1
|
Shabani P, Dong F, Yun J, Shin SY, Dinchman A, Kundu D, Goodwill A, Gadd J, Pucci T, Kolz C, Shockling L, Yin L, Chilian W, Ohanyan V. Does coronary microvascular dysfunction play a role in heart failure with reduced ejection fraction? J Mol Cell Cardiol 2025; 200:61-67. [PMID: 39884552 DOI: 10.1016/j.yjmcc.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/31/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025]
Abstract
Heart failure (HF) is a conundrum in that, current therapies only slow the progression of the disease. We posit, if the causal mechanism were targeted, progression of the disease could be stopped and potentially reversed. We hypothesize that insufficient myocardial blood flow (MBF) produces minute areas of ischemia, that lead to an accumulating injury culminating in HF. Accordingly, we determined the relationship between MBF and cardiac work (wall stress-rate product [WSRP]) in control C57Bl6/J mice (Control), mice with transaortic constriction to produce HF (TAC-HF) and HF mice treated with the coronary vasodilator, chromonar (4 weeks of treatment, TAC-Chromonar). MBF and WSRP were measured during norepinephrine infusion in anesthetized mice. In Controls, MBF increased when work/WSRP was increased with norepinephrine, however, when cardiac work was increased in TAC-HF, MBF did not increase. After chromonar treatment, when work increased, MBF increased. Changes in cardiac function paralleled MBF, i.e., decrement in cardiac function occurred in TAC-HF (ejection fraction), but 4 weeks of chromonar treatment reversed this functional decline. We also found in a model of cardiac hypoxia fate-mapping, a 5-fold increase in the number of hypoxic cardiac myocytes (TAC-HF vs Control), which was reversed by chromonar. Capillary densities also followed this trend with a decrease from Control in TAC-HF, which was restored by Chromonar. We propose that a cause of HF is inadequate MBF to meet the metabolic demands of the working heart. Pharmacological coronary vasodilation with chromonar to increase MBF in HF can reverse the functional decline and improve cardiac function.
Collapse
Affiliation(s)
- Parisa Shabani
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - June Yun
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Song Yi Shin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Amber Dinchman
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Dipan Kundu
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Adam Goodwill
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - James Gadd
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Thomas Pucci
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Christopher Kolz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Lindsay Shockling
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - William Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH 44272, USA.
| |
Collapse
|
2
|
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
|
3
|
Bekele AT. Natriuretic Peptide Receptors (NPRs) as a Potential Target for the Treatment of Heart Failure. Curr Heart Fail Rep 2023; 20:429-440. [PMID: 37710133 DOI: 10.1007/s11897-023-00628-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE OF REVIEW Heart failure is defined as a complex clinical syndrome that results from any structural or functional impairment of ventricular filling or ejection of blood. The natriuretic peptide is known to exert its biological action on the kidney, heart, blood vessels, renin-angiotensin system, autonomous nervous system, and central nervous system. The natriuretic peptide-natriuretic receptor system plays an important role in the regulation of blood pressure and body fluid volume through its pleiotropic effects. RECENT FINDINGS The clinical and animal studies suggest that natriuretic peptide-natriuretic receptors are important targets for the treatment of heart failure and other cardiovascular diseases. Even though attempts targeting natriuretic peptide receptors are underway for heart failure treatment, they seem insufficient despite the receptor systems' potential. This review summarizes natriuretic peptide-natriuretic receptor system's physiological actions and potential target for the treatment of heart failure. Natriuretic peptides play multiple roles in different parts of the body, almost all of the activities related to this receptor system appear to have the potential to be harnessed to treat heart failure or symptoms associated with heart failure.
Collapse
Affiliation(s)
- Adamu T Bekele
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University, P.O. Box 9086, Addis Ababa, Ethiopia.
| |
Collapse
|
4
|
Bachmann JC, Kirchhoff JE, Napolitano JE, Sorota S, Gordon WM, Feric N, Aschar‐Sobbi R, Lv J, Cao Z, Coppieters K, Borghetti G, Nyberg M. C-type natriuretic peptide induces inotropic and lusitropic effects in human 3D-engineered cardiac tissue: Implications for the regulation of cardiac function in humans. Exp Physiol 2023; 108:1172-1188. [PMID: 37493451 PMCID: PMC10988518 DOI: 10.1113/ep091303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/06/2023] [Indexed: 07/27/2023]
Abstract
The role of C-type natriuretic peptide (CNP) in the regulation of cardiac function in humans remains to be established as previous investigations have been confined to animal model systems. Here, we used well-characterized engineered cardiac tissues (ECTs) generated from human stem cell-derived cardiomyocytes and fibroblasts to study the acute effects of CNP on contractility. Application of CNP elicited a positive inotropic response as evidenced by increases in maximum twitch amplitude, maximum contraction slope and maximum calcium amplitude. This inotropic response was accompanied by a positive lusitropic response as demonstrated by reductions in time from peak contraction to 90% of relaxation and time from peak calcium transient to 90% of decay that paralleled increases in maximum contraction decay slope and maximum calcium decay slope. To establish translatability, CNP-induced changes in contractility were also assessed in rat ex vivo (isolated heart) and in vivo models. Here, the effects on force kinetics observed in ECTs mirrored those observed in both the ex vivo and in vivo model systems, whereas the increase in maximal force generation with CNP application was only detected in ECTs. In conclusion, CNP induces a positive inotropic and lusitropic response in ECTs, thus supporting an important role for CNP in the regulation of human cardiac function. The high degree of translatability between ECTs, ex vivo and in vivo models further supports a regulatory role for CNP and expands the current understanding of the translational value of human ECTs. NEW FINDINGS: What is the central question of this study? What are the acute responses to C-type natriuretic peptide (CNP) in human-engineered cardiac tissues (ECTs) on cardiac function and how well do they translate to matched concentrations in animal ex vivo and in vivo models? What is the main finding and its importance? Acute stimulation of ECTs with CNP induced positive lusitropic and inotropic effects on cardiac contractility, which closely reflected the changes observed in rat ex vivo and in vivo cardiac models. These findings support an important role for CNP in the regulation of human cardiac function and highlight the translational value of ECTs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Juan Lv
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | - Zhiyou Cao
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | - Ken Coppieters
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| | | | - Michael Nyberg
- Research & Early DevelopmentNovo Nordisk A/SMaaloevDenmark
| |
Collapse
|
5
|
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: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [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
|
6
|
Cachorro E, Günscht M, Schubert M, Sadek MS, Siegert J, Dutt F, Bauermeister C, Quickert S, Berning H, Nowakowski F, Lämmle S, Firneburg R, Luo X, Künzel SR, Klapproth E, Mirtschink P, Mayr M, Dewenter M, Vettel C, Heijman J, Lorenz K, Guan K, El-Armouche A, Wagner M, Kämmerer S. CNP Promotes Antiarrhythmic Effects via Phosphodiesterase 2. Circ Res 2023; 132:400-414. [PMID: 36715019 PMCID: PMC9930893 DOI: 10.1161/circresaha.122.322031] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Ventricular arrhythmia and sudden cardiac death are the most common lethal complications after myocardial infarction. Antiarrhythmic pharmacotherapy remains a clinical challenge and novel concepts are highly desired. Here, we focus on the cardioprotective CNP (C-type natriuretic peptide) as a novel antiarrhythmic principle. We hypothesize that antiarrhythmic effects of CNP are mediated by PDE2 (phosphodiesterase 2), which has the unique property to be stimulated by cGMP to primarily hydrolyze cAMP. Thus, CNP might promote beneficial effects of PDE2-mediated negative crosstalk between cAMP and cGMP signaling pathways. METHODS To determine antiarrhythmic effects of cGMP-mediated PDE2 stimulation by CNP, we analyzed arrhythmic events and intracellular trigger mechanisms in mice in vivo, at organ level and in isolated cardiomyocytes as well as in human-induced pluripotent stem cell-derived cardiomyocytes. RESULTS In ex vivo perfused mouse hearts, CNP abrogated arrhythmia after ischemia/reperfusion injury. Upon high-dose catecholamine injections in mice, PDE2 inhibition prevented the antiarrhythmic effect of CNP. In mouse ventricular cardiomyocytes, CNP blunted the catecholamine-mediated increase in arrhythmogenic events as well as in ICaL, INaL, and Ca2+ spark frequency. Mechanistically, this was driven by reduced cellular cAMP levels and decreased phosphorylation of Ca2+ handling proteins. Key experiments were confirmed in human iPSC-derived cardiomyocytes. Accordingly, the protective CNP effects were reversed by either specific pharmacological PDE2 inhibition or cardiomyocyte-specific PDE2 deletion. CONCLUSIONS CNP shows strong PDE2-dependent antiarrhythmic effects. Consequently, the CNP-PDE2 axis represents a novel and attractive target for future antiarrhythmic strategies.
Collapse
Affiliation(s)
- Eleder Cachorro
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Günscht
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mario Schubert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Mirna S. Sadek
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Johanna Siegert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Fabian Dutt
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Carla Bauermeister
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Susann Quickert
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Henrik Berning
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Felix Nowakowski
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Simon Lämmle
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Rebecca Firneburg
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Xiaojing Luo
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Stephan R. Künzel
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Erik Klapproth
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Peter Mirtschink
- Institute of Clinical Chemistry and Laboratory Medicine, Department of Clinical Pathobiochemistry, University Hospital Dresden, Germany (P.M.)
| | - Manuel Mayr
- The James Black Centre, King’s College, University of London, United Kingdom (M.M.)
- Internal Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany (M.M.)
| | - Matthias Dewenter
- Department of Molecular Cardiology and Epigenetics, Heidelberg University, Germany (M.D.)
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
| | - Christiane Vettel
- DZHK (German Center for Cardiovascular Research), Partner Site, Heidelberg-Mannheim, Germany (M.D., C.V.)
- Institute of Experimental and Clinical Pharmacology and Toxicology, University Medical Center Mannheim, Germany (C.V.)
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine, and Life Sciences, Maastricht University, The Netherlands (J.H.)
| | - Kristina Lorenz
- Institut für Pharmakologie und Toxikologie, Julius-Maximilians-Universität Würzburg, Germany (K.L.)
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany (K.L.)
| | - Kaomei Guan
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Ali El-Armouche
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| | - Michael Wagner
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
- Bereich Rhythmologie, Klinik für Innere Medizin und Kardiologie, Herzzentrum Dresden, Dresden University of Technology, Germany (M.W.)
| | - Susanne Kämmerer
- Institut für Pharmakologie und Toxikologie, Technische Universität Dresden, Germany (E.C., M.G., M.S., M.S.S., J.S., F.D., C.B., S.Q., H.B., F.N., S.L., R.F., X.L., S.R.K., E.K., K.G., A.E.-A., M.W., S.K.)
| |
Collapse
|
7
|
Zou S, Khoo BL. Subtyping based on immune cell fractions reveal heterogeneity of cardiac fibrosis in end-stage heart failure. Front Immunol 2023; 14:1053793. [PMID: 36875078 PMCID: PMC9975711 DOI: 10.3389/fimmu.2023.1053793] [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: 09/26/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Background A central issue hindering the development of effective anti-fibrosis drugs for heart failure is the unclear interrelationship between fibrosis and the immune cells. This study aims at providing precise subtyping of heart failure based on immune cell fractions, elaborating their differences in fibrotic mechanisms, and proposing a biomarker panel for evaluating intrinsic features of patients' physiological statuses through subtype classification, thereby promoting the precision medicine for cardiac fibrosis. Methods We inferred immune cell type abundance of the ventricular samples by a computational method (CIBERSORTx) based on ventricular tissue samples from 103 patients with heart failure, and applied K-means clustering to divide patients into two subtypes based on their immune cell type abundance. We also designed a novel analytic strategy: Large-Scale Functional Score and Association Analysis (LAFSAA), to study fibrotic mechanisms in the two subtypes. Results Two subtypes of immune cell fractions: pro-inflammatory and pro-remodeling subtypes, were identified. LAFSAA identified 11 subtype-specific pro-fibrotic functional gene sets as the basis for personalised targeted treatments. Based on feature selection, a 30-gene biomarker panel (ImmunCard30) established for diagnosing patient subtypes achieved high classification performance, with the area under the receiver operator characteristic curve corresponding to 0.954 and 0.803 for the discovery and validation sets, respectively. Conclusion Patients with the two subtypes of cardiac immune cell fractions were likely having different fibrotic mechanisms. Patients' subtypes can be predicted based on the ImmunCard30 biomarker panel. We envision that our unique stratification strategy revealed in this study will unravel advance diagnostic techniques for personalised anti-fibrotic therapy.
