, but also important in cardiac remodeling and heart failure. Systematic studies on the expression of ligands and their corresponding receptors in different cell types showed that for 60% of the expressed ligands in a particular cell, the receptor is also expressed. The fact that many ligand-receptor pairs are present in most cells, including the major cell types in the heart, indicates that autocrine signaling is a widespread phenomenon. Autocrine signaling in cardiac remodeling and heart failure is involved in all pathophysiological mechanisms generally observed: hypertrophy, fibrosis, angiogenesis, cell survival, and inflammation. Herein, we review ligand-receptor pairs present in the major cardiac cell types based on RNA-sequencing expression databases, and we review current literature on extracellular signaling proteins with an autocrine function in the heart; these include C-type natriuretic peptide, fibroblast growth factors 2, F21, and 23, macrophage migration inhibitory factor, heparin binding-epidermal growth factor, angiopoietin-like protein 2, leptin, adiponectin, follistatin-like 1, apelin, neuregulin 1, vascular endothelial growth factor, transforming growth factor β, wingless-type integration site family, member 1-induced secreted protein-1, interleukin 11, connective tissue growth factor/cellular communication network factor, and calcitonin gene‒related peptide. The large number of autocrine signaling factors that have been studied in the literature supports the concept that autocrine signaling is an essential part of myocardial biology and disease.
Collapse
Affiliation(s)
- Vincent F. M. Segers
- Laboratory of PhysiopharmacologyUniversity of AntwerpBelgium
- Department of CardiologyUniversity Hospital AntwerpEdegemBelgium
| | - Gilles W. De Keulenaer
- Laboratory of PhysiopharmacologyUniversity of AntwerpBelgium
- Department of CardiologyZNA HospitalAntwerpBelgium
| |
Collapse
|
21
|
Vitiello A, La Porta R, Ferrara F. Scientific hypothesis and rational pharmacological for the use of sacubitril/valsartan in cardiac damage caused by COVID-19. Med Hypotheses 2021; 147:110486. [PMID: 33460992 PMCID: PMC7788318 DOI: 10.1016/j.mehy.2021.110486] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/30/2020] [Accepted: 01/02/2021] [Indexed: 12/17/2022]
Abstract
On March 11, 2020 the World Health Organization (WHO) declared the state of global pandemic caused by the new SARS-CoV-2 (COVID-19). To date, no antivirals directed against SARS-CoV-2 or effective vaccines to combat the viral infection are available. Severe acute respiratory syndrome caused by SARS-CoV-2 is treated empirically with antivirals, anti-inflammatory, anticoagulants. The approval of an effective vaccine still takes time. In this state, it may be useful to find new therapeutic solutions from drugs already on the market. Recent hypotheses suggest that the use of AT-1 receptor antagonists (ARB) in combination with neprilisin inhibitors (NEPi) could indirectly provide clinical benefits to patients with SARS-CoV-2 and cardiac involvement. In this article we investigate and describe a possible innovative pharmacological approach for the treatment of the most severe stages of COVID-19 infection.
Collapse
Affiliation(s)
- Antonio Vitiello
- Pharmaceutical Department, Usl Umbria 1, XIV Settembre Street, 06132 Perugia, Italy.
| | - Raffaele La Porta
- Clinical Pathology, Asur Marche, Viale Guido Da Montefeltro, Urbino, Italy.
| | - Francesco Ferrara
- Pharmaceutical Department, Usl Umbria 1, A. Migliorati Street, 06132 Perugia, Italy.
| |
Collapse
|
22
|
Michel K, Herwig M, Werner F, Špiranec Spes K, Abeßer M, Schuh K, Dabral S, Mügge A, Baba HA, Skryabin BV, Hamdani N, Kuhn M. C-type natriuretic peptide moderates titin-based cardiomyocyte stiffness. JCI Insight 2020; 5:139910. [PMID: 33055420 PMCID: PMC7710274 DOI: 10.1172/jci.insight.139910] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Heart failure is often accompanied by titin-dependent myocardial stiffness. Phosphorylation of titin by cGMP-dependent protein kinase I (PKGI) increases cardiomyocyte distensibility. The upstream pathways stimulating PKGI-mediated titin phosphorylation are unclear. We studied whether C-type natriuretic peptide (CNP), via its guanylyl cyclase-B (GC-B) receptor and cGMP/PKGI signaling, modulates titin-based ventricular compliance. To dissect GC-B–mediated effects of endogenous CNP in cardiomyocytes, we generated mice with cardiomyocyte-restricted GC-B deletion (CM GC-B–KO mice). The impact on heart morphology and function, myocyte passive tension, and titin isoform expression and phosphorylation was studied at baseline and after increased afterload induced by transverse aortic constriction (TAC). Pressure overload increased left ventricular endothelial CNP expression, with an early peak after 3 days. Concomitantly, titin phosphorylation at Ser4080, the site phosphorylated by PKGI, was augmented. Notably, in CM GC-B–KO mice this titin response was abolished. TAC-induced hypertrophy and fibrosis were not different between genotypes. However, the KO mice presented mild systolic and diastolic dysfunction together with myocyte stiffness, which were not observed in control littermates. In vitro, recombinant PKGI rescued reduced titin-Ser4080 phosphorylation and reverted passive stiffness of GC-B–deficient cardiomyocytes. CNP-induced activation of GC-B/cGMP/PKGI signaling in cardiomyocytes provides a protecting regulatory circuit preventing titin-based myocyte stiffening during early phases of pressure overload. C-type natriuretic peptide via GC-B/cGMP/PKGI signalling in cardiomyocytes attenuates titin-based cardiomyocyte stiffening during early phases of pressure-overload.
Collapse
Affiliation(s)
- Konstanze Michel
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Melissa Herwig
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Franziska Werner
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Marco Abeßer
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Kai Schuh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Swati Dabral
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Andreas Mügge
- Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Hideo A Baba
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Boris V Skryabin
- Medical Faculty, Core Facility TRAnsgenic animal and genetic engineering Models (TRAM), University of Münster, Münster, Germany
| | - Nazha Hamdani
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
23
|
Murphy SP, Prescott MF, Camacho A, Iyer SR, Maisel AS, Felker GM, Butler J, Piña IL, Ibrahim NE, Abbas C, Burnett JC, Solomon SD, Januzzi JL. Atrial Natriuretic Peptide and Treatment With Sacubitril/Valsartan in Heart Failure With Reduced Ejection Fraction. JACC-HEART FAILURE 2020; 9:127-136. [PMID: 33189632 DOI: 10.1016/j.jchf.2020.09.013] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVES This study sought to assess associations between longitudinal change in atrial natriuretic peptide (ANP) and reverse cardiac remodeling following initiation of sacubitril/valsartan in patients with heart failure with reduced ejection fraction (HFrEF). BACKGROUND Neprilysin inhibition results in an increase of several vasoactive peptides that may mediate the beneficial effects of sacubitril/valsartan, including ANP. METHODS In a prospective study of initiation and titration of sacubitril/valsartan in patients with HFrEF, blood was collected at scheduled time points into tubes containing protease inhibitors. This pre-specified exploratory analysis included patients in whom ANP was measured at baseline and serially through 12 months of treatment. RESULTS Among 144 participants (mean age: 64.5 years; left ventricular ejection fraction: 30.8%), following initiation of sacubitril/valsartan, there was an early and significant increase in ANP, with the majority of rise from 99 pg/ml at baseline to 156 pg/ml at day 14 (p < 0.001). There was a further trend toward a second increase from day 30 to day 45 (p = 0.07). At maximal rise, ANP had doubled. In longitudinal analyses, early rise in ANP was followed by a subsequent increase in urinary cycle guanosine monophosphate. Larger early increase in ANP was associated with larger later improvements in left ventricular ejection fraction and left atrial volume index (p < 0.001 for both). CONCLUSIONS Concentrations of ANP doubled after initiation of sacubitril/valsartan in patients with HFrEF. Larger early increases in ANP were associated with a greater magnitude of subsequent reverse cardiac remodeling. (Effects of Sacubitril/Valsartan Therapy on Biomarkers, Myocardial Remodeling and Outcomes [PROVE-HF]; NCT02887183).
