1
|
Chen S, Wang Q, Bakker D, Hu X, Zhang L, van der Made I, Tebbens AM, Kovácsházi C, Giricz Z, Brenner GB, Ferdinandy P, Schaart G, Gemmink A, Hesselink MKC, Rivaud MR, Pieper MP, Hollmann MW, Weber NC, Balligand JL, Creemers EE, Coronel R, Zuurbier CJ. Empagliflozin prevents heart failure through inhibition of the NHE1-NO pathway, independent of SGLT2. Basic Res Cardiol 2024; 119:751-772. [PMID: 39046464 PMCID: PMC11461573 DOI: 10.1007/s00395-024-01067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
Sodium glucose cotransporter 2 inhibitors (SGLT2i) constitute the only medication class that consistently prevents or attenuates human heart failure (HF) independent of ejection fraction. We have suggested earlier that the protective mechanisms of the SGLT2i Empagliflozin (EMPA) are mediated through reductions in the sodium hydrogen exchanger 1 (NHE1)-nitric oxide (NO) pathway, independent of SGLT2. Here, we examined the role of SGLT2, NHE1 and NO in a murine TAC/DOCA model of HF. SGLT2 knockout mice only showed attenuated systolic dysfunction without having an effect on other signs of HF. EMPA protected against systolic and diastolic dysfunction, hypertrophy, fibrosis, increased Nppa/Nppb mRNA expression and lung/liver edema. In addition, EMPA prevented increases in oxidative stress, sodium calcium exchanger expression and calcium/calmodulin-dependent protein kinase II activation to an equal degree in WT and SGLT2 KO animals. In particular, while NHE1 activity was increased in isolated cardiomyocytes from untreated HF, EMPA treatment prevented this. Since SGLT2 is not required for the protective effects of EMPA, the pathway between NHE1 and NO was further explored in SGLT2 KO animals. In vivo treatment with the specific NHE1-inhibitor Cariporide mimicked the protection by EMPA, without additional protection by EMPA. On the other hand, in vivo inhibition of NOS with L-NAME deteriorated HF and prevented protection by EMPA. In conclusion, the data support that the beneficial effects of EMPA are mediated through the NHE1-NO pathway in TAC/DOCA-induced heart failure and not through SGLT2 inhibition.
Collapse
Affiliation(s)
- Sha Chen
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Qian Wang
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Diane Bakker
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Xin Hu
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Liping Zhang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, USA
| | - Ingeborg van der Made
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Anna M Tebbens
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Csenger Kovácsházi
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary
| | - Zoltán Giricz
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary
- Pharmahungary Group, 6722, Szeged, Hungary
| | - Gábor B Brenner
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary
| | - Peter Ferdinandy
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary
- Pharmahungary Group, 6722, Szeged, Hungary
| | - Gert Schaart
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Anne Gemmink
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Mathilde R Rivaud
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michael P Pieper
- CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Markus W Hollmann
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Nina C Weber
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics, Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Esther E Creemers
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ruben Coronel
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology (L.E.I.C.A.), Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Aziz MK, Molony D, Monlezun D, Holder T, Brunckhorst O, Higgason N, Roland J, Magill R, Fatakdawala M, Iacobucci A, Mody-Bailey N, Owen C, Zarker A, Thames E, Swaby J, Xiao D, Choi L, Desai S, Galan J, Deng B, Hartshorne T, Nichols A, Zhang A, Imber J, Song J, Jones W, Rivas A, Sanchez D, Guhan M, Gandaglia G, Ranganath S, Jacob J, Howell S, Plana J, van den Bergh R, Roberts M, Sommer SG, Oldenburg J, Ploussard G, Tilki D, Schoots I, Briers E, Stranne J, Rouviere O, van Oort I, Oprea-Lager D, De Santis M, Cornford P. Prostate Cancer Therapy Cardiotoxicity Map (PROXMAP) for Advanced Disease States: A Systematic Review and Network Meta-analysis with Bayesian Modeling of Treatment Histories. Eur Urol 2024:S0302-2838(24)02569-7. [PMID: 39299896 DOI: 10.1016/j.eururo.2024.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/22/2024] [Accepted: 08/23/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND OBJECTIVE Recommendations of first-line therapies for metastatic hormone-sensitive (mHSPC), nonmetastatic castrate-resistant (M0CRPC), and metastatic castrate-resistant (mCRPC) prostate cancer do not account for cardiotoxicity due to a lack of clear prior evidence. This manuscript assesses cardiotoxicity of these therapies. METHODS We searched Ovid Medline, Elsevier Embase, and the Cochrane Library for randomized clinical trials (RCTs) from database inception to January 14, 2024. Network meta-analyses of first-line mHSPC, M0CRPC, and mCRPC therapies were constructed for the five cardiotoxicity metrics defined by the International Cardio-Oncology Society: heart failure, myocarditis, vascular toxicity, hypertension, and arrhythmias. Additional Bayesian network meta-analyses