Collapse
Affiliation(s)
- Shangjie Zou
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, Hong Kong SAR, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, Hong Kong SAR, China.,Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong-Shenzhen Futian Research Institute, Shenzhen, China
| |
Collapse
|
8
|
Ilieva M, Panella R, Uchida S. MicroRNAs in Cancer and Cardiovascular Disease. Cells 2022; 11:3551. [PMID: 36428980 PMCID: PMC9688578 DOI: 10.3390/cells11223551] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Although cardiac tumor formation is rare, accumulating evidence suggests that the two leading causes of deaths, cancers, and cardiovascular diseases are similar in terms of pathogenesis, including angiogenesis, immune responses, and fibrosis. These similarities have led to the creation of new exciting field of study called cardio-oncology. Here, we review the similarities between cancer and cardiovascular disease from the perspective of microRNAs (miRNAs). As miRNAs are well-known regulators of translation by binding to the 3'-untranslated regions (UTRs) of messenger RNAs (mRNAs), we carefully dissect how a specific set of miRNAs are both oncomiRs (miRNAs in cancer) and myomiRs (muscle-related miRNAs). Furthermore, from the standpoint of similar pathogenesis, miRNAs categories related to the similar pathogenesis are discussed; namely, angiomiRs, Immune-miRs, and fibromiRs.
Collapse
Affiliation(s)
| | | | - Shizuka Uchida
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, DK-2450 Copenhagen SV, Denmark
| |
Collapse
|
9
|
Wang C, Taskinen JH, Segersvärd H, Immonen K, Kosonen R, Tolva JM, Mäyränpää MI, Kovanen PT, Olkkonen VM, Sinisalo J, Laine M, Tikkanen I, Lakkisto P. Alterations of Cardiac Protein Kinases in Cyclic Nucleotide-Dependent Signaling Pathways in Human Ischemic Heart Failure. Front Cardiovasc Med 2022; 9:919355. [PMID: 35783854 PMCID: PMC9247256 DOI: 10.3389/fcvm.2022.919355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/13/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives Impaired protein kinase signaling is a hallmark of ischemic heart disease (IHD). Inadequate understanding of the pathological mechanisms limits the development of therapeutic approaches. We aimed to identify the key cardiac kinases and signaling pathways in patients with IHD with an effort to discover potential therapeutic strategies. Methods Cardiac kinase activity in IHD left ventricle (LV) and the related signaling pathways were investigated by kinomics, transcriptomics, proteomics, and integrated multi-omics approach. Results Protein kinase A (PKA) and protein kinase G (PKG) ranked on top in the activity shift among the cardiac kinases. In the IHD LVs, PKA activity decreased markedly compared with that of controls (62% reduction, p = 0.0034), whereas PKG activity remained stable, although the amount of PKG protein increased remarkably (65%, p = 0.003). mRNA levels of adenylate cyclases (ADCY 1, 3, 5, 9) and cAMP-hydrolysing phosphodiesterases (PDE4A, PDE4D) decreased significantly, although no statistically significant alterations were observed in that of PKGs (PRKG1 and PRKG2) and guanylate cyclases (GUCYs). The gene expression of natriuretic peptide CNP decreased remarkably, whereas those of BNP, ANP, and neprilysin increased significantly in the IHD LVs. Proteomics analysis revealed a significant reduction in protein levels of “Energy metabolism” and “Muscle contraction” in the patients. Multi-omics integration highlighted intracellular signaling by second messengers as the top enriched Reactome pathway. Conclusion The deficiency in cAMP/PKA signaling pathway is strongly implicated in the pathogenesis of IHD. Natriuretic peptide CNP could be a potential therapeutic target for the modulation of cGMP/PKG signaling.
Collapse
Affiliation(s)
- Chunguang Wang
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- *Correspondence: Chunguang Wang
| | - Juuso H. Taskinen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Heli Segersvärd
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Katariina Immonen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Riikka Kosonen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
| | - Johanna M. Tolva
- Transplantation Laboratory, Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Mikko I. Mäyränpää
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Petri T. Kovanen
- Atherosclerosis Research Laboratory, Wihuri Research Institute, Helsinki, Finland
| | - Vesa M. Olkkonen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juha Sinisalo
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mika Laine
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ilkka Tikkanen
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Päivi Lakkisto
- Minerva Foundation Institute for Medical Research, Biomedicum Helsinki 2 U, Helsinki, Finland
- Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Päivi Lakkisto
| |
Collapse
|
10
|
Natriuretic Peptide-Based Novel Therapeutics: Long Journeys of Drug Developments Optimized for Disease States. BIOLOGY 2022; 11:biology11060859. [PMID: 35741380 PMCID: PMC9219923 DOI: 10.3390/biology11060859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 12/11/2022]
Abstract
Simple Summary Natriuretic peptides are endogenous hormones produced in the heart and vascular endothelium, and they enable cardiorenal protective actions or bone growth via cGMP stimulation through their receptor guanylyl cyclase receptor A or B. To optimize the drug for each disease state, we must consider drug metabolism, delivery systems, and target receptor(s). This review summarizes attempts to develop novel natriuretic peptide-based therapeutics, including novel designer natriuretic peptides and oral drugs to enhance endogenous natriuretic peptides. We introduce some therapeutics that have been successful in clinical practice, as well as the prospective drug developments in the natriuretic peptide system for disease states. Abstract The field of natriuretic peptides (NPs) as an endocrine hormone has been developing since 1979. There are three peptides in humans: atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP), which bind to the guanylyl cyclase-A (GC-A) receptor (also called natriuretic peptide receptor-A (NPR-A)), and C-type natriuretic peptide (CNP), which binds to the GC-B receptor (also called the NPR-B) and then synthesizes intracellular cGMP. GC-A receptor stimulation has natriuretic, vasodilatory, cardiorenal protective and anti-renin–angiotensin–aldosterone system actions, and GC-B receptor stimulation can suppress myocardial fibrosis and can activate bone growth before epiphyseal plate closure. These physiological effects are useful as therapeutics for some disease states, such as heart failure, hypertension, and dwarfism. To optimize the therapeutics for each disease state, we must consider drug metabolism, delivery systems, and target receptor(s). We review the cardiac NP system; new designer NPs, such as modified/combined NPs and modified peptides that can bind to not only NP receptors but receptors for other systems; and oral drugs that enhance endogenous NP activity. Finally, we discuss prospective drug discoveries and the development of novel NP therapeutics.
Collapse
|
11
|
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
|
12
|
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
|
13
|
Polymers and Nanoparticles for Statin Delivery: Current Use and Future Perspectives in Cardiovascular Disease. Polymers (Basel) 2021; 13:polym13050711. [PMID: 33652927 PMCID: PMC7956757 DOI: 10.3390/polym13050711] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/21/2021] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis-related coronary artery disease (CAD) is one of the leading sources of mortality and morbidity in the world. Primary and secondary prevention appear crucial to reduce CAD-related complications. In this scenario, statin treatment was shown to be clinically effective in the reduction of adverse events, but systemic administration provides suboptimal results. As an attempt to improve bioavailability and effectiveness, polymers and nanoparticles for statin delivery were recently investigated. Polymers and nanoparticles can help statin delivery and their effects by increasing oral bioavailability or enhancing target-specific interaction, leading to reduced vascular endothelial dysfunction, reduced intimal hyperplasia, reduced ischemia-reperfusion injury, increased cardiac regeneration, positive remodeling in the extracellular matrix, reduced neointimal growth and increased re-endothelization. Moreover, some innovative aspects described in other cardiovascular fields could be translated into the CAD scenario. Recent preclinical studies are underlining the effect of statins in the stimulation and differentiation of endogenous cardiac stem cells, as well as in targeting of local adverse conditions implicated in atherosclerosis, and statin delivery through poly-lactic-co-glycolic acid (PLGA) appears the most promising aspect of current research to enhance drug activity. The present review intends to summarize the current evidence about polymers and nanoparticles for statin delivery in the field of cardiovascular disease, trying to shed light on this topic and identify new avenues for future studies.
Collapse
|
14
|
Garnier A, Bork NI, Jacquet E, Zipfel S, Muñoz-Guijosa C, Baczkó I, Reichenspurner H, Donzeau-Gouge P, Maier LS, Dobrev D, Girdauskas E, Nikolaev VO, Fischmeister R, Molina CE. Mapping genetic changes in the cAMP-signaling cascade in human atria. J Mol Cell Cardiol 2021; 155:10-20. [PMID: 33631188 DOI: 10.1016/j.yjmcc.2021.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/04/2021] [Accepted: 02/11/2021] [Indexed: 11/15/2022]
Abstract
AIM To obtain a quantitative expression profile of the main genes involved in the cAMP-signaling cascade in human control atria and in different cardiac pathologies. METHODS AND RESULTS Expression of 48 target genes playing a relevant role in the cAMP-signaling cascade was assessed by RT-qPCR. 113 samples were obtained from right atrial appendages (RAA) of patients in sinus rhythm (SR) with or without atrium dilation, paroxysmal atrial fibrillation (AF), persistent AF or heart failure (HF); and left atrial appendages (LAA) from patients in SR or with AF. Our results show that right and left atrial appendages in donor hearts or from SR patients have similar expression values except for AC7 and PDE2A. Despite the enormous chamber-dependent variability in the gene-expression changes between pathologies, several distinguishable patterns could be identified. PDE8A, PI3Kγ and EPAC2 were upregulated in AF. Different phosphodiesterase (PDE) families showed specific pathology-dependent changes. CONCLUSION By comparing mRNA-expression patterns of the cAMP-signaling cascade related genes in right and left atrial appendages of human hearts and across different pathologies, we show that 1) gene expression is not significantly affected by cardioplegic solution content, 2) it is appropriate to use SR atrial samples as controls, and 3) many genes in the cAMP-signaling cascade are affected in AF and HF but only few of them appear to be chamber (right or left) specific. TOPIC Genetic changes in human diseased atria. TRANSLATIONAL PERSPECTIVE The cyclic AMP signaling pathway is important for atrial function. However, expression patterns of the genes involved in the atria of healthy and diseased hearts are still unclear. We give here a general overview of how different pathologies affect the expression of key genes in the cAMP signaling pathway in human right and left atria appendages. Our study may help identifying new genes of interest as potential therapeutic targets or clinical biomarkers for these pathologies and could serve as a guide in future gene therapy studies.