Collapse
Affiliation(s)
- Sean P Murphy
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | | - Alan S Maisel
- University of California, San Diego School of Medicine, San Diego, California, USA
| | - G Michael Felker
- Duke University Medical Center and Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Javed Butler
- University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | - Nasrien E Ibrahim
- Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Cheryl Abbas
- Novartis Pharmaceuticals, East Hanover, New Jersey, USA
| | | | - Scott D Solomon
- Harvard Medical School, Boston, Massachusetts, USA; Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - James L Januzzi
- Massachusetts General Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA; Baim Institute for Clinical Research, Boston, Massachusetts, USA.
| |
Collapse
|
24
|
Li LYF, Lou Q, Liu GZ, Lv JC, Yun FX, Li TK, Yang W, Zhao HY, Zhang L, Bai N, Zhan CC, Yu J, Zang YX, Li WM. Sacubitril/valsartan attenuates atrial electrical and structural remodelling in a rabbit model of atrial fibrillation. Eur J Pharmacol 2020; 881:173120. [PMID: 32325147 DOI: 10.1016/j.ejphar.2020.173120] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 01/09/2023]
Abstract
Atrial structural and electrical remodelling play important roles in atrial fibrillation (AF). Sacubitril/valsartan attenuates cardiac remodelling in heart failure. However, the effect of sacubitril/valsartan on AF is unclear. The aim of this study was to evaluate the effect of sacubitril/valsartan on atrial electrical and structural remodelling in AF and investigate the underlying mechanism of action. Thirty-three rabbits were randomized into sham, RAP, and sac/val groups. HL-1 cells were subjected to control treatment or rapid pacing with or without LBQ657 and valsartan. Echocardiography, atrial electrophysiology, and histological examination were performed. The concentration of Ca2+ and expression levels of calcineurin, NFAT, p-NFAT, Cav1.2, collagen Ⅰ and Ⅲ, ANP, BNP, CNP, NT-proBNP, and ST2 in HL-1 cells, and IcaL in left atrial cells, were determined. We observed that compared to that in the sham group, the atrium and right ventricle were enlarged, myocardial fibrosis was markedly higher, AF inducibility was significantly elevated, and atrial effective refractory periods were shortened in the RAP group. These effects were significantly reversed by sacubitril/valsartan. Compared to that in the sham group, collagen Ⅰ and Ⅲ, NT-proBNP, ST2, calcineurin, and NFAT were significantly up-regulated, while p-NFAT and Cav1.2 were down-regulated in the RAP group, and sacubitril/valsartan inhibited these changes. Ca2+ concentration increased and ICaL density decreased in in vivo and in vitro AF models, reversed by sacubitril/valsartan. Sacubitril/valsartan attenuates atrial electrical remodelling and ameliorates structure remodelling in AF. This study paves the way for the possibility of clinical use of sacubitril/valsartan in AF patients.
Collapse
Affiliation(s)
- Lu-Yi-Fei Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Qi Lou
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Guang-Zhong Liu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jia-Chen Lv
- Department of Colorectal Surgery, The Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Feng-Xiang Yun
- Department of Internal Critic Care, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tian-Kai Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wen Yang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Hong-Yan Zhao
- Department of Cardiology, The People's Hospital of Liaoning Province, Shenyang, China
| | - Li Zhang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Nan Bai
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Cheng-Chuang Zhan
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Jia Yu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Yan-Xiang Zang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Wei-Min Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China.
| |
Collapse
|
25
|
Moyes AJ, Chu SM, Aubdool AA, Dukinfield MS, Margulies KB, Bedi KC, Hodivala-Dilke K, Baliga RS, Hobbs AJ. C-type natriuretic peptide co-ordinates cardiac structure and function. Eur Heart J 2020; 41:1006-1020. [PMID: 30903134 PMCID: PMC7068173 DOI: 10.1093/eurheartj/ehz093] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/21/2018] [Accepted: 02/26/2019] [Indexed: 12/11/2022] Open
Abstract
AIMS C-type natriuretic peptide (CNP) is an essential endothelium-derived signalling species that governs vascular homoeostasis; CNP is also expressed in the heart but an intrinsic role for the peptide in cardiac function is not established. Herein, we employ unique transgenic strains with cell-specific deletion of CNP to define a central (patho)physiological capacity of CNP in maintaining heart morphology and contractility. METHODS AND RESULTS Cardiac structure and function were explored in wild type (WT), cardiomyocyte (cmCNP-/-), endothelium (ecCNP-/-), and fibroblast (fbCNP-/-)-specific CNP knockout mice, and global natriuretic peptide receptor (NPR)-B-/-, and NPR-C-/- animals at baseline and in experimental models of myocardial infarction and heart failure (HF). Endothelium-specific deletion of CNP resulted in impaired coronary responsiveness to endothelium-dependent- and flow-mediated-dilatation; changes mirrored in NPR-C-/- mice. Ex vivo, global ischaemia resulted in larger infarcts and diminished functional recovery in cmCNP-/- and NPR-C-/-, but not ecCNP-/-, vs. WT. The cardiac phenotype of cmCNP-/-, fbCNP-/-, and NPR-C-/- (but not ecCNP-/- or NPR-B-/-) mice was more severe in pressure overload- and sympathetic hyperactivation-induced HF compared with WT; these adverse effects were rescued by pharmacological CNP administration in WT, but not NPR-C-/-, mice. At a molecular level, CNP/NPR-C signalling is impaired in human HF but attenuates activation of well-validated pro-hypertrophic and pro-fibrotic pathways. CONCLUSION C-type natriuretic peptide of cardiomyocyte, endothelial and fibroblast origins co-ordinates and preserves cardiac structure, function, and coronary vasoreactivity via activation of NPR-C. Targeting NPR-C may prove an innovative approach to treating HF and ischaemic cardiovascular disorders.
Collapse
Affiliation(s)
- Amie J Moyes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sandy M Chu
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Aisah A Aubdool
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Matthew S Dukinfield
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Kenneth B Margulies
- Heart Failure and Transplant Program, Perelman School of Medicine, University of Pennsylvania, Translational Research Center, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Kenneth C Bedi
- Heart Failure and Transplant Program, Perelman School of Medicine, University of Pennsylvania, Translational Research Center, 3400 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Reshma S Baliga
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| |
Collapse
|
26
|
Ding DZ, Jia YN, Zhang B, Guan CM, Zhou S, Li X, Cui X. C‑type natriuretic peptide prevents angiotensin II‑induced atrial connexin 40 and 43 dysregulation by activating AMP‑activated kinase signaling. Mol Med Rep 2019; 20:5091-5099. [PMID: 31638216 PMCID: PMC6854524 DOI: 10.3892/mmr.2019.10744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 08/14/2019] [Indexed: 12/13/2022] Open
Abstract
C‑type natriuretic peptide (CNP), from the family of natriuretic peptides (NPs), has been shown to induce antihypertrophic and antifibrotic effects in cardiomyocytes. However, the roles of CNP in the atrial dysregulation of connexin (Cx)40 and Cx43 remain to be elucidated. The present study aimed to investigate the effects of CNP on angiotensin (Ang) II‑induced Cx40 and Cx43 dysregulation in isolated perfused beating rat left atria. A rat isolated perfused beating atrial model was used and the protein levels were determined via western blotting. Ang II significantly upregulated NF‑κB, activator protein‑1, transforming growth factor‑β1 (TGF‑β1), collagen I and matrix metalloproteinase 2, leading to atrial fibrosis, and downregulated expression of Cx40 and Cx43. The changes in Cx40 and Cx43 induced by Ang II were abolished by CNP through upregulation of phosphorylated AMP‑activated kinase a1 (AMPK) and downregulation of TGF‑β1. The effects of CNP on AMPK and TGF‑β1 levels were inhibited by KT5823 and pertussis toxin, inhibitors of protein kinase G (PKG) and NP receptor type C (NPR‑C), respectively. Thus, CNP can prevent Ang II‑induced dysregulation of Cx40 and Cx43 through activation of AMPK via the CNP‑PKG and CNP‑NPR‑C pathways in isolated beating rat atria. The present findings suggested that CNP may be therapeutically useful for clinical conditions involving cardiac dysregulation of Cx expression‑related diseases.
Collapse
Affiliation(s)
- Da-Zhi Ding
- Department of Cardiology, Institute of Clinical Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Ya-Nan Jia
- Department of Cardiology, Institute of Clinical Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Bo Zhang
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Cheng-Ming Guan
- Department of Cardiology, Institute of Clinical Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Shuai Zhou
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Xiang Li
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
| | - Xun Cui
- Department of Physiology, College of Medicine, Yanbian University, Yanji, Jilin 133002, P.R. China
- Key Laboratory of Organism Functional Factors of The Changbai Mountain, Ministry of Education, Yanbian University, Yanji, Jilin 133002, P.R. China
- Cellular Function Research Center, Yanbian University, Yanji, Jilin 133002, P.R. China
| |
Collapse
|
27
|
Rubattu S, Volpe M. Natriuretic Peptides in the Cardiovascular System: Multifaceted Roles in Physiology, Pathology and Therapeutics. Int J Mol Sci 2019; 20:ijms20163991. [PMID: 31426320 PMCID: PMC6721730 DOI: 10.3390/ijms20163991] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
The natriuretic peptides (NPs) family includes a class of hormones and their receptors needed for the physiological control of cardiovascular functions. The discovery of NPs provided a fundamental contribution into our understanding of the physiological regulation of blood pressure, and of heart and kidney functions. NPs have also been implicated in the pathogenesis of several cardiovascular diseases (CVDs), including hypertension, atherosclerosis, heart failure, and stroke. A fine comprehension of the molecular mechanisms dependent from NPs and underlying the promotion of cardiovascular damage has contributed to improve our understanding of the molecular basis of all major CVDs. Finally, the opportunity to target NPs in order to develop new therapeutic tools for a better treatment of CVDs has been developed over the years. The current Special Issue of the Journal covers all major aspects of the molecular implications of NPs in physiology and pathology of the cardiovascular system, including NP-based therapeutic approaches.
Collapse
Affiliation(s)
- Speranza Rubattu
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy.
- IRCCS Neuromed, 86077 Pozzilli (Isernia), Italy.
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy.
- IRCCS Neuromed, 86077 Pozzilli (Isernia), Italy.
| |
Collapse
|
28
|
Forte M, Madonna M, Schiavon S, Valenti V, Versaci F, Zoccai GB, Frati G, Sciarretta S. Cardiovascular Pleiotropic Effects of Natriuretic Peptides. Int J Mol Sci 2019; 20:3874. [PMID: 31398927 PMCID: PMC6719167 DOI: 10.3390/ijms20163874] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023] Open
Abstract
Atrial natriuretic peptide (ANP) is a cardiac hormone belonging to the family of natriuretic peptides (NPs). ANP exerts diuretic, natriuretic, and vasodilatory effects that contribute to maintain water-salt balance and regulate blood pressure. Besides these systemic properties, ANP displays important pleiotropic effects in the heart and in the vascular system that are independent of blood pressure regulation. These functions occur through autocrine and paracrine mechanisms. Previous works examining the cardiac phenotype of loss-of-function mouse models of ANP signaling showed that both mice with gene deletion of ANP or its receptor natriuretic peptide receptor A (NPR-A) developed cardiac hypertrophy and dysfunction in response to pressure overload and chronic ischemic remodeling. Conversely, ANP administration has been shown to improve cardiac function in response to remodeling and reduces ischemia-reperfusion (I/R) injury. ANP also acts as a pro-angiogenetic, anti-inflammatory, and anti-atherosclerotic factor in the vascular system. Pleiotropic effects regarding brain natriuretic peptide (BNP) and C-type natriuretic peptide (CNP) were also reported. In this review, we discuss the current evidence underlying the pleiotropic effects of NPs, underlying their importance in cardiovascular homeostasis.
Collapse
Affiliation(s)
| | | | - Sonia Schiavon
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Valentina Valenti
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy
| | - Francesco Versaci
- Department of Cardiology, Santa Maria Goretti Hospital, 04100 Latina, Italy
| | - Giuseppe Biondi Zoccai
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
- Mediterranea Cardiocentro, 80122 Napoli, Italy
| | - Giacomo Frati
- IRCCS NEUROMED, 86077 Pozzilli, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Sebastiano Sciarretta
- IRCCS NEUROMED, 86077 Pozzilli, Italy.
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy.
| |
Collapse
|
29
|
Moyes AJ, Hobbs AJ. C-type Natriuretic Peptide: A Multifaceted Paracrine Regulator in the Heart and Vasculature. Int J Mol Sci 2019; 20:E2281. [PMID: 31072047 PMCID: PMC6539462 DOI: 10.3390/ijms20092281] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/11/2022] Open
Abstract
C-type natriuretic peptide (CNP) is an autocrine and paracrine mediator released by endothelial cells, cardiomyocytes and fibroblasts that regulates vital physiological functions in the cardiovascular system. These roles are conveyed via two cognate receptors, natriuretic peptide receptor B (NPR-B) and natriuretic peptide receptor C (NPR-C), which activate different signalling pathways that mediate complementary yet distinct cellular responses. Traditionally, CNP has been deemed the endothelial component of the natriuretic peptide system, while its sibling peptides, atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), are considered the endocrine guardians of cardiac function and blood volume. However, accumulating evidence indicates that CNP not only modulates vascular tone and blood pressure, but also governs a wide range of cardiovascular effects including the control of inflammation, angiogenesis, smooth muscle and endothelial cell proliferation, atherosclerosis, cardiomyocyte contractility, hypertrophy, fibrosis, and cardiac electrophysiology. This review will focus on the novel physiological functions ascribed to CNP, the receptors/signalling mechanisms involved in mediating its cardioprotective effects, and the development of therapeutics targeting CNP signalling pathways in different disease pathologies.
Collapse
Affiliation(s)
- Amie J Moyes
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Adrian J Hobbs
- William Harvey Research Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
30
|
Chen Y, Zheng Y, Iyer SR, Harders GE, Pan S, Chen HH, Ichiki T, Burnett JC, Sangaralingham SJ. C53: A novel particulate guanylyl cyclase B receptor activator that has sustained activity in vivo with anti-fibrotic actions in human cardiac and renal fibroblasts. J Mol Cell Cardiol 2019; 130:140-150. [PMID: 30954448 DOI: 10.1016/j.yjmcc.2019.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/08/2019] [Accepted: 03/31/2019] [Indexed: 01/01/2023]
Abstract
The native particulate guanylyl cyclase B receptor (pGC-B) activator, C-type natriuretic peptide (CNP), induces anti-remodeling actions in the heart and kidney through the generation of the second messenger 3', 5' cyclic guanosine monophosphate (cGMP). Indeed fibrotic remodeling, particularly in cardiorenal disease states, contributes to disease progression and thus, has been a key target for drug discovery and development. Although the pGC-B/cGMP system has been perceived as a promising anti-fibrotic pathway, its therapeutic potential is limited due to the rapid degradation and catabolism of CNP by neprilysin (NEP) and natriuretic peptide clearance receptor (NPRC). The goal of this study was to bioengineer and test in vitro and in vivo a novel pGC-B activator, C53. Here we established that C53 selectively generates cGMP via the pGC-B receptor and is highly resistant to NEP and has less interaction with NPRC in vitro. Furthermore in vivo, C53 had enhanced cGMP-generating actions that paralleled elevated plasma CNP-like levels, thus indicating a longer circulating half-life compared to CNP. Importantly in human cardiac fibroblasts (HCFs) and renal fibroblasts (HRFs), C53 exerted robust cGMP-generating actions, inhibited TGFβ-1 stimulated HCFs and HRFs proliferation chronically and suppressed the differentiation of HCFs and HRFs to myofibroblasts. The current findings advance innovation in drug discovery and highlight C53 as a novel pGC-B activator with sustained in vivo activity and anti-fibrotic actions in vitro. Future studies are warranted to explore the efficacy and therapeutic opportunity of C53 targeting fibrosis in cardiorenal disease states and beyond.