Collapse
Affiliation(s)
- Anne Garnier
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France
| | - Nadja I Bork
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | - Eric Jacquet
- Université Paris-Saclay, Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Gif-sur-Yvette, France
| | - Svante Zipfel
- Dept. of Cardiovascular Surgery, University Heart Center Hamburg, Germany
| | | | - Istvan Baczkó
- Dept. Pharmacology and Pharmacotherapy, Univ. of Szeged, Hungary
| | | | | | - Lars S Maier
- Dept. Internal Medicine II, University Heart Center, University Hospital Regensburg, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West-German Heart and Vascular Center, Faculty of Medicine, University Duisburg-, Essen, Germany
| | - Evaldas Girdauskas
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany; Dept. of Cardiovascular Surgery, University Heart Center Hamburg, Germany
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| | | | - Cristina E Molina
- Université Paris-Saclay, Inserm, UMR-S 1180, Châtenay-Malabry, France; Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Germany; German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Germany
| |
Collapse
|
15
|
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: 18] [Impact Index Per Article: 3.6] [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
|
16
|
Affiliation(s)
- S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Yang Chen
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
17
|
Abstract
Heart failure (HF) is a common consequence of several cardiovascular diseases and is understood as a vicious cycle of cardiac and hemodynamic decline. The current inventory of treatments either alleviates the pathophysiological features (eg, cardiac dysfunction, neurohumoral activation, and ventricular remodeling) and/or targets any underlying pathologies (eg, hypertension and myocardial infarction). Yet, since these do not provide a cure, the morbidity and mortality associated with HF remains high. Therefore, the disease constitutes an unmet medical need, and novel therapies are desperately needed. Cyclic guanosine-3',5'-monophosphate (cGMP), synthesized by nitric oxide (NO)- and natriuretic peptide (NP)-responsive guanylyl cyclase (GC) enzymes, exerts numerous protective effects on cardiac contractility, hypertrophy, fibrosis, and apoptosis. Impaired cGMP signaling, which can occur after GC deactivation and the upregulation of cyclic nucleotide-hydrolyzing phosphodiesterases (PDEs), promotes cardiac dysfunction. In this study, we review the role that NO/cGMP and NP/cGMP signaling plays in HF. After considering disease etiology, the physiological effects of cGMP in the heart are discussed. We then assess the evidence from preclinical models and patients that compromised cGMP signaling contributes to the HF phenotype. Finally, the potential of pharmacologically harnessing cardioprotective cGMP to rectify the present paucity of effective HF treatments is examined.
Collapse
|
18
|
Krylatov AV, Tsibulnikov SY, Mukhomedzyanov AV, Boshchenko AA, Goldberg VE, Jaggi AS, Erben RG, Maslov LN. The Role of Natriuretic Peptides in the Regulation of Cardiac Tolerance to Ischemia/Reperfusion and Postinfarction Heart Remodeling. J Cardiovasc Pharmacol Ther 2020; 26:131-148. [PMID: 32840121 DOI: 10.1177/1074248420952243] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In the past 10 years, mortality from acute myocardial infarction has not decreased despite the widespread introduction of percutaneous coronary intervention. The reason for this situation is the absence in clinical practice of drugs capable of preventing reperfusion injury of the heart with high efficiency. In this regard, noteworthy natriuretic peptides (NPs) which have the infarct-limiting effect, prevent reperfusion cardiac injury, prevent adverse post-infarction remodeling of the heart. Atrial natriuretic peptide does not have the infarct-reducing effect in rats with alloxan-induced diabetes mellitus. NPs have the anti-apoptotic and anti-inflammatory effects. There is indirect evidence that NPs inhibit pyroptosis and autophagy. Published data indicate that NPs inhibit reactive oxygen species production in cardiomyocytes, aorta, heart, kidney and the endothelial cells. NPs can suppress aldosterone, angiotensin II, endothelin-1 synthesize and secretion. NPs inhibit the effects aldosterone, angiotensin II on the post-receptor level through intracellular signaling events. NPs activate guanylyl cyclase, protein kinase G and protein kinase A, and reduce phosphodiesterase 3 activity. NO-synthase and soluble guanylyl cyclase are involved in the cardioprotective effect of NPs. The cardioprotective effect of natriuretic peptides is mediated via activation of kinases (AMPK, PKC, PI3 K, ERK1/2, p70s6 k, Akt) and inhibition of glycogen synthase kinase 3β. The cardioprotective effect of NPs is mediated via sarcolemmal KATP channel and mitochondrial KATP channel opening. The cardioprotective effect of brain natriuretic peptide is mediated via MPT pore closing. The anti-fibrotic effect of NPs may be mediated through inhibition TGF-β1 expression. Natriuretic peptides can inhibit NF-κB activity and activate GATA. Hemeoxygenase-1 and peroxisome proliferator-activated receptor γ may be involved in the infarct-reducing effect of NPs. NPs exhibit the infarct-limiting effect in patients with acute myocardial infarction. NPs prevent post-infarction remodeling of the heart. To finally resolve the question of the feasibility of using NPs in AMI, a multicenter, randomized, blind, placebo-controlled study is needed to assess the effect of NPs on the mortality of patients after AMI.
Collapse
Affiliation(s)
- Andrey V Krylatov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Sergey Y Tsibulnikov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | | | - Alla A Boshchenko
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Victor E Goldberg
- Cancer Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| | - Amteshwar S Jaggi
- 429174Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Reinhold G Erben
- Department of Biomedical Research, Institute of Physiology, Pathophysiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - Leonid N Maslov
- Cardiology Research Institute, 164253Tomsk National Research Medical Center of the RAS, Tomsk, Russia
| |
Collapse
|
19
|
Watson CJ, Glezeva N, Horgan S, Gallagher J, Phelan D, McDonald K, Tolan M, Baugh J, Collier P, Ledwidge M. Atrial Tissue Pro-Fibrotic M2 Macrophage Marker CD163+, Gene Expression of Procollagen and B-Type Natriuretic Peptide. J Am Heart Assoc 2020; 9:e013416. [PMID: 32431194 PMCID: PMC7428985 DOI: 10.1161/jaha.119.013416] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Atrial tissue fibrosis is linked to inflammatory cells, yet is incompletely understood. A growing body of literature associates peripheral blood levels of the antifibrotic hormone BNP (B‐type natriuretic peptide) with atrial fibrillation (AF). We investigated the relationship between pro‐fibrotic tissue M2 macrophage marker Cluster of Differentiation (CD)163+, atrial procollagen expression, and BNP gene expression in patients with and without AF. Methods and Results In a cross‐sectional study design, right atrial tissue was procured from 37 consecutive, consenting, stable patients without heart failure or left ventricular systolic dysfunction, of whom 10 had AF and 27 were non‐AF controls. Samples were analyzed for BNP and fibro‐inflammatory gene expression, as well as fibrosis and CD163+. Primary analyses showed strong correlations (all P<0.008) between M2 macrophage CD163+ staining, procollagen gene expression, and myocardial BNP gene expression across the entire cohort. In secondary analyses without multiplicity adjustments, AF patients had greater left atrial volume index, more valve disease, higher serum BNP, and altered collagen turnover markers versus controls (all P<0.05). AF patients also showed higher atrial tissue M2 macrophage CD163+, collagen volume fraction, gene expression of procollagen 1 and 3, as well as reduced expression of the BNP clearance receptor NPRC (all P<0.05). Atrial procollagen 3 gene expression was correlated with fibrosis and BNP gene expression was correlated with serum BNP. Conclusions Elevated atrial tissue pro‐fibrotic M2 macrophage CD163+ is associated with increased myocardial gene expression of procollagen and anti‐fibrotic BNP and is higher in patients with AF. More work on modulation of BNP signaling for treatment and prevention of AF may be warranted.
Collapse
Affiliation(s)
- Chris J Watson
- Centre for Experimental Medicine Queen's University Belfast Belfast Northern Ireland.,Chronic Cardiovascular Disease Management Unit and Heart Failure Unit St Vincent's Healthcare Group Dublin Ireland.,School of Medicine UCD Conway Institute University College Dublin Dublin Ireland
| | - Nadezhda Glezeva
- Chronic Cardiovascular Disease Management Unit and Heart Failure Unit St Vincent's Healthcare Group Dublin Ireland.,School of Medicine UCD Conway Institute University College Dublin Dublin Ireland
| | - Stephen Horgan
- Sidney Kimmel Medical College Thomas Jefferson University Philadelphia PA
| | - Joe Gallagher
- Chronic Cardiovascular Disease Management Unit and Heart Failure Unit St Vincent's Healthcare Group Dublin Ireland.,School of Medicine UCD Conway Institute University College Dublin Dublin Ireland
| | - Dermot Phelan
- Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH
| | - Ken McDonald
- Chronic Cardiovascular Disease Management Unit and Heart Failure Unit St Vincent's Healthcare Group Dublin Ireland.,School of Medicine UCD Conway Institute University College Dublin Dublin Ireland
| | - Michael Tolan
- Cardiology Department Blackrock Clinic Dublin Ireland
| | - John Baugh
- School of Medicine UCD Conway Institute University College Dublin Dublin Ireland
| | - Patrick Collier
- Department of Cardiovascular Medicine Cleveland Clinic Cleveland OH
| | - Mark Ledwidge
- Chronic Cardiovascular Disease Management Unit and Heart Failure Unit St Vincent's Healthcare Group Dublin Ireland.,School of Medicine UCD Conway Institute University College Dublin Dublin Ireland
| |
Collapse
|
20
|
Ibrahim NE, McCarthy CP, Shrestha S, Gaggin HK, Mukai R, Szymonifka J, Apple FS, Burnett JC, Iyer S, Januzzi JL. Effect of Neprilysin Inhibition on Various Natriuretic Peptide Assays. J Am Coll Cardiol 2020; 73:1273-1284. [PMID: 30898202 DOI: 10.1016/j.jacc.2018.12.063] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND With sacubitril/valsartan treatment, B-type natriuretic peptide (BNP) concentrations increase; it remains unclear whether change in BNP concentrations is similar across all assays for its measurement. Effects of sacubitril/valsartan on atrial natriuretic peptide (ANP) concentrations in patients are unknown. Lastly, the impact of neprilysin inhibition on mid-regional pro-ANP (MR-proANP), N-terminal pro-BNP (NT-proBNP), proBNP1-108, or C-type natriuretic peptide (CNP) is not well understood. OBJECTIVES This study sought to examine the effects of sacubitril/valsartan on results from different natriuretic peptide assays. METHODS Twenty-three consecutive stable patients with heart failure and reduced ejection fraction were initiated and titrated on sacubitril/valsartan. Change in ANP, MR-proANP, BNP (using 5 assays), NT-proBNP (3 assays), proBNP1-108, and CNP were measured over 3 visits. RESULTS Average time to 3 follow-up visits was 22, 46, and 84 days. ANP rapidly and substantially increased with initiation and titration of sacubitril/valsartan, more than doubling by the first follow-up visit (+105.8%). Magnitude of ANP increase was greatest in those with concentrations above the median at baseline (+188%) compared with those with lower baseline concentrations (+44%); ANP increases were sustained. Treatment with sacubitril/valsartan led to inconsistent changes in BNP, which varied across methods assessed. Concentrations of MR-proANP, NT-proBNP, and proBNP1-108 variably declined after treatment; whereas CNP concentrations showed no consistent change. CONCLUSIONS Initiation and titration of sacubitril/valsartan led to variable changes in concentrations of multiple natriuretic peptides. These results provide important insights into the effects of sacubitril/valsartan treatment on individual patient results, and further suggest the benefit of neprilysin inhibition may be partially mediated by increased ANP concentrations.
Collapse
Affiliation(s)
- Nasrien E Ibrahim
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Cian P McCarthy
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
| | - Shreya Shrestha
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
| | - Hanna K Gaggin
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Renata Mukai
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Fred S Apple
- Laboratory Medicine & Pathology, Hennepin County Medical Center & University of Minnesota, Minneapolis, Minnesota
| | | | | | - James L Januzzi
- Cardiology Division, Massachusetts General Hospital, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts; Baim Institute for Clinical Research, Boston, Massachusetts.
| |
Collapse
|
21
|
Sridharan S, Kini RM, Richards AM. Venom natriuretic peptides guide the design of heart failure therapeutics. Pharmacol Res 2020; 155:104687. [PMID: 32057893 DOI: 10.1016/j.phrs.2020.104687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/24/2020] [Accepted: 02/07/2020] [Indexed: 12/29/2022]
Abstract
Heart failure (HF) affects over 26 million people world-wide. It is a syndrome triggered by loss of normal cardiac function due to many acute (eg myocardial infarction) and/or chronic (eg hypertension) causes and characterized by mixed beneficial and deleterious activation of a complex of multifaceted neurohormonal systems the net effect of which frequently is further adverse disruption of pressure-volume homeostasis. Unlike the situation in chronic heart failure, current strategies for treatment of acute heart failure are empirical and lack a strong evidence base. Management includes any of a combination of vasodilators, diuretics and ionotropic agents depending on the hemodynamic profile of the patient. Despite the improvement in the options available to improve outcomes in patients with chronic HF, for several decades little gain has been made in the treatment of the acute decompensated state. Morbidity and mortality rates remain high necessitating new therapeutic agents. The cardiac natriuretic peptides (NPs) are key hormones in pressure-volume homoeostasis. There are three isoforms of mammalian NPs, namely ANP, BNP and CNP. These peptides bind to membrane-bound NP receptors (NPRs) on the heart, vasculature and kidney to lower blood pressure and circulating volume. Intravenous infusion of NPs in HF patients improves hemodynamic status but is associated with occasional severe hypotension. Apart from mammalian NPs, snake venom NPs are an excellent source of pharmacologically distinct ligands that offer the possibility of engineering NPs for therapeutic purposes. Venom NPs have long half-lives, differential NPR activation profiles and varied NPR specificity. The scaffolds of venom NPs encode the molecular information for designing NPs with longer half-lives and improved and differential vascular and renal functions. This review focuses on the structure-function paradigm of mammalian and venom NPs and the different peptide engineering strategies that have been utilized in the design of clinically relevant new NP-analogues.
Collapse
Affiliation(s)
- Sindhuja Sridharan
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - R Manjunatha Kini
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore.
| | - Arthur Mark Richards
- Cardiac Department, National University Hospital, Cardiovascular Research Institute, National University Heart Centre, National University Health System, Singapore; Christchurch Heart Institute, University of Otago, NZ, United States.
| |
Collapse
|
22
|
Bartoli F, Bailey MA, Rode B, Mateo P, Antigny F, Bedouet K, Gerbaud P, Gosain R, Plante J, Norman K, Gomez S, Lefebvre F, Rucker-Martin C, Ainscough JFX, Kearney MT, Bruns AF, Shi J, Appleby HL, Young RS, Shawer HM, Debant M, Gomez AM, Beech DJ, Foster R, Benitah JP, Sabourin J. Orai1 Channel Inhibition Preserves Left Ventricular Systolic Function and Normal Ca 2+ Handling After Pressure Overload. Circulation 2020; 141:199-216. [PMID: 31906693 PMCID: PMC6970549 DOI: 10.1161/circulationaha.118.038891] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Orai1 is a critical ion channel subunit, best recognized as a mediator of store-operated Ca2+ entry (SOCE) in nonexcitable cells. SOCE has recently emerged as a key contributor of cardiac hypertrophy and heart failure but the relevance of Orai1 is still unclear. METHODS To test the role of these Orai1 channels in the cardiac pathophysiology, a transgenic mouse was generated with cardiomyocyte-specific expression of an ion pore-disruptive Orai1R91W mutant (C-dnO1). Synthetic chemistry and channel screening strategies were used to develop 4-(2,5-dimethoxyphenyl)-N-[(pyridin-4-yl)methyl]aniline (hereafter referred to as JPIII), a small-molecule Orai1 channel inhibitor suitable for in vivo delivery. RESULTS Adult mice subjected to transverse aortic constriction (TAC) developed cardiac hypertrophy and reduced ventricular function associated with increased Orai1 expression and Orai1-dependent SOCE (assessed by Mn2+ influx). C-dnO1 mice displayed normal cardiac electromechanical function and cellular excitation-contraction coupling despite reduced Orai1-dependent SOCE. Five weeks after TAC, C-dnO1 mice were protected from systolic dysfunction (assessed by preserved left ventricular fractional shortening and ejection fraction) even if increased cardiac mass and prohypertrophic markers induction were observed. This is correlated with a protection from TAC-induced cellular Ca2+ signaling alterations (increased SOCE, decreased [Ca2+]i transients amplitude and decay rate, lower SR Ca2+ load and depressed cellular contractility) and SERCA2a downregulation in ventricular cardiomyocytes from C-dnO1 mice, associated with blunted Pyk2 signaling. There was also less fibrosis in heart sections from C-dnO1 mice after TAC. Moreover, 3 weeks treatment with JPIII following 5 weeks of TAC confirmed the translational relevance of an Orai1 inhibition strategy during hypertrophic insult. CONCLUSIONS The findings suggest a key role of cardiac Orai1 channels and the potential for Orai1 channel inhibitors as inotropic therapies for maintaining contractility reserve after hypertrophic stress.
Collapse
Affiliation(s)
- Fiona Bartoli
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - Marc A Bailey
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Baptiste Rode
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Philippe Mateo
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - Fabrice Antigny
- Inserm, UMR-S 999, Université Paris-Saclay, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (F.A., C.R.M.)
| | - Kaveen Bedouet
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - Pascale Gerbaud
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - Rajendra Gosain
- School of Chemistry, University of Leeds, United Kingdom (R.G., J.P., K.N., R.F.)
| | - Jeffrey Plante
- School of Chemistry, University of Leeds, United Kingdom (R.G., J.P., K.N., R.F.)
| | - Katherine Norman
- School of Chemistry, University of Leeds, United Kingdom (R.G., J.P., K.N., R.F.)
| | - Susana Gomez
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - Florence Lefebvre
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - Catherine Rucker-Martin
- Inserm, UMR-S 999, Université Paris-Saclay, Centre Chirurgical Marie Lannelongue, Le Plessis-Robinson, France (F.A., C.R.M.)
| | - Justin F X Ainscough
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Alexander-Francisco Bruns
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Jian Shi
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Hollie L Appleby
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Richard S Young
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Heba M Shawer
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Marjolaine Debant
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Ana-Maria Gomez
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - David J Beech
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom (M.A.B., B.R., J.F.X.A., M.T.K., A.-F.B., J. Shi, H.L.A., R.S.Y., H.M.S., M.D., D.J.B.)
| | - Richard Foster
- School of Chemistry, University of Leeds, United Kingdom (R.G., J.P., K.N., R.F.)
| | - Jean-Pierre Benitah
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| | - Jessica Sabourin
- Inserm, UMR-S 1180, Signalisation et Physiopathologie Cardiovasculaire, Université Paris-Saclay, Châtenay-Malabry, France (F.B., P.M., K.B., P.G., S.G., F.L., A.-M.G., J.P.B., J. Sabourin)
| |
Collapse
|
23
|
Fu S, Chang Z, Luo L, Deng J. Therapeutic Progress and Knowledge Basis on the Natriuretic Peptide System in Heart Failure. Curr Top Med Chem 2019; 19:1850-1866. [PMID: 31448711 DOI: 10.2174/1568026619666190826163536] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 06/25/2019] [Accepted: 07/25/2019] [Indexed: 01/26/2023]
Abstract
Notwithstanding substantial improvements in diagnosis and treatment, Heart Failure (HF) remains a major disease burden with high prevalence and poor outcomes worldwide. Natriuretic Peptides (NPs) modulate whole cardiovascular system and exhibit multiple cardio-protective effects, including the counteraction of the Renin-Angiotensin-Aldosterone System (RAAS) and Sympathetic Nervous System (SNS), promotion of vasodilatation and natriuresis, and inhibition of hypertrophy and fibrosis. Novel pharmacological therapies based on NPs may achieve a valuable shift in managing patients with HF from inhibiting RAAS and SNS to a reversal of neurohormonal imbalance. Enhancing NP bioavailability through exogenous NP administration and inhibiting Neutral Endopeptidase (NEP) denotes valuable therapeutic strategies for HF. On the one hand, NEP-resistant NPs may be more specific as therapeutic choices in patients with HF. On the other hand, NEP Inhibitors (NEPIs) combined with RAAS inhibitors have proved to exert beneficial effects and reduce adverse events in patients with HF. Highly effective and potentially safe Angiotensin Receptor Blocker Neprilysin Inhibitors (ARNIs) have been developed after the failure of NEPIs and Vasopeptidase Inhibitors (VPIs) due to lacking efficacy and safety. Therapeutic progress and knowledge basis on the NP system in HF are summarized in the current review.