Collapse
Affiliation(s)
- Yang Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States; Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States.
| | - Ye Zheng
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Seethalakshmi R Iyer
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Gerald E Harders
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Shuchong Pan
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Horng H Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States; Department of Physiology and Biomedical Engineering, United States
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, United States; Department of Physiology and Biomedical Engineering, United States.
| |
Collapse
|
31
|
Wu K, Mei C, Chen Y, Guo L, Yu Y, Huang D. C-type natriuretic peptide regulates sperm capacitation by the cGMP/PKG signalling pathway via Ca 2+ influx and tyrosine phosphorylation. Reprod Biomed Online 2019; 38:289-299. [PMID: 30655075 DOI: 10.1016/j.rbmo.2018.11.025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 11/21/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022]
Abstract
RESEARCH QUESTION What is the effect of C-type natriuretic peptide (CNP) on human sperm capacitation in vitro and what is the mechanism of this effect? DESIGN CNP/NPR-B expression in the female rat genital tract was examined by immunohistochemistry and western blot assay, and then the role of CNP in human sperm capacitation was determined. The signal transduction pathway of CNP in the process was determined to elucidate the regulation mechanism of CNP by enzyme-linked immunosorbent assay and flow cytometry. RESULTS Both CNP and NPR-B were expressed in the genital tract of female rats, especially in the mucosa epithelium cell of the oviduct; the CNP level in the rat oviduct was higher than that in the cervix. Both CNP and NPR-B level in the rat oviduct varied during the oestrus cycle, maximal expression being observed at proestrus. Furthermore, intracellular cGMP level in spermatozoa was significantly enhanced by CNP (P < 0.01). PKG activity was detected in the spermatozoa, and it can be activated by the CNP and 8-Br-cGMP (cGMP analogue). The PKG inhibitor KT5823 inhibited the effect of CNP on sperm hyperactivation and the acrosome reaction. Finally, Ca2+ and tyrosine phosphorylation levels in spermatozoa were markedly improved by CNP and 8-Br-cGMP but significantly inhibited by the addition of KT5823 (P < 0.05). CONCLUSIONS CNP secreted by the female genital tract might bind to NPR-B on the spermatozoa. It successively stimulated intracellular cGMP/PKG signalling, increased Ca2+ and tyrosine-phosphorylated proteins, promoted hyperactivation and induced the acrosome reaction, which ultimately facilitated sperm capacitation.
Collapse
Affiliation(s)
- Kejia Wu
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Zhongnan Hospital, Wuhan University, Wuhan 430071, China
| | - Chunlei Mei
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yao Chen
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Reproductive Medicine Centre of Jingzhou Central Hospital, Jingzhou 434000, China
| | - Lidan Guo
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuejin Yu
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Donghui Huang
- Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
32
|
Nakagawa Y, Nishikimi T, Kuwahara K. Atrial and brain natriuretic peptides: Hormones secreted from the heart. Peptides 2019; 111:18-25. [PMID: 29859763 DOI: 10.1016/j.peptides.2018.05.012] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/23/2018] [Accepted: 05/27/2018] [Indexed: 02/01/2023]
Abstract
The natriuretic peptide family consists of three biologically active peptides: atrial natriuretic peptide (ANP), brain (or B-type) natriuretic peptide (BNP), and C-type natriuretic peptide (CNP). Among these, ANP and BNP are secreted by the heart and act as cardiac hormones. Both ANP and BNP preferentially bind to natriuretic peptide receptor-A (NPR-A or guanylyl cyslase-A) and exert similar effects through increases in intracellular cyclic guanosine monophosphate (cGMP) within target tissues. Expression and secretion of ANP and BNP are stimulated by various factors and are regulated via multiple signaling pathways. Human ANP has three molecular forms, α-ANP, β-ANP, and proANP (or γ-ANP), with proANP predominating in healthy atrial tissue. During secretion proANP is proteolytically processed by corin, resulting in secretion of bioactive α-ANP into the peripheral circulation. ProANP and β-ANP are minor forms in the circulation but are increased in patients with heart failure. The human BNP precursor proBNP is proteolytically processed to BNP1-32 and N-terminal proBNP (NT-proBNP) within ventricular myocytes. Uncleaved proBNP as well as mature BNP1-32 and NT-proBNP is secreted from the heart, and its secretion is increased in patients with heart failure. Mature BNP, its metabolites including BNP3-32, BNP4-32, and BNP5-32, and proBNP are all detected as immunoreactive-BNP by the current BNP assay system. We recently developed an assay system that specifically detects human proBNP. Using this assay system, we observed that miR30-GALNTs-dependent O-glycosylation in the N-terminal region of proBNP contributes to regulation of the processing and secretion of proBNP from the heart.
Collapse
Affiliation(s)
- Yasuaki Nakagawa
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Japan
| | - Toshio Nishikimi
- Department of Cardiovascular Medicine, Kyoto University Graduate School of Medicine, Japan; Department of Internal Medicine, Wakakusa-Tatsuma Rehabilitation Hospital, Japan
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, Shinshu University School of Medicine, Japan.
| |
Collapse
|
33
|
Matsuo A, Nagai-Okatani C, Nishigori M, Kangawa K, Minamino N. Natriuretic peptides in human heart: Novel insight into their molecular forms, functions, and diagnostic use. Peptides 2019; 111:3-17. [PMID: 30120963 DOI: 10.1016/j.peptides.2018.08.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/12/2018] [Accepted: 08/13/2018] [Indexed: 01/16/2023]
Abstract
Among the three natriuretic peptides, atrial/A-type natriuretic peptide (ANP) and brain/B-type natriuretic peptide (BNP) are primarily produced by, and secreted from, heart tissue. They maintain cardiovascular homeostasis by binding to natriuretic peptide receptor-A. Since plasma ANP and BNP concentrations, as well as expression, are elevated in response to increased body fluid volume and pressure load on the heart wall, these peptides are widely utilized as diagnostic biomarkers for evaluating heart failure. Regardless of their high utility, differences in their molecular forms between healthy and diseased subjects and how these relate to pathophysiology have not well been examined. Recent studies have shown that the circulating molecular forms of ANP and BNP are not uniform; bioactive α-ANP is the major ANP form, whereas the weakly active proBNP is the major BNP form. The relative ratios of the different molecular forms are altered under different pathophysiological conditions. These facts indicate that detailed measurements of each form may provide useful information on the pathophysiological state of heart tissue. Here, we revisit the relationship between the molecular forms of, and pathophysiological alterations in, human ANP and BNP and discuss the possible utility of the measurement of each of the molecular forms. The third peptide, C-type natriuretic peptide, activates natriuretic peptide receptor-B, but little is known about its production and function in the heart because of its extremely low levels. However, through recent studies, its role in the heart is gradually becoming clear. Here, we summarize its molecular forms, assay systems, and functions in the heart.
Collapse
Affiliation(s)
- Ayaka Matsuo
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Chiaki Nagai-Okatani
- Biotechnology Research Institute for Drug Discovery, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki 305-8568, Japan
| | - Mitsuhiro Nishigori
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Kenji Kangawa
- Research Institute, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan
| | - Naoto Minamino
- Omics Research Center, National Cerebral and Cardiovascular Center, Suita, Osaka 565-8565, Japan.
| |
Collapse
|
34
|
Natriuretic Peptide Receptor-C Protects Against Angiotensin II-Mediated Sinoatrial Node Disease in Mice. JACC Basic Transl Sci 2018; 3:824-843. [PMID: 30623142 PMCID: PMC6314975 DOI: 10.1016/j.jacbts.2018.08.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022]
Abstract
SAN disease is prevalent in hypertension and heart failure and can be induced by chronic Ang II treatment in mice. Ang II caused SAN disease in mice in association with impaired electrical conduction, reduction in the hyperpolarization-activated current (If) in SAN myocytes, and increased SAN fibrosis. Ang II-induced SAN disease was worsened in mice lacking NPR-C in association with enhanced SAN fibrosis. Mice co-treated with Ang II and an NPR-C agonist (cANF) were protected from SAN disease. NPR-C may represent a new target to protect against Ang II-induced SAN disease.