Collapse
Affiliation(s)
- Shihui Fu
- Department of Geriatric Cardiology, National Clinical Research Center of Geriatrics Disease, Beijing Key Laboratory of Precision Medicine for Chronic Heart Failure, Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhenyu Chang
- Department of Hepatobiliary and Pancreatic Surgical Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Leiming Luo
- Department of Geriatric Cardiology, National Clinical Research Center of Geriatrics Disease, Beijing Key Laboratory of Precision Medicine for Chronic Heart Failure, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Juelin Deng
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
24
|
Emelyanova L, Sra A, Schmuck EG, Raval AN, Downey FX, Jahangir A, Rizvi F, Ross GR. Impact of statins on cellular respiration and de-differentiation of myofibroblasts in human failing hearts. ESC Heart Fail 2019; 6:1027-1040. [PMID: 31520523 PMCID: PMC6816080 DOI: 10.1002/ehf2.12509] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 05/24/2019] [Accepted: 07/30/2019] [Indexed: 12/16/2022] Open
Abstract
AIMS Fibroblast to myofibroblast trans-differentiation with altered bioenergetics precedes cardiac fibrosis (CF). Either prevention of differentiation or promotion of de-differentiation could mitigate CF-related pathologies. We determined whether 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG-CoA) reductase inhibitors-statins, commonly prescribed to patients at risk of heart failure (HF)-can de-differentiate myofibroblasts, alter cellular bioenergetics, and impact the human ventricular fibroblasts (hVFs) in HF patients. METHODS AND RESULTS Either in vitro statin treatment of differentiated myofibroblasts (n = 3-6) or hVFs, isolated from human HF patients under statin therapy (HF + statin) vs. without statins (HF) were randomly used (n = 4-12). In vitro, hVFs were differentiated by transforming growth factor-β1 (TGF-β1) for 72 h (TGF-72 h). Differentiation status and cellular oxygen consumption rate (OCR) were determined by α-smooth muscle actin (α-SMA) expression and Seahorse assay, respectively. Data are mean ± SEM except Seahorse (mean ± SD); P < 0.05, considered significant. In vitro, statins concentration-dependently de-differentiated the myofibroblasts. The respective half-maximal effective concentrations were 729 ± 13 nmol/L (atorvastatin), 3.6 ± 1 μmol/L (rosuvastatin), and 185 ± 13 nmol/L (simvastatin). Mevalonic acid (300 μmol/L), the reduced product of HMG-CoA, prevented the statin-induced de-differentiation (α-SMA expression: 31.4 ± 10% vs. 58.6 ± 12%). Geranylgeranyl pyrophosphate (GGPP, 20 μmol/L), a cholesterol synthesis-independent HMG-CoA reductase pathway intermediate, completely prevented the statin-induced de-differentiation (α-SMA/GAPDH ratios: 0.89 ± 0.05 [TGF-72 h + 72 h], 0.63 ± 0.02 [TGF-72 h + simvastatin], and 1.2 ± 0.08 [TGF-72 h + simvastatin + GGPP]). Cellular metabolism involvement was observed when co-incubation of simvastatin (200 nmol/L) with glibenclamide (10 μmol/L), a KATP channel inhibitor, attenuated the simvastatin-induced de-differentiation (0.84 ± 0.05). Direct inhibition of mitochondrial respiration by oligomycin (1 ng/mL) also produced a de-differentiation effect (0.33 ± 0.02). OCR (pmol O2 /min/μg protein) was significantly decreased in the simvastatin-treated hVFs, including basal (P = 0.002), ATP-linked (P = 0.01), proton leak-linked (P = 0.01), and maximal (P < 0.001). The OCR inhibition was prevented by GGPP (basal OCR [P = 0.02], spare capacity OCR [P = 0.008], and maximal OCR [P = 0.003]). Congruently, hVFs from HF showed an increased population of myofibroblasts while HF + statin group showed significantly reduced cellular respiration (basal OCR [P = 0.021], ATP-linked OCR [P = 0.047], maximal OCR [P = 0.02], and spare capacity OCR [P = 0.025]) and myofibroblast differentiation (α-SMA/GAPDH: 1 ± 0.19 vs. 0.23 ± 0.06, P = 0.01). CONCLUSIONS This study demonstrates the de-differentiating effect of statins, the underlying GGPP sensitivity, reduced OCR with potential activation of KATP channels, and their impact on the differentiation magnitude of hVFs in HF patients. This novel pleiotropic effect of statins may be exploited to reduce excessive CF in patients at risk of HF.
Collapse
Affiliation(s)
- Larisa Emelyanova
- Center for Integrative Research on Cardiovascular Aging, Aurora Health Care, St. Luke's Medical Center, 2900 W. Oklahoma Ave, Milwaukee, WI, 53215, USA
| | - Amar Sra
- Center for Integrative Research on Cardiovascular Aging, Aurora Health Care, St. Luke's Medical Center, 2900 W. Oklahoma Ave, Milwaukee, WI, 53215, USA
| | - Eric G Schmuck
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Amish N Raval
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Francis X Downey
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, WI, USA
| | - Arshad Jahangir
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, WI, USA.,Center for Advanced Atrial Fibrillation Therapies, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, WI, USA
| | - Farhan Rizvi
- Center for Integrative Research on Cardiovascular Aging, Aurora Health Care, St. Luke's Medical Center, 2900 W. Oklahoma Ave, Milwaukee, WI, 53215, USA
| | - Gracious R Ross
- Center for Integrative Research on Cardiovascular Aging, Aurora Health Care, St. Luke's Medical Center, 2900 W. Oklahoma Ave, Milwaukee, WI, 53215, USA
| |
Collapse
|
25
|
Ross GR, Edwards S, Warner C, Homar P, Downey FX, Emelyanova L, Rizvi F, Jahangir A. Deletion of transcription factor AP-2α gene attenuates fibroblast differentiation into myofibroblast. J Cell Mol Med 2019; 23:6494-6498. [PMID: 31339227 PMCID: PMC6714505 DOI: 10.1111/jcmm.14421] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/26/2019] [Accepted: 04/18/2019] [Indexed: 12/18/2022] Open
Affiliation(s)
- Gracious R Ross
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Stacie Edwards
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Catherine Warner
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Peter Homar
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Francis X Downey
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Larisa Emelyanova
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Farhan Rizvi
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Arshad Jahangir
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| |
Collapse
|
26
|
Mohis M, Edwards S, Ryan S, Rizvi F, Tajik AJ, Jahangir A, Ross GR. Aging-related increase in store-operated Ca 2+ influx in human ventricular fibroblasts. Am J Physiol Heart Circ Physiol 2019; 315:H83-H91. [PMID: 29985070 DOI: 10.1152/ajpheart.00588.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Senescence-related fibrosis contributes to cardiac dysfunction. Profibrotic processes are Ca2+ dependent. The effect of aging on the Ca2+ mobilization processes of human ventricular fibroblasts (hVFs) is unclear. Therefore, we tested whether aging altered intracellular Ca2+ release and store-operated Ca2+ entry (SOCE). Disease-free hVFs from 2- to 63-yr-old trauma victims were assessed for cytosolic Ca2+ dynamics with fluo 3/confocal imaging. Angiotensin II or thapsigargin was used to release endoplasmic reticulum Ca2+ in Ca2+-free solution; CaCl2 (2 mM) was then added to assess SOCE, which was normalized to ionomycin-induced maximal Ca2+. The angiotensin II experiments were repeated after phosphoenolpyruvate pretreatment to determine the role of energy status. The expression of genes encoding SOCE-related ion channel subunits was assessed by quantitative PCR, and protein expression was assessed by immunoblot analysis. Age groups of <50 and ≥50 yr were compared using unpaired t-test or regression analysis. Ca2+ release by angiotensin II or thapsigargin was not different between the groups, but SOCE was significantly elevated in the ≥50-yr group. Regression analysis showed an age-dependent phosphoenolpyruvate-sensitive increase in SOCE of hVFs. Aging did not alter the mRNA expression of SOCE-related genes. The profibrotic phenotype of hVFs was evident by sprouty1 downregulation with age. Thus, an age-associated increase in angiotensin II- and thapsigargin-induced SOCE occurs in hVFs, independent of receptor mechanisms or alterations of mRNA expression level of SOCE-related ion channel subunits but related to the cellular bioenergetics status. Elucidation of mechanisms underlying enhanced hVF SOCE with aging may refine SOCE targets to limit aging-related progression of Ca2+-dependent cardiac fibrosis. NEW & NOTEWORTHY Human ventricular fibroblasts exhibit an age-related increase in store-operated Ca2+ influx induced by angiotensin II, an endogenous vasoactive hormone, or thapsigargin, an inhibitor of endoplasmic reticulum Ca2+-ATPase, independent of receptor mechanisms or genes encoding store-operated Ca2+ entry-related ion channel subunits. Selective inhibition of this augmented store-operated Ca2+ entry could therapeutically limit aging-related cardiac fibrosis.
Collapse
Affiliation(s)
- Momin Mohis
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Stacie Edwards
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Sean Ryan
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - Farhan Rizvi
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| | - A Jamil Tajik
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin.,Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers , Milwaukee, Wisconsin
| | - Arshad Jahangir
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin.,Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers , Milwaukee, Wisconsin
| | - Gracious R Ross
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin
| |
Collapse
|
27
|
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: 86] [Impact Index Per Article: 14.3] [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
|
28
|
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
|
29
|
Effects of the (Pro)renin Receptor on Cardiac Remodeling and Function in a Rat Alcoholic Cardiomyopathy Model via the PRR-ERK1/2-NOX4 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4546975. [PMID: 31049135 PMCID: PMC6462324 DOI: 10.1155/2019/4546975] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/09/2019] [Accepted: 01/27/2019] [Indexed: 12/14/2022]
Abstract
Alcoholic cardiomyopathy (ACM) caused by alcohol consumption manifests mainly as by maladaptive myocardial function, which eventually leads to heart failure and causes serious public health problems. The (pro)renin receptor (PRR) is an important member of the local tissue renin-angiotensin system and plays a vital role in many cardiovascular diseases. However, the mechanism responsible for the effects of PRR on ACM remains unclear. The purpose of this study was to determine the role of PRR in myocardial fibrosis and the deterioration of cardiac function in alcoholic cardiomyopathy. Wistar rats were fed a liquid diet containing 9% v/v alcohol to establish an alcoholic cardiomyopathy model. Eight weeks later, rats were injected with 1 × 109v.g./100 μl of recombinant adenovirus containing EGFP (scramble-shRNA), PRR, and PRR-shRNA via the tail vein. Cardiac function was assessed by echocardiography. Cardiac histopathology was measured by Masson's trichrome staining, immunohistochemical staining, and dihydroethidium staining. In addition, cardiac fibroblasts (CFs) were cultured to evaluate the effects of alcohol stimulation on the production of the extracellular matrix and their underlying mechanisms. Our results indicated that overexpression of PRR in rats with alcoholic cardiomyopathy exacerbates myocardial oxidative stress and myocardial fibrosis. Silencing of PRR expression with short hairpin RNA (shRNA) technology reversed the myocardial damage mediated by PRR. Additionally, PRR activated phosphorylation of ERK1/2 and increased NOX4-derived reactive oxygen species and collagen expression in CFs with alcohol stimulation. Administration of the ERK kinase inhibitor (PD98059) significantly reduced NOX4 protein expression and collagen production, which indicated that PRR increases collagen production primarily through the PRR-ERK1/2-NOX4 pathway in CFs. In conclusion, our study demonstrated that PRR induces myocardial fibrosis and deteriorates cardiac function through ROS from the PRR-ERK1/2-NOX4 pathway during ACM development.
Collapse
|
30
|
Keng BMH, Gao F, Tan RS, Ewe SH, Teo LLY, Xie BQ, Goh GBB, Koh WP, Koh AS. N-Terminal pro C-Type Natriuretic Peptide (NTproCNP) and myocardial function in ageing. PLoS One 2018; 13:e0209517. [PMID: 30566484 PMCID: PMC6300279 DOI: 10.1371/journal.pone.0209517] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/06/2018] [Indexed: 11/19/2022] Open
Abstract
Ageing-related alterations in cardiovascular structure and function are commonly associated with chronic inflammation. A potential blood-based biomarker indicative of a chronic inflammatory state is N-Terminal Pro C-Type Natriuretic Peptide (NTproCNP). We aim to investigate associations between NTproCNP and ageing-related impairments in cardiovascular function. Community-based participants underwent same-day assessment of cardiovascular function and circulating profiles of plasma NTproCNP. Associations between cardiovascular and biomarker profiles were studied in adjusted models including standard covariates. We studied 93 participants (mean age 73 ± 5.3 years, 36 women), of whom 55 (59%) had impaired myocardial relaxation (ratio of peak velocity flow in early diastole E (m/s) to peak velocity flow in late diastole by atrial contraction A (m/s) <0.84). Participants with impaired myocardial relaxation were also found to have lower peak early phase filling velocity (0.6 ± 0.1 vs 0.7 ± 0.1, p < 0.0001) and higher peak atrial phase filling velocity (0.9 ± 0.1 vs 0.7 ± 0.1, p < 0.0001). NTproCNP levelswere significantly lower among participants with impaired myocardial relaxation (16.4% vs 39.5% with NTproCNP ≥ 19, p = 0.012). After multivariable adjustments, NTproCNP was independently associated with impaired myocardial relaxation (OR 2.99, 95%CI 1.12–8.01, p = 0.029). Community elderly adults with myocardial ageing have lower NTproCNP levels compared to those with preserved myocardial function. Given that impaired myocardial relaxation probably represents early changes within the myocardium with ageing, NTproCNP may be useful as an ‘upstream’ biomarker useful for charting myocardial ageing.
Collapse
Affiliation(s)
| | - Fei Gao
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Ru San Tan
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - See Hooi Ewe
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | | | - Bei Qi Xie
- National Heart Centre Singapore, Singapore, Singapore
| | - George B. B. Goh
- Duke-NUS Medical School, Singapore, Singapore
- Singapore General Hospital, Singapore, Singapore
| | - Woon-Puay Koh
- Duke-NUS Medical School, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Angela S. Koh
- National Heart Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- * E-mail:
| |
Collapse
|
31
|
Day A, Jameson Z, Hyde C, Simbi B, Fowkes R, Lawson C. C-Type Natriuretic Peptide (CNP) Inhibition of Interferon-γ-Mediated Gene Expression in Human Endothelial Cells In Vitro. BIOSENSORS-BASEL 2018; 8:bios8030086. [PMID: 30223437 PMCID: PMC6164118 DOI: 10.3390/bios8030086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/24/2018] [Accepted: 09/11/2018] [Indexed: 01/14/2023]
Abstract
Cardiovascular diseases, including atherosclerosis, now account for more deaths in the Western world than from any other cause. Atherosclerosis has a chronic inflammatory component involving Th1 pro-inflammatory cytokines such as IFN-γ, which is known to induce endothelial cell inflammatory responses. On the other hand CNP, which acts via its receptors to elevate intracellular cGMP, is produced by endothelium and endocardium and is upregulated in atherosclerosis. It is believed to be protective, however its role in vascular inflammation is not well understood. The aim of this study was to investigate the effects of CNP on human endothelial cell inflammatory responses following IFN-γ stimulation. Human umbilical vein endothelial cells were treated with either IFN-γ (10 ng/mL) or CNP (100 nm), or both in combination, followed by analysis by flow cytometry for expression of MHC class I and ICAM-1. IFN-γ significantly increased expression of both molecules, which was significantly inhibited by CNP or the cGMP donor 8-Bromoguanosine 3',5'-cyclic monophosphate (1 µm). CNP also reduced IFN-γ mediated kynurenine generation by the IFN-γ regulated enzyme indoleamine-2,3-deoxygenase (IDO). We conclude that CNP downmodulates IFN-γ induced pro-inflammatory gene expression in human endothelial cells via a cGMP-mediated pathway. Thus, CNP may have a protective role in vascular inflammation and novel therapeutic strategies for CVD based on upregulation of endothelial CNP expression could reduce chronic EC inflammation.
Collapse
Affiliation(s)
- Amy Day
- Cardiovascular and Inflammation Biology Group, Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street London, NW1 0TU, UK.
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Zoe Jameson
- Cardiovascular and Inflammation Biology Group, Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street London, NW1 0TU, UK.
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Carolyn Hyde
- Bio-Analysis Centre, London Bioscience Innovation Centre, Royal College Street, London NW1 0NH, UK.
| | - Bigboy Simbi
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Robert Fowkes
- Endocrine Signalling Group, Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK.
| | - Charlotte Lawson
- Cardiovascular and Inflammation Biology Group, Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street London, NW1 0TU, UK.
| |
Collapse
|
32
|
Kodziszewska K, Sochanowicz B, Brzóska K, Kuśmierczyk M, Kuśmierski K, Śmigielski W, Piotrowski W, Kruszewski M, Leszek P. Natriuretic peptides and their receptors in failing heart - Functional changes and implications for treatment. Int J Cardiol 2018; 265:135-140. [PMID: 29724568 DOI: 10.1016/j.ijcard.2018.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 02/19/2018] [Accepted: 04/05/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Katarzyna Kodziszewska
- Department of Heart Failure and Transplantology, Institute of Cardiology, Warsaw, Poland.
| | - Barbara Sochanowicz
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Kamil Brzóska
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Mariusz Kuśmierczyk
- Department of Cardiac Surgery and Transplantology, Institute of Cardiology, Warsaw, Poland
| | - Krzysztof Kuśmierski
- Department of Cardiac Surgery and Transplantology, Institute of Cardiology, Warsaw, Poland
| | - Witold Śmigielski
- Unit of Demography and Social Gerontology at the University of Lodz, Poland; Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, Institute of Cardiology, Warsaw, Poland
| | - Walerian Piotrowski
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, Institute of Cardiology, Warsaw, Poland
| | - Marcin Kruszewski
- Centre for Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, Institute of Cardiology, Warsaw, Poland
| |
Collapse
|
33
|
Ichiki T, Dzhoyashvili N, Burnett JC. Natriuretic peptide based therapeutics for heart failure: Cenderitide: A novel first-in-class designer natriuretic peptide. Int J Cardiol 2018; 281:166-171. [PMID: 29941213 DOI: 10.1016/j.ijcard.2018.06.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 06/01/2018] [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 co-activate the two NP receptors, particulate guanylyl cyclase (pGC)-A and pGC-B. The rationale for its design was to achieve the renal-enhancing and anti-fibrotic properties of dual receptor activation, but without clinically significant hypotension. Here, we review the biology of the NPs and the rationale for their use in heart failure. Most importantly, we present the key studies related to the discovery of Cenderitide. Finally, we review the key clinical studies that have advanced this first-in-class dual NP receptor activator for heart failure.
Collapse
Affiliation(s)
- Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Department of Physiology and Bioengineering, College of Medicine Mayo Clinic, Rochester, MN, USA
| | - Nina Dzhoyashvili
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Department of Physiology and Bioengineering, College of Medicine Mayo Clinic, Rochester, MN, USA
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Department of Physiology and Bioengineering, College of Medicine Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
34
|
Di Marca S, Rando A, Cataudella E, Pulvirenti A, Alaimo S, Terranova V, Corriere T, Pisano M, Di Quattro R, Ronsisvalle ML, Giraffa CM, Giordano M, Stancanelli B, Malatino L. B-type natriuretic peptide may predict prognosis in older adults admitted with a diagnosis other than heart failure. Nutr Metab Cardiovasc Dis 2018; 28:636-642. [PMID: 29625779 DOI: 10.1016/j.numecd.2018.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/16/2018] [Accepted: 02/23/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS The diagnosis of heart failure (HF) in elderly patients is often difficult, due to overlap of typical signs and symptoms with those of comorbidities. B-type Natriuretic Peptide (BNP) predicts diagnosis and prognosis of HF, but little is known on its predictive role of short-term prognosis when admission diagnosis is other than HF. METHODS AND RESULTS We prospectively recruited 404 consecutive patients (aged≥65 years) hospitalized in the Unit of Internal Medicine, University of Catania, Catania, Italy, with an admission diagnosis other than HF. Clinical examination, laboratory data and BNP were evaluated at the admission. The predictive value of BNP and other variables for in-hospital mortality, thirty-day mortality and three month re-hospitalization was assessed. During hospitalization 48 (12%) patients died; by logistic regression analysis, in-hospital mortality was not predicted by BNP>600 pg/ml (OR = 1.36; CI 95% = 0.60-2.80; p = 0.4), while it was by chronic kidney disease (CKD, p < 0.001), WBC count (p < 0.001), immobilization syndrome (p < 0.008) and age (p = 0.012). After discharge, 54 patients (15%) died within 30 days; in these patients thirty-day mortality was significantly predicted by BNP>600 pg/ml (OR = 2.70; CI 95% = 1.40-5.00; p = 0.001), CKD (p < 0.001), malnutrition (p = 0.029) and age (p = 0.033). Re-hospitalized patients were 97 (32%); three month re-hospitalization was predicted by BNP>600 pg/ml (OR = 12.28; CI 95% = 6.00-24.90; p < 0.001) and anamnestic HF (p = 0.002). CONCLUSIONS Our study shows that BNP>600 pg/ml, CKD, malnutrition and age predict thirty-day mortality after discharge in elderly patients with an admission diagnosis other than HF, while CKD, WBC count, immobilization syndrome and age predict in-hospital mortality. Three-month re-hospitalization was predicted by BNP>600 pg/ml and anamnestic HF.
Collapse
Affiliation(s)
- S Di Marca
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - A Rando
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - E Cataudella
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - A Pulvirenti
- Unit of Bioinformatics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - S Alaimo
- Unit of Bioinformatics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - V Terranova
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - T Corriere
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - M Pisano
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - R Di Quattro
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - M L Ronsisvalle
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - C M Giraffa
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - M Giordano
- School of Emergency Medicine, Department of Medical, Surgical, Neurologic, Metabolic and Geriatric Sciences, Second University of Naples, Naples, Italy
| | - B Stancanelli
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy
| | - L Malatino
- Academic Unit of Internal Medicine, School of Emergency Medicine, Department of Clinical and Experimental Medicine, University of Catania, c/o Cannizzaro Hospital, Catania, Italy.
| |
Collapse
|
35
|
Fu S, Ping P, Wang F, Luo L. Synthesis, secretion, function, metabolism and application of natriuretic peptides in heart failure. J Biol Eng 2018; 12:2. [PMID: 29344085 PMCID: PMC5766980 DOI: 10.1186/s13036-017-0093-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
As a family of hormones with pleiotropic effects, natriuretic peptide (NP) system includes atrial NP (ANP), B-type NP (BNP), C-type NP (CNP), dendroaspis NP and urodilatin, with NP receptor-A (guanylate cyclase-A), NP receptor-B (guanylate cyclase-B) and NP receptor-C (clearance receptor). These peptides are genetically distinct, but structurally and functionally related for regulating circulatory homeostasis in vertebrates. In humans, ANP and BNP are encoded by NP precursor A (NPPA) and NPPB genes on chromosome 1, whereas CNP is encoded by NPPC on chromosome 2. NPs are synthesized and secreted through certain mechanisms by cardiomyocytes, fibroblasts, endotheliocytes, immune cells (neutrophils, T-cells and macrophages) and immature cells (embryonic stem cells, muscle satellite cells and cardiac precursor cells). They are mainly produced by cardiovascular, brain and renal tissues in response to wall stretch and other causes. NPs provide natriuresis, diuresis, vasodilation, antiproliferation, antihypertrophy, antifibrosis and other cardiometabolic protection. NPs represent body's own antihypertensive system, and provide compensatory protection to counterbalance vasoconstrictor-mitogenic-sodium retaining hormones, released by renin-angiotensin-aldosterone system (RAAS) and sympathetic nervous system (SNS). NPs play central roles in regulation of heart failure (HF), and are inactivated through not only NP receptor-C, but also neutral endopeptidase (NEP), dipeptidyl peptidase-4 and insulin degrading enzyme. Both BNP and N-terminal proBNP are useful biomarkers to not only make the diagnosis and assess the severity of HF, but also guide the therapy and predict the prognosis in patients with HF. Current NP-augmenting strategies include the synthesis of NPs or agonists to increase NP bioactivity and inhibition of NEP to reduce NP breakdown. Nesiritide has been established as an available therapy, and angiotensin receptor blocker NEP inhibitor (ARNI, LCZ696) has obtained extremely encouraging results with decreased morbidity and mortality. Novel pharmacological approaches based on NPs may promote a therapeutic shift from suppressing the RAAS and SNS to re-balancing neuroendocrine dysregulation in patients with HF. The current review discussed the synthesis, secretion, function and metabolism of NPs, and their diagnostic, therapeutic and prognostic values in HF.