Sinoatrial node (SAN) disease mechanisms are poorly understood, and therapeutic options are limited. Natriuretic peptide(s) (NP) are cardioprotective hormones whose effects can be mediated partly by the NP receptor C (NPR-C). We investigated the role of NPR-C in angiotensin II (Ang II)-mediated SAN disease in mice. Ang II caused SAN disease due to impaired electrical activity in SAN myocytes and increased SAN fibrosis. Strikingly, Ang II treatment in NPR-C−/− mice worsened SAN disease, whereas co-treatment of wild-type mice with Ang II and a selective NPR-C agonist (cANF) prevented SAN dysfunction. NPR-C may represent a new target to protect against the development of Ang II-induced SAN disease.
Collapse
Key Words
- AP, action potential
- Ang II, angiotensin II
- CV, conduction velocity
- DD, diastolic depolarization
- Gmax, maximum conductance
- HR, heart rate
- ICa,L, L-type calcium current
- ICa,T, T-type calcium current
- INCX, sodium–calcium exchanger current
- IV, current voltage relationship
- If, hyperpolarization-activated current
- NP, natriuretic peptide
- NPR, natriuretic peptide receptor
- NPR-C, natriuretic peptide receptor C
- SAN, sinoatrial node
- SBP, systolic blood pressure
- V1/2(act), voltage for 50% channel activation
- cSNRT, corrected sinoatrial node recovery time
- fibrosis
- hypertension
- ion currents
- natriuretic peptide
- sinoatrial node
Collapse
|
35
|
Ohukainen P, Ruskoaho H, Rysa J. Cellular Mechanisms of Valvular Thickening in Early and Intermediate Calcific Aortic Valve Disease. Curr Cardiol Rev 2018; 14:264-271. [PMID: 30124158 PMCID: PMC6300797 DOI: 10.2174/1573403x14666180820151325] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 08/08/2018] [Accepted: 08/09/2018] [Indexed: 01/23/2023] Open
Abstract
Background: Calcific aortic valve disease is common in an aging population. It is an ac-tive atheroinflammatory process that has an initial pathophysiology and similar risk factors as athero-sclerosis. However, the ultimate disease phenotypes are markedly different. While coronary heart dis-ease results in rupture-prone plaques, calcific aortic valve disease leads to heavily calcified and ossi-fied valves. Both are initiated by the retention of low-density lipoprotein particles in the subendotheli-al matrix leading to sterile inflammation. In calcific aortic valve disease, the process towards calcifica-tion and ossification is preceded by valvular thickening, which can cause the first clinical symptoms. This is attributable to the accumulation of lipids, inflammatory cells and subsequently disturbances in the valvular extracellular matrix. Fibrosis is also increased but the innermost extracellular matrix layer is simultaneously loosened. Ultimately, the pathological changes in the valve cause massive calcifica-tion and bone formation - the main reasons for the loss of valvular function and the subsequent myo-cardial pathology. Conclusion: Calcification may be irreversible, and no drug treatments have been found to be effec-tive, thus it is imperative to emphasize lifestyle prevention of the disease. Here we review the mecha-nisms underpinning the early stages of the disease.
Collapse
Affiliation(s)
- Pauli Ohukainen
- Computational Medicine, Faculty of Medicine, University of Oulu and Biocenter Oulu, Oulu, Finland
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Jaana Rysa
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
36
|
Wu YF, Zhang DD, Liu SY, Luo HH, Jiang GM, Xu Y, Wu Y, Wang JJ, Liu FF, Samadli S, Wei W, Hu B, Hu P. C-Type Natriuretic Peptide Dampens Fibroblast Growth Factor-23 Expression Through MAPK Signaling Pathway in Human Mesangial Cells. J Interferon Cytokine Res 2018; 38:500-509. [PMID: 30335543 DOI: 10.1089/jir.2018.0051] [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: 01/20/2023] Open
Abstract
C-type natriuretic peptide (CNP) is believed to be produced locally in the kidneys and possess several renoprotective properties. In contrast, fibroblast growth factor (FGF) -23 elevates in the early stage of chronic kidney disease and predicts its outcomes. Currently, several studies have demonstrated that CNP and FGF-23 act through a close pathway, and moreover, FGF-23/mitogen-activated protein kinase (MAPK) can be obviously suppressed by CNP. In the present study, human mesangial cells (MCs) were incubated in serum-containing medium in the absence or presence of CNP (0, 10 and 100 pM) for 24, 48 and 72 h, respectively. CNP administration significantly suppresses MCs proliferation in a time- and dose-dependent manner. As a down-stream signaling of CNP activation, the expressions of natriuretic peptide receptor (NPR)-B, cyclic guanosine monophosphate-dependent protein kinases II and NPR-C were obviously augmented, whereas neutral endopeptidase expression was significantly decreased after CNP treatment in MCs. FGF-23, FGF receptor-1 and RAF-1 experienced a pronounced down-regulation in MCs with different doses of CNP throughout the whole observational period. CNP may dampen FGF-23 expression via MAPK signaling pathway in MCs.
Collapse
Affiliation(s)
- Yang Fang Wu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Dong Dong Zhang
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Si Yan Liu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Huang Huang Luo
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Guang Mei Jiang
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Yao Xu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Yue Wu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Jing Jing Wang
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Fei Fei Liu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Sama Samadli
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Wei Wei
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Bo Hu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| | - Peng Hu
- Department of Pediatrics, First Affiliated Hospital of Anhui Medical University , Hefei, People's Republic of China
| |
Collapse
|
37
|
Grassini DR, Lagendijk AK, De Angelis JE, Da Silva J, Jeanes A, Zettler N, Bower NI, Hogan BM, Smith KA. Nppa and Nppb act redundantly during zebrafish cardiac development to confine AVC marker expression and reduce cardiac jelly volume. Development 2018; 145:dev.160739. [PMID: 29752386 DOI: 10.1242/dev.160739] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 05/02/2018] [Indexed: 12/30/2022]
Abstract
Atrial natriuretic peptide (nppa/anf) and brain natriuretic peptide (nppb/bnp) form a gene cluster with expression in the chambers of the developing heart. Despite restricted expression, a function in cardiac development has not been demonstrated by mutant analysis. This is attributed to functional redundancy; however, their genomic location in cis has impeded formal analysis. Using genome editing, we have generated mutants for nppa and nppb, and found that single mutants were indistinguishable from wild type, whereas nppa/nppb double mutants displayed heart morphogenesis defects and pericardial oedema. Analysis of atrioventricular canal (AVC) markers show expansion of bmp4, tbx2b, has2 and versican expression into the atrium of double mutants. This expanded expression correlates with increased extracellular matrix in the atrium. Using a biosensor for hyaluronic acid to measure the cardiac jelly (cardiac extracellular matrix), we confirmed cardiac jelly expansion in nppa/nppb double mutants. Finally, bmp4 knockdown rescued the expansion of has2 expression and cardiac jelly in double mutants. This definitively shows that nppa and nppb function redundantly during cardiac development to restrict gene expression to the AVC, preventing excessive cardiac jelly synthesis in the atrial chamber.
Collapse
Affiliation(s)
- Daniela R Grassini
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Anne K Lagendijk
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jessica E De Angelis
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Jason Da Silva
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Angela Jeanes
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Nicole Zettler
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Neil I Bower
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Benjamin M Hogan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Kelly A Smith
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
38
|
Kawakami R, Lee CYW, Scott C, Bailey KR, Schirger JA, Chen HH, Benike SL, Cannone V, Martin FL, Sangaralingham SJ, Ichiki T, Burnett JC. A Human Study to Evaluate Safety, Tolerability, and Cyclic GMP Activating Properties of Cenderitide in Subjects With Stable Chronic Heart Failure. Clin Pharmacol Ther 2018; 104:546-552. [PMID: 29226471 PMCID: PMC5995613 DOI: 10.1002/cpt.974] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/20/2017] [Accepted: 11/28/2017] [Indexed: 12/11/2022]
Abstract
Cenderitide is a novel designer natriuretic peptide (NP) composed of C‐type natriuretic peptide (CNP) fused to the C‐terminus of Dendroaspis natriuretic peptide (DNP). Cenderitide was engineered to coactivate the two NP receptors, particulate guanylyl cyclase (pGC)‐A and ‐B. The rationale for its design was to achieve the renal‐enhancing and antifibrotic properties of dual receptor activation, but without clinically significant hypotension. Here we report the first clinical trial on the safety, tolerability, and cyclic guanosine monophosphate (cGMP) activating properties of Cenderitide in subjects with stable heart failure (HF). Four‐hour infusion of Cenderitide was safe, well‐tolerated, and significantly increased plasma cGMP levels and urinary cGMP excretion without adverse effects with no change in blood pressure. Thus, Cenderitide has a favorable safety profile and expected pharmacological effects in stable human HF. Our results support further investigations of Cenderitide in HF as a potential future cGMP‐enhancing therapeutic strategy.