Collapse
Affiliation(s)
- Shihui Fu
- Department of Geriatric Cardiology, Chinese People’s Liberation Army General Hospital, Beijing, 100853 China
- Department of Cardiology and Hainan Branch, Chinese People’s Liberation Army, General Hospital, Beijing, China
| | - Ping Ping
- Department of Pharmaceutical Care, Chinese People’s, Liberation Army General Hospital, Beijing, China
| | - Fengqi Wang
- Department of Cardiology and Hainan Branch, Chinese People’s Liberation Army, General Hospital, Beijing, China
| | - Leiming Luo
- Department of Geriatric Cardiology, Chinese People’s Liberation Army General Hospital, Beijing, 100853 China
| |
Collapse
|
36
|
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
|
37
|
The evolution of heart failure with reduced ejection fraction pharmacotherapy: What do we have and where are we going? Pharmacol Ther 2017; 178:67-82. [DOI: 10.1016/j.pharmthera.2017.03.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
38
|
Sharma V, Dogra N, Saikia UN, Khullar M. Transcriptional regulation of endothelial-to-mesenchymal transition in cardiac fibrosis: role of myocardin-related transcription factor A and activating transcription factor 3. Can J Physiol Pharmacol 2017; 95:1263-1270. [PMID: 28686848 DOI: 10.1139/cjpp-2016-0634] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The etiology of cardiac fibrogenesis is quite diverse, but a common feature is the presence of activated fibroblasts. Experimental evidence suggests that a subset of cardiac fibroblasts is derived via transition of vascular endothelial cells into fibroblasts by endothelial-to-mesenchymal transition (EndMT). During EndMT, endothelial cells lose their endothelial characteristics and acquire a mesenchymal phenotype. Molecular mechanisms and the transcriptional mediators controlling EndMT in heart during development or disease remain relatively undefined. Myocardin-related transcription factor A facilitates the transcription of cytoskeletal genes by serum response factor during fibrosis; therefore, its specific role in cardiac EndMT might be of importance. Activation of activating transcription factor 3 (ATF-3) during cardiac EndMT is speculative, since ATF-3 responds to a transforming growth factor β (TGF-β) stimulus and controls the expression of the primary epithelial-to-mesenchymal transition markers Snail, Slug, and Twist. Although the role of TGF-β in EndMT-mediated cardiac fibrosis has been established, targeting of the TGF-β ligand has not proven to be a viable anti-fibrotic strategy owing to the broad functional importance of this ligand. Thus, targeting of downstream transcriptional mediators may be a useful therapeutic approach in attenuating cardiac fibrosis. Here, we discuss some of the transcription factors that may regulate EndMT-mediated cardiac fibrosis and their involvement in type 2 diabetes.
Collapse
Affiliation(s)
- Vibhuti Sharma
- a Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160 012, India
| | - Nilambra Dogra
- b Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160 012, India
| | - Uma Nahar Saikia
- a Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160 012, India
| | - Madhu Khullar
- b Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh 160 012, India
| |
Collapse
|
39
|
Ichiki T, Huntley BK, Harty GJ, Sangaralingham SJ, Burnett JC. Early activation of deleterious molecular pathways in the kidney in experimental heart failure with atrial remodeling. Physiol Rep 2017; 5:5/9/e13283. [PMID: 28507167 PMCID: PMC5430128 DOI: 10.14814/phy2.13283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 04/20/2017] [Accepted: 04/22/2017] [Indexed: 12/20/2022] Open
Abstract
Heart failure (HF) is a major health problem with worsening outcomes when renal impairment is present. Therapeutics for early phase HF may be effective for cardiorenal protection, however the detailed characteristics of the kidney in early‐stage HF (ES‐HF), and therefore treatment for potential renal protection, are poorly defined. We sought to determine the gene and protein expression profiles of specific maladaptive pathways of ES‐HF in the kidney and heart. Experimental canine ES‐HF, characterized by de‐novo HF with atrial remodeling but not ventricular fibrosis, was induced by right ventricular pacing for 10 days. Kidney cortex (KC), medulla (KM), left ventricle (LV), and left atrial (LA) tissues from ES‐HF versus normal canines (n = 4 of each) were analyzed using RT‐PCR microarrays and protein assays to assess genes and proteins related to inflammation, renal injury, apoptosis, and fibrosis. ES‐HF was characterized by increased circulating natriuretic peptides and components of the renin‐angiotensin‐aldosterone system and decreased sodium and water excretion with mild renal injury and up‐regulation of CNP and renin genes in the kidney. Compared to normals, widespread genes, especially genes of the inflammatory pathways, were up‐regulated in KC similar to increases seen in LA. Protein expressions related to inflammatory cytokines were also augmented in the KC. Gene and protein changes were less prominent in the LV and KM. The ES‐HF displayed mild renal injury with widespread gene changes and increased inflammatory cytokines. These changes may provide important clues into the pathophysiology of ES‐HF and for therapeutic molecular targets in the kidney of ES‐HF.
Collapse
Affiliation(s)
- Tomoko Ichiki
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Brenda K Huntley
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Gail J Harty
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
40
|
Holditch SJ, Schreiber CA, Harris PC, LaRusso NF, Ramirez-Alvarado M, Cataliotti A, Torres VE, Ikeda Y. B-type natriuretic peptide overexpression ameliorates hepatorenal fibrocystic disease in a rat model of polycystic kidney disease. Kidney Int 2017; 92:657-668. [PMID: 28416225 DOI: 10.1016/j.kint.2017.02.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 02/05/2017] [Accepted: 02/16/2017] [Indexed: 12/15/2022]
Abstract
Polycystic kidney disease (PKD) involves progressive hepatorenal cyst expansion and fibrosis, frequently leading to end-stage renal disease. Increased vasopressin and cAMP signaling, dysregulated calcium homeostasis, and hypertension play major roles in PKD progression. The guanylyl cyclase A agonist, B-type natriuretic peptide (BNP), stimulates cGMP and shows anti-fibrotic, anti-hypertensive, and vasopressin-suppressive effects, potentially counteracting PKD pathogenesis. Here, we assessed the impacts of guanylyl cyclase A activation on PKD progression in a rat model of PKD. Sustained BNP production significantly reduced kidney weight, renal cystic indexes and fibrosis, in concert with suppressed hepatic cystogenesis in vivo. In vitro, BNP decreased cystic epithelial cell proliferation, suppressed fibrotic gene expression, and increased intracellular calcium. Together, our data demonstrate multifaceted effects of sustained activation of guanylyl cyclase A on polycystic kidney and liver disease. Thus, targeting the guanylyl cyclase A-cGMP axis may provide a novel therapeutic strategy for hepatorenal fibrocystic diseases.
Collapse
Affiliation(s)
- Sara J Holditch
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Claire A Schreiber
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic Translational Polycystic Kidney Disease Center, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic Translational Polycystic Kidney Disease Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Yasuhiro Ikeda
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
41
|
Ross GR, Bajwa T, Edwards S, Emelyanova L, Rizvi F, Holmuhamedov EL, Werner P, Downey FX, Tajik AJ, Jahangir A. Enhanced store-operated Ca 2+ influx and ORAI1 expression in ventricular fibroblasts from human failing heart. Biol Open 2017; 6:326-332. [PMID: 28126709 PMCID: PMC5374400 DOI: 10.1242/bio.022632] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Excessive cardiac fibrosis, characterized by increased collagen-rich extracellular matrix (ECM) deposition, is a major predisposing factor for mechanical and electrical dysfunction in heart failure (HF). The human ventricular fibroblast (hVF) remodeling mechanisms that cause excessive collagen deposition in HF are unclear, although reports suggest a role for intracellular free Ca2+ in fibrosis. Therefore, we determined the association of differences in cellular Ca2+ dynamics and collagen secretion/deposition between hVFs from failing and normal (control) hearts. Histology of left ventricle sections (Masson trichrome) confirmed excessive fibrosis in HF versus normal. In vitro, hVFs from HF showed increased secretion/deposition of soluble collagen in 48 h of culture compared with control [85.9±7.4 µg/106 cells vs 58.5±8.8 µg/106 cells, P<0.05; (Sircol™ assay)]. However, collagen gene expressions (COL1A1 and COL1A2; RT-PCR) were not different. Ca2+ imaging (fluo-3) of isolated hVFs showed no difference in the thapsigargin-induced intracellular Ca2+ release capacity (control 16±1.4% vs HF 17±1.1%); however, Ca2+ influx via store-operated Ca2+ entry/Ca2+ release-activated channels (SOCE/CRAC) was significantly (P≤0.05) greater in HF-hVFs (47±3%) compared with non-failing (35±5%). Immunoblotting for ICRAC channel components showed increased ORAI1 expression in HF-hVFs compared with normal without any difference in STIM1 expression. The Pearson's correlation coefficient for co-localization of STIM1/ORAI1 was significantly (P<0.01) greater in HF (0.5±0.01) than control (0.4±0.01) hVFs. The increase in collagen secretion of HF versus control hVFs was eliminated by incubation of hVFs with YM58483 (10 µM), a selective ICRAC inhibitor, for 48 h (66.78±5.87 µg/106 cells vs 55.81±7.09 µg/106 cells, P=0.27). In conclusion, hVFs from HF have increased collagen secretion capacity versus non-failing hearts and this is related to increase in Ca2+ entry via SOCE and enhanced expression of ORAI, the pore-forming subunit. Therapeutic inhibition of SOCE may reduce the progression of cardiac fibrosis/HF. Summary: The excessive collagen secretory phenotype found in failing human hearts is associated with ventricular fibroblast remodeling, caused by an elevated influx of intracellular calcium via SOC channels.
Collapse
Affiliation(s)
- Gracious R Ross
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, WI 53215, USA
| | - Tanvir Bajwa
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, WI 53215, USA
| | - Stacie Edwards
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, WI 53215, USA
| | - Larisa Emelyanova
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, WI 53215, USA
| | - Farhan Rizvi
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, WI 53215, USA
| | - Ekhson L Holmuhamedov
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, WI 53215, USA
| | - Paul Werner
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, WI 53215, USA
| | - Francis X Downey
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, WI 53215, USA
| | - A Jamil Tajik
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, WI 53215, USA
| | - Arshad Jahangir
- Center for Integrative Research on Cardiovascular Aging, Aurora Research Institute, Aurora Health Care, Milwaukee, WI 53215, USA .,Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, WI 53215, USA
| |
Collapse
|
42
|
Abstract
Endogenous natriuretic peptides serve as potent activators of particulate guanylyl cyclase receptors and the second messenger cGMP. Natriuretic peptides are essential in maintenance of volume homeostasis, and can be of myocardial, renal and endothelial origin. Advances in peptide engineering have permitted the ability to pursue highly innovative drug discovery strategies. This has resulted in designer natriuretic peptides that go beyond native peptides in efficacy, specificity, and resistance to enzymatic degradation. Together with recent improvements in peptide delivery systems, which have improved bioavailability, further advances in this field have been made. Therefore, designer natriuretic peptides with pleotropic actions together with strategies of chronic delivery have provided an unparalleled opportunity for the treatment of cardiovascular disease. In this review, we report the conceptual framework of peptide engineering of the natriuretic peptides that resulted in designer peptides for cardiovascular disease. We specifically provide an update on those currently in clinical trials for heart failure and hypertension, which include Cenderitide, ANX042 and ZD100.
Collapse
Affiliation(s)
- Laura M G Meems
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, College of Medicine Mayo Clinic, Rochester, MN
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, College of Medicine Mayo Clinic, Rochester, MN
| |
Collapse
|
43
|
Chen Y, Burnett JC. Biochemistry, Therapeutics, and Biomarker Implications of Neprilysin in Cardiorenal Disease. Clin Chem 2016; 63:108-115. [PMID: 28062615 DOI: 10.1373/clinchem.2016.262907] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/27/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Neprilysin (NEP) is a membrane-bound neutral endopeptidase that degrades a variety of bioactive peptides. The substrates include natriuretic peptides (NPs), which are important regulating mediators for cardiovascular and renal biology. Inhibition of NEP activity and exogenous NP administration thus have emerged as potential therapeutic strategies for treating cardiorenal diseases. More recently, B-type natriuretic peptide (BNP) or N-terminal-proBNP (NT-proBNP), 3'-5' cyclic guanosine monophosphate (cGMP), and soluble NEP as biomarkers have also been investigated in heart failure (HF) trials and their predictive value are beginning to be recognized. CONTENT The biological functions of NEP and NPs are discussed. Enhancing NPs through NEP inhibition combined with renin-angiotensin-aldosterone system (RAAS) antagonism has proved to be successful in HF treatment, although future surveillance studies will be required. Direct NP enhancement through peptide delivery may have fewer potentially hazardous effects compared to NEP inhibition. Strategies of combined inhibition on NEP with other cardiorenal pathophysiological pathways are promising. Finally, monitoring BNP/NT-proBNP/cGMP concentrations during NEP inhibition treatment may provide supplemental benefits to conventional biomarkers, and the identification of soluble NEP as a novel biomarker for HF needs further investigation. SUMMARY In this review, the biology of NEP is summarized, with a focus on NP regulation. The degradation of NPs by NEP provides the rationale for NEP inhibition as a strategy for cardiorenal disease treatment. We also describe the current therapeutic strategies of NEP inhibition and NP therapeutics in cardiorenal diseases. Moreover, the discovery of its circulating form, soluble NEP, as a biomarker is also discussed.
Collapse
Affiliation(s)
- Yang Chen
- Biochemistry and Molecular Biology Graduate Program, Mayo Graduate School, Rochester, MN; .,Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester MN
| | - John C Burnett
- Cardiorenal Research Laboratory, Department of Cardiovascular Diseases, Mayo Clinic, Rochester MN
| |
Collapse
|
44
|
Abstract
Despite >100 clinical trials, only 2 new drugs had been approved by the US Food and Drug Administration for the treatment of chronic heart failure in more than a decade: the aldosterone antagonist eplerenone in 2003 and a fixed dose combination of hydralazine-isosorbide dinitrate in 2005. In contrast, 2015 has witnessed the Food and Drug Administration approval of 2 new drugs, both for the treatment of chronic heart failure with reduced ejection fraction: ivabradine and another combination drug, sacubitril/valsartan or LCZ696. Seemingly overnight, a range of therapeutic possibilities, evoking new physiological mechanisms, promise great hope for a disease that often carries a prognosis worse than many forms of cancer. Importantly, the newly available therapies represent a culmination of basic and translational research that actually spans many decades. This review will summarize newer drugs currently being used in the treatment of heart failure, as well as newer strategies increasingly explored for their utility during the stages of the heart failure syndrome.
Collapse
Affiliation(s)
- Anjali Tiku Owens
- From the Cardiovascular Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Susan C Brozena
- From the Cardiovascular Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Mariell Jessup
- From the Cardiovascular Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia.
| |
Collapse
|
45
|
Peroxisome Proliferator-Activated Receptor-γ Is Critical to Cardiac Fibrosis. PPAR Res 2016; 2016:2198645. [PMID: 27293418 PMCID: PMC4880703 DOI: 10.1155/2016/2198645] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 04/16/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) is a ligand-activated transcription factor belonging to the nuclear receptor superfamily, which plays a central role in regulating lipid and glucose metabolism. However, accumulating evidence demonstrates that PPARγ agonists have potential to reduce inflammation, influence the balance of immune cells, suppress oxidative stress, and improve endothelial function, which are all involved in the cellular and molecular mechanisms of cardiac fibrosis. Thus, in this review we discuss the role of PPARγ in various cardiovascular conditions associated with cardiac fibrosis, including diabetes mellitus, hypertension, myocardial infarction, heart failure, ischemia/reperfusion injury, atrial fibrillation, and several other cardiovascular disease (CVD) conditions, and summarize the developmental status of PPARγ agonists for the clinical management of CVD.
Collapse
|
46
|
Vivar R, Humeres C, Muñoz C, Boza P, Bolivar S, Tapia F, Lavandero S, Chiong M, Diaz-Araya G. FoxO1 mediates TGF-beta1-dependent cardiac myofibroblast differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:128-38. [DOI: 10.1016/j.bbamcr.2015.10.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 10/02/2015] [Accepted: 10/26/2015] [Indexed: 12/31/2022]
|
47
|
Lee CYW, Huntley BK, McCormick DJ, Ichiki T, Sangaralingham SJ, Lisy O, Burnett JC. Cenderitide: structural requirements for the creation of a novel dual particulate guanylyl cyclase receptor agonist with renal-enhancing in vivo and ex vivo actions. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2015; 2:98-105. [PMID: 27340557 DOI: 10.1093/ehjcvp/pvv040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 09/23/2015] [Indexed: 12/11/2022]
Abstract
AIMS Cenderitide is a novel dual natriuretic peptide (NP) receptor chimeric peptide activator, which targets the particulate guanylyl cyclase B (pGC-B) receptor and pGC-A unlike native NPs. Cenderitide was engineered to retain the anti-fibrotic properties of C-type natriuretic peptide (CNP)/pGC-B with renal-enhancing actions facilitated by fusion to the carboxyl terminus of Dendroaspis NP (DNP), a pGC-A agonist, to CNP. Here, we address significance of the DNP carboxyl terminus in dual pGC receptor activation and actions of cenderitide compared with CNP on renal function and cyclic guanosine monophosphate (cGMP) in vivo and ex vivo in normal canines. METHODS AND RESULTS In vitro, only cenderitide and not CNP or three CNP-based variants was a potent dual pGC-A/pGC-B activator of cGMP production (from 5 to 237 pmol/mL) in human embryonic kidney (HEK) 293 cells overexpressing human pGC-A while in pGC-B overexpressing cells cenderitide increased cGMP production (from 4 to 321 pmol/mL) while the three CNP-based variants were weak agonists. Based upon our finding that the DNP carboxyl terminus is a key structural requirement for dual pGC-A/pGC-B activation, we defined in vivo the renal-enhancing actions of cenderitide compared with CNP. Cenderitide increased urinary cGMP excretion (from 989 to 5977 pmol/mL), net generation of renal cGMP (821-4124 pmol/min), natriuresis (12-242 μEq/min), and glomerular filtration rate (GFR) (37-51 mL/min) while CNP did not. We then demonstrated the transformation of CNP ex vivo into a renal cGMP-activating peptide which increased cGMP in freshly isolated glomeruli eight-fold greater than CNP. CONCLUSION The current study establishes that dual pGC-A and pGC-B activation with CNP requires the specific carboxyl terminus of DNP. In normal canines in vivo and in glomeruli ex vivo, the carboxyl terminus of DNP transforms CNP into a natriuretic and GFR-enhancing peptide. Future studies of cenderitide are warranted in cardiorenal disease states to explore its efficacy in overall cardiorenal homeostasis.
Collapse
Affiliation(s)
- Candace Y W Lee
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine, Bioengineering & Physiology, and Biochemistry and Molecular Biology, College of Medicine Mayo Clinic, Guggenheim 915, 200 First Street S.W., Rochester, MN 55905, USA
| | - Brenda K Huntley
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine, Bioengineering & Physiology, and Biochemistry and Molecular Biology, College of Medicine Mayo Clinic, Guggenheim 915, 200 First Street S.W., Rochester, MN 55905, USA
| | - Daniel J McCormick
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine, Bioengineering & Physiology, and Biochemistry and Molecular Biology, College of Medicine Mayo Clinic, Guggenheim 915, 200 First Street S.W., Rochester, MN 55905, USA
| | - Tomoko Ichiki
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine, Bioengineering & Physiology, and Biochemistry and Molecular Biology, College of Medicine Mayo Clinic, Guggenheim 915, 200 First Street S.W., Rochester, MN 55905, USA
| | - S Jeson Sangaralingham
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine, Bioengineering & Physiology, and Biochemistry and Molecular Biology, College of Medicine Mayo Clinic, Guggenheim 915, 200 First Street S.W., Rochester, MN 55905, USA
| | - Ondrej Lisy
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine, Bioengineering & Physiology, and Biochemistry and Molecular Biology, College of Medicine Mayo Clinic, Guggenheim 915, 200 First Street S.W., Rochester, MN 55905, USA
| | - John C Burnett
- Cardiorenal Research Laboratory, Division of Cardiovascular Diseases, Departments of Medicine, Bioengineering & Physiology, and Biochemistry and Molecular Biology, College of Medicine Mayo Clinic, Guggenheim 915, 200 First Street S.W., Rochester, MN 55905, USA
| |
Collapse
|
48
|
Sangaralingham SJ, McKie PM, Ichiki T, Scott CG, Heublein DM, Chen HH, Bailey KR, Redfield MM, Rodeheffer RJ, Burnett JC. Circulating C-type natriuretic peptide and its relationship to cardiovascular disease in the general population. Hypertension 2015; 65:1187-94. [PMID: 25895587 DOI: 10.1161/hypertensionaha.115.05366] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/31/2015] [Indexed: 11/16/2022]
Abstract
C-type natriuretic peptide (CNP) is an endothelium-derived peptide that is released as a protective mechanism in response cardiovascular injury or disease. However, no studies have investigated circulating CNP, identifying clinical factors that may influence CNP and its relationship to cardiovascular disease in the general population. We studied 1841 randomly selected subjects from Olmsted County, MN (mean age, 63±11 years; 48% men). Plasma CNP was measured by a well-established radioimmunoassay and echocardiography, clinical characterization, and detailed medical record review were performed. We report that CNP circulates at various concentrations (median, 13 pg/mL), was unaffected by sex, was weakly associated by age, and that highest quartile of CNP identified a high-risk phenotype. Subjects with CNP in the highest quartile were associated with increased risk of myocardial infarction (multivariable-adjusted hazard ratio, 1.51; 95% confidence interval, 1.09-2.09; P=0.01) but not heart failure, cerebrovascular accidents, or death during a follow-up of 12 years. Addition of the highest quartile of CNP to clinical variables led to a modest increase in the integrated discrimination improvement for risk of myocardial infarction. In a large community-based cohort, elevated circulating CNP identified a high-risk phenotype that included cardiovascular comorbidities and left ventricular dysfunction, and provided evidence that highest concentrations of CNP potentially has prognostic value in predicting future risk of myocardial infarction. Together, these data from the general population highlight the potential value of CNP and support the need for additional studies to evaluate whether mechanisms regulating CNP could improve outcomes.
Collapse
Affiliation(s)
- S Jeson Sangaralingham
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN.
| | - Paul M McKie
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - Tomoko Ichiki
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - Christopher G Scott
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - Denise M Heublein
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - Horng H Chen
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - Kent R Bailey
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - Margaret M Redfield
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - Richard J Rodeheffer
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| | - John C Burnett
- From the Cardiorenal Research Laboratory (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., J.C.B.), Divisions of Cardiovascular Diseases (S.J.S., P.M.M., T.I., D.M.H., H.H.C., M.M.R., R.J.R., J.C.B.), and Biomedical Statistics and Informatics (C.G.S., K.R.B.), Mayo Clinic, Rochester, MN
| |
Collapse
|