Collapse
Affiliation(s)
- Rika Kawakami
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Candace Y W Lee
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Christopher Scott
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Kent R Bailey
- Department of Health Science Research, Mayo Clinic, Rochester, Minnesota, USA
| | - John A Schirger
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Horng H Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sherry L Benike
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Valentina Cannone
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Clinical and Experimental Medicine, University of Parma Medical School, Parma, Italy
| | - Fernando L Martin
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
39
|
Evolving Role of Natriuretic Peptides from Diagnostic Tool to Therapeutic Modality. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1067:109-131. [PMID: 29411335 DOI: 10.1007/5584_2018_143] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Natriuretic peptides (NP) are widely recognized as key regulators of blood pressure, water and salt homeostasis. In addition, they play a critical role in physiological cardiac growth and mediate a variety of biological effects including antiproliferative and anti-inflammatory effects in other organs and tissues. The cardiac release of NPs ANP and BNP represents an important compensatory mechanism during acute and chronic cardiac overload and during the pathogenesis of heart failure where their actions counteract the sustained activation of renin-angiotensin-aldosterone and other neurohormonal systems. Elevated circulating plasma NP levels correlate with the severity of heart failure and particularly BNP and the pro-peptide, NT-proBNP have been established as biomarkers for the diagnosis of heart failure as well as prognostic markers for cardiovascular risk. Despite activation of the NP system in heart failure it is inadequate to prevent progressive fluid and sodium retention and cardiac remodeling. Therapeutic approaches included administration of synthetic peptide analogs and the inhibition of NP-degrading enzyme neutral endopeptidase (NEP). Of all strategies only the combined NEP/ARB inhibition with sacubitril/valsartan had shown clinical success in reducing cardiovascular mortality and morbidity in patients with heart failure.
Collapse
|
40
|
Blaser MC, Wei K, Adams RLE, Zhou YQ, Caruso LL, Mirzaei Z, Lam AYL, Tam RKK, Zhang H, Heximer SP, Henkelman RM, Simmons CA. Deficiency of Natriuretic Peptide Receptor 2 Promotes Bicuspid Aortic Valves, Aortic Valve Disease, Left Ventricular Dysfunction, and Ascending Aortic Dilatations in Mice. Circ Res 2017; 122:405-416. [PMID: 29273600 DOI: 10.1161/circresaha.117.311194] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 01/25/2023]
Abstract
RATIONALE Aortic valve disease is a cell-mediated process without effective pharmacotherapy. CNP (C-type natriuretic peptide) inhibits myofibrogenesis and osteogenesis of cultured valve interstitial cells and is downregulated in stenotic aortic valves. However, it is unknown whether CNP signaling regulates aortic valve health in vivo. OBJECTIVE The aim of this study is to determine whether a deficient CNP signaling axis in mice causes accelerated progression of aortic valve disease. METHODS AND RESULTS In cultured porcine valve interstitial cells, CNP inhibited pathological differentiation via the guanylate cyclase NPR2 (natriuretic peptide receptor 2) and not the G-protein-coupled clearance receptor NPR3 (natriuretic peptide receptor 3). We used Npr2+/- and Npr2+/-;Ldlr-/- mice and wild-type littermate controls to examine the valvular effects of deficient CNP/NPR2 signaling in vivo, in the context of both moderate and advanced aortic valve disease. Myofibrogenesis in cultured Npr2+/- fibroblasts was insensitive to CNP treatment, whereas aged Npr2+/- and Npr2+/-;Ldlr-/- mice developed cardiac dysfunction and ventricular fibrosis. Aortic valve function was significantly impaired in Npr2+/- and Npr2+/-;Ldlr-/- mice versus wild-type littermates, with increased valve thickening, myofibrogenesis, osteogenesis, proteoglycan synthesis, collagen accumulation, and calcification. 9.4% of mice heterozygous for Npr2 had congenital bicuspid aortic valves, with worse aortic valve function, fibrosis, and calcification than those Npr2+/- with typical tricuspid aortic valves or all wild-type littermate controls. Moreover, cGK (cGMP-dependent protein kinase) activity was downregulated in Npr2+/- valves, and CNP triggered synthesis of cGMP and activation of cGK1 (cGMP-dependent protein kinase 1) in cultured porcine valve interstitial cells. Finally, aged Npr2+/-;Ldlr-/- mice developed dilatation of the ascending aortic, with greater aneurysmal progression in Npr2+/- mice with bicuspid aortic valves than those with tricuspid valves. CONCLUSIONS Our data establish CNP/NPR2 signaling as a novel regulator of aortic valve development and disease and elucidate the therapeutic potential of targeting this pathway to arrest disease progression.
Collapse
Affiliation(s)
- Mark C Blaser
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Kuiru Wei
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Rachel L E Adams
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Yu-Qing Zhou
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Laura-Lee Caruso
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Zahra Mirzaei
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Alan Y-L Lam
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Richard K K Tam
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Hangjun Zhang
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Scott P Heximer
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - R Mark Henkelman
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.)
| | - Craig A Simmons
- From the Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, Canada (M.C.B., R.L.E.A., Y.-Q.Z., L.-l.C., Z.M., A.Y.-L.L., R.K.K.T., H.Z., S.P.H., C.A.S.); Institute of Biomaterials and Biomedical Engineering (M.C.B., K.W., R.L.E.A., A.Y.-L.L., R.K.K.T., C.A.S.), Department of Physiology (H.Z., S.P.H.), and Department of Mechanical and Industrial Engineering (L.-l.C., Z.M., C.A.S.), University of Toronto, Ontario, Canada; and Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada (Y.-Q.Z., R.M.H.).
| |
Collapse
|
41
|
Compartmentation of Natriuretic Peptide Signalling in Cardiac Myocytes: Effects on Cardiac Contractility and Hypertrophy. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-3-319-54579-0_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
|
42
|
Lu J, Pan SS. Elevated C-type natriuretic peptide elicits exercise preconditioning-induced cardioprotection against myocardial injury probably via the up-regulation of NPR-B. J Physiol Sci 2017; 67:475-487. [PMID: 27557795 PMCID: PMC10717239 DOI: 10.1007/s12576-016-0477-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 08/08/2016] [Indexed: 02/02/2023]
Abstract
To evaluate exercise preconditioning (EP)-induced cardioprotective effects against exercise-induced acute myocardial injury and investigate the alterations of C-type natriuretic peptide (CNP) and its specific receptor, natriuretic peptide receptor B (NPR-B), during EP-induced cardioprotection. Rats were subjected to treadmill exercise as an EP model (4 periods of 10 min each at 30 m/min with intervening periods of rest lasting 10 min). High-intensity exercise was performed 0.5 and 24 h after the EP. EP attenuated high-intensity exercise-induced myocardial injury in both the early and late phases. After EP and high-intensity exercise, CNP and NPR-B levels increased robustly, but no alterations in the plasma CNP were observed. The enhanced NPR-B, plasma and tissue CNP, and its mRNA levels after high-intensity exercise were significantly elevated by EP. These results suggest that cardiac CNP and NPR-B play an important role in EP-mediated cardioprotection against high-intensity exercise-induced myocardial injury in rats.
Collapse
Affiliation(s)
- Jiao Lu
- School of Kinesiology, Shanghai University of Sport, 399 Changhai Road, Shanghai, 200438, China
| | - Shan-Shan Pan
- School of Kinesiology, Shanghai University of Sport, 399 Changhai Road, Shanghai, 200438, China.
| |
Collapse
|
43
|
Kuang DB, Zhou JP, Li MP, Tang J, Chen XP. Association of NPR3 polymorphism with risk of essential hypertension in a Chinese population. J Clin Pharm Ther 2017; 42:554-560. [PMID: 28497617 DOI: 10.1111/jcpt.12549] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 04/05/2017] [Indexed: 12/24/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Essential hypertension (EH) is a common disease exhibiting large individual difference in occurrence, development and treatment response. Genetic factors are implicated in the development and progression of EH. This study aimed to explore the association between NPR3 single nucleotide polymorphism rs2270915 (A/G, Asn521Asp) and the risk of EH in a Chinese Han population by a case-control study. METHODS The study was a single-centre, case-control trial, in which a total of 287 EH patients and 289 age- and sex-matched healthy controls were enrolled. The inclusion criteria were as follows: Han Chinese origin, male or female patients, systolic blood pressure (SBP) ≥140 mm Hg and/or diastolic blood pressure (DBP) ≥90 mm Hg. The healthy controls were subjects without histories of cardiovascular or cerebrovascular diseases. NPR3 rs2270915 polymorphism was genotyped by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). In addition, primary human umbilical vein endothelial cells (HUVECs) were isolated from 19 fresh human umbilical cords and cultured. Atrial natriuretic peptide (ANP) concentration in cell medium was determined by enzyme-linked immunosorbent assay (ELISA). NPR3 mRNA expression was determined by real-time semi-quantitative PCR. RESULTS AND DISCUSSION No significant difference in genotype distribution of NPR3 rs2270915 polymorphism was observed between cases and controls (P>.05). Patients carrying the rs2270915 G allele showed decreased SBP, and the difference was marginal. As compared with cells carrying the rs2270915 AA genotype, those with the AG genotype showed significantly lower NPR3 mRNA expression levels (P<.05) and lower medium ANP concentration (P<.001). WHAT IS NEW AND CONCLUSION This study suggested that NPR3 rs2270915 polymorphism was associated with decreased SBP level marginally in EH patients in a Chinese Han population, and the polymorphism may function through decreasing NPR3 mRNA expression and ANP level.
Collapse
Affiliation(s)
- D-B Kuang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
| | - J-P Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - M-P Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
| | - J Tang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
| | - X-P Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
| |
Collapse
|
44
|
Entresto (Sacubitril/Valsartan): Angiotensin Receptor Neprilysin Inhibition for Treating Heart Failure. CURRENT EMERGENCY AND HOSPITAL MEDICINE REPORTS 2017. [DOI: 10.1007/s40138-017-0137-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Rignault-Clerc S, Bielmann C, Liaudet L, Waeber B, Feihl F, Rosenblatt-Velin N. Natriuretic Peptide Receptor B modulates the proliferation of the cardiac cells expressing the Stem Cell Antigen-1. Sci Rep 2017; 7:41936. [PMID: 28181511 PMCID: PMC5299447 DOI: 10.1038/srep41936] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 01/04/2017] [Indexed: 12/21/2022] Open
Abstract
Brain Natriuretic Peptide (BNP) injections in adult “healthy” or infarcted mice led to increased number of non-myocyte cells (NMCs) expressing the nuclear transcription factor Nkx2.5. The aim of this study was to identify the nature of the cells able to respond to BNP as well as the signaling pathway involved. BNP treatment of neonatal mouse NMCs stimulated Sca-1+ cell proliferation. The Sca-1+ cells were characterized as being a mixed cell population involving fibroblasts and multipotent precursor cells. Thus, BNP treatment led also to increased number of Sca-1+ cells expressing Nkx2.5, in Sca-1+ cell cultures in vitro and in vivo, in the hearts of neonatal and adult infarcted mice. Whereas BNP induced Sca-1+ cell proliferation via NPR-B receptor and protein kinase G activation, CNP stimulated Sca-1+ cell proliferation via NPR-B and a PKG-independent mechanism. We highlighted here a new role for the natriuretic peptide receptor B which was identified as a target able to modulate the proliferation of the Sca-1+ cells. The involvement of NPR-B signaling in heart regeneration has, however, to be further investigated.
Collapse
Affiliation(s)
- Stéphanie Rignault-Clerc
- Unité de Physiopathologie Clinique Centre Hospitalier Universitaire Vaudois and University of Lausanne, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Christelle Bielmann
- Unité de Physiopathologie Clinique Centre Hospitalier Universitaire Vaudois and University of Lausanne, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Lucas Liaudet
- Service de Médecine Intensive Adulte, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Switzerland
| | - Bernard Waeber
- Unité de Physiopathologie Clinique Centre Hospitalier Universitaire Vaudois and University of Lausanne, Bugnon 7a, 1005 Lausanne, Switzerland
| | - François Feihl
- Unité de Physiopathologie Clinique Centre Hospitalier Universitaire Vaudois and University of Lausanne, Bugnon 7a, 1005 Lausanne, Switzerland
| | - Nathalie Rosenblatt-Velin
- Unité de Physiopathologie Clinique Centre Hospitalier Universitaire Vaudois and University of Lausanne, Bugnon 7a, 1005 Lausanne, Switzerland
| |
Collapse
|
46
|
Abstract
cGMP controls many cellular functions ranging from growth, viability, and differentiation to contractility, secretion, and ion transport. The mammalian genome encodes seven transmembrane guanylyl cyclases (GCs), GC-A to GC-G, which mainly modulate submembrane cGMP microdomains. These GCs share a unique topology comprising an extracellular domain, a short transmembrane region, and an intracellular COOH-terminal catalytic (cGMP synthesizing) region. GC-A mediates the endocrine effects of atrial and B-type natriuretic peptides regulating arterial blood pressure/volume and energy balance. GC-B is activated by C-type natriuretic peptide, stimulating endochondral ossification in autocrine way. GC-C mediates the paracrine effects of guanylins on intestinal ion transport and epithelial turnover. GC-E and GC-F are expressed in photoreceptor cells of the retina, and their activation by intracellular Ca(2+)-regulated proteins is essential for vision. Finally, in the rodent system two olfactorial GCs, GC-D and GC-G, are activated by low concentrations of CO2and by peptidergic (guanylins) and nonpeptidergic odorants as well as by coolness, which has implications for social behaviors. In the past years advances in human and mouse genetics as well as the development of sensitive biosensors monitoring the spatiotemporal dynamics of cGMP in living cells have provided novel relevant information about this receptor family. This increased our understanding of the mechanisms of signal transduction, regulation, and (dys)function of the membrane GCs, clarified their relevance for genetic and acquired diseases and, importantly, has revealed novel targets for therapies. The present review aims to illustrate these different features of membrane GCs and the main open questions in this field.
Collapse
Affiliation(s)
- Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
47
|
Chen S, Huang J, Zhao Q, Chen J, Jaquish CE, He J, Lu X, Yang X, Gu CC, Hixson JE, Liu F, Rice TK, Cao J, Chen J, Gu D. Associations Between Genetic Variants of the Natriuretic Peptide System and Blood Pressure Response to Dietary Sodium Intervention: The GenSalt Study. Am J Hypertens 2016. [PMID: 26224401 DOI: 10.1093/ajh/hpv129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The aim of this study was to comprehensively test the association of genetic variants in the natriuretic peptide (NP) system with blood pressure (BP) response to dietary sodium intervention in a Chinese population. METHODS We conducted a 7-day low-sodium intervention followed by a 7-day high-sodium intervention among 1,906 participants in rural China. BP measurements were obtained at baseline and each dietary intervention using a random-zero sphygmomanometer. Linear mixed-effect models were used to assess the associations of 48 single-nucleotide polymorphisms (SNPs) in 6 genes of NP system with BP response to dietary sodium intervention. RESULTS SNP rs5063 in the NPPA gene and SNP rs2077386 in the NPPC gene exhibited significant associations with BP response to low-sodium dietary intervention under recessive genetic model. For rs5063, absolute mean arterial pressure responses (95% confidence interval) to the low-sodium intervention were 1.31 (-1.08, 3.70) mm Hg for TT genotype and -3.74 (-4.01, -3.46) mm Hg for CC or TC genotype, respectively (P = 4.1 × 10(-5)). Individuals with at least one copy of the C allele of rs2077386 had significantly reduction in systolic BP during the low-sodium intervention compared to those with genotype GG with responses of -5.48 (-5.83, -5.14) vs. -2.76 (-3.52, -2.00) mm Hg, respectively (P = 1.9 × 10(-13)). CONCLUSIONS These novel findings suggested that genetic variants of NP system may contribute to the variation of BP response to sodium intervention in Chinese population. Certainly, replication of these results in other populations and further functional studies are warranted to clarify their role in the regulation of BP and hypertension.
Collapse
Affiliation(s)
- Shufeng Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China;
| | - Jianfeng Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Zhao
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jing Chen
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Cashell E Jaquish
- National Heart, Lung, and Blood Institute, National Institute of Health, Bethesda, Maryland, USA
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Xiangfeng Lu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xueli Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Charles C Gu
- School of Medicine, Washington University, St. Louis, Missouri, USA
| | - James E Hixson
- School of Public Health, University of Texas, Houston, Texas, USA
| | - Fangchao Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Treva K Rice
- School of Medicine, Washington University, St. Louis, Missouri, USA
| | - Jie Cao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jichun Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongfeng Gu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
48
|
Kimura T, Nojiri T, Hino J, Hosoda H, Miura K, Shintani Y, Inoue M, Zenitani M, Takabatake H, Miyazato M, Okumura M, Kangawa K. C-type natriuretic peptide ameliorates pulmonary fibrosis by acting on lung fibroblasts in mice. Respir Res 2016; 17:19. [PMID: 26895702 PMCID: PMC4761143 DOI: 10.1186/s12931-016-0335-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/13/2016] [Indexed: 12/31/2022] Open
Abstract
Background Pulmonary fibrosis has high rates of mortality and morbidity; however, no effective pharmacological therapy has been established. C-type natriuretic peptide (CNP), a member of the natriuretic peptide family, selectively binds to the transmembrane guanylyl cyclase (GC)-B receptor and exerts anti-inflammatory and anti-fibrotic effects in various organs through vascular endothelial cells and fibroblasts that have a cell-surface GC-B receptor. Given the pathophysiological importance of fibroblast activation in pulmonary fibrosis, we hypothesized that the anti-fibrotic and anti-inflammatory effects of exogenous CNP against bleomycin (BLM)-induced pulmonary fibrosis were exerted in part by the effect of CNP on pulmonary fibroblasts. Methods C57BL/6 mice were divided into two groups, CNP-treated (2.5 μg/kg/min) and vehicle, to evaluate BLM-induced (1 mg/kg) pulmonary fibrosis and inflammation. A periostin-CNP transgenic mouse model exhibiting CNP overexpression in fibroblasts was generated and examined for the anti-inflammatory and anti-fibrotic effects of CNP via fibroblasts in vivo. Additionally, we assessed CNP attenuation of TGF-β-induced differentiation into myofibroblasts by using immortalized human lung fibroblasts stably expressing GC-B receptors. Furthermore, to investigate whether CNP acts on human lung fibroblasts in a clinical setting, we obtained primary-cultured fibroblasts from surgically resected lungs of patients with lung cancer and analyzed levels of GC-B mRNA transcription. Results CNP reduced mRNA levels of the profibrotic cytokines interleukin (IL)-1β and IL-6, as well as collagen deposition and the fibrotic area in lungs of mice with bleomycin-induced pulmonary fibrosis. Furthermore, similar CNP effects were observed in transgenic mice exhibiting fibroblast-specific CNP overexpression. In cultured-lung fibroblasts, CNP treatment attenuated TGF-β–induced phosphorylation of Smad2 and increased mRNA and protein expression of α-smooth muscle actin and SM22α, indicating that CNP suppresses fibroblast differentiation into myofibroblasts. Furthermore, human lung fibroblasts from patients with or without interstitial lung disease substantially expressed GC-B receptor mRNA. Conclusions These data suggest that CNP ameliorates bleomycin-induced pulmonary fibrosis by suppressing TGF-β signaling and myofibroblastic differentiation in lung fibroblasts. Therefore, we propose consideration of CNP for clinical application to pulmonary fibrosis treatment.
Collapse
Affiliation(s)
- Toru Kimura
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan. .,Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita-City, Osaka, Japan.
| | - Takashi Nojiri
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan. .,Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita-City, Osaka, Japan.
| | - Jun Hino
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan.
| | - Hiroshi Hosoda
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita-City, Osaka, Japan.
| | - Koichi Miura
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan.
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita-City, Osaka, Japan.
| | - Masayoshi Inoue
- Department of General Thoracic Surgery, Kyoto Prefectural University of Medicine, Kyoto-City, Kyoto, Japan.
| | - Masahiro Zenitani
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan.
| | - Hiroyuki Takabatake
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan. .,Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita-City, Osaka, Japan.
| | - Mikiya Miyazato
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan.
| | - Meinoshin Okumura
- Department of General Thoracic Surgery, Osaka University Graduate School of Medicine, Suita-City, Osaka, Japan.
| | - Kenji Kangawa
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 5-7-1, Fujishirodai, Suita-city, Osaka, 565-8565, Japan.
| |
Collapse
|
49
|
Sangaralingham SJ, Wang BH, Huang L, Kumfu S, Ichiki T, Krum H, Burnett JC. Cardiorenal fibrosis and dysfunction in aging: Imbalance in mediators and regulators of collagen. Peptides 2016; 76:108-14. [PMID: 26774586 PMCID: PMC4754975 DOI: 10.1016/j.peptides.2016.01.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 12/19/2015] [Accepted: 01/08/2016] [Indexed: 01/01/2023]
Abstract
Cardiorenal fibrosis is a biological process that increases with age and contributes to dysfunction of the heart and kidney. While numerous circulating and tissue hormones, cytokines and enzymes have been identified in the development of cardiorenal fibrosis, several reports have suggested that the anti-fibrotic natriuretic peptide system (NPS), pro-fibrotic renin-angiotensin-aldosterone system (RAAS), transforming growth factor-beta 1 (TGF-β1), matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinases (TIMPs) are fundamental regulators and mediators of this process. However, the simultaneous assessment of these components in the development of age-mediated cardiorenal fibrotic remodeling is not completely understood. Thus, we assessed cardiorenal structure and function, the circulating NPS and RAAS and the cardiorenal tissue gene expression of collagen (Col) I, Col III, TGF-β1, MMP-9 and TIMP-1 in 2 and 20 month old Fischer rats. Our studies determined that aging was characterized by an increase in cardiorenal fibrosis that was accompanied with cardiorenal dysfunction. These alterations were associated with lower circulating atrial and C-type natriuretic peptides and higher angiotensin II and aldosterone levels in the aged rats. Moreover, we observed a decrease in Col I and III and an increase in TIMP- mRNA expressions in the aged heart and kidney, while TGF-β1 expression increased and MMP-9 decreased only in the aged kidney. We conclude that the age-mediated alterations in these fibrotic regulator and mediator profiles favors collagen accumulation due to an imbalance between the NPS and RAAS as well as a decline in the degradative pathway, thus suggesting a therapeutic opportunity to target these components.
Collapse
Affiliation(s)
- S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| | - Bing H Wang
- Centre of Cardiovascular Research and Education in Therapeutics, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Li Huang
- Centre of Cardiovascular Research and Education in Therapeutics, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Sirinart Kumfu
- Centre of Cardiovascular Research and Education in Therapeutics, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - Tomoko Ichiki
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | - Henry Krum
- Centre of Cardiovascular Research and Education in Therapeutics, School of Epidemiology and Preventive Medicine, Monash University, Melbourne, Australia
| | - John C Burnett
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
50
|
Moghtadaei M, Polina I, Rose RA. Electrophysiological effects of natriuretic peptides in the heart are mediated by multiple receptor subtypes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2016; 120:37-49. [DOI: 10.1016/j.pbiomolbio.2015.12.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/25/2015] [Accepted: 12/02/2015] [Indexed: 12/13/2022]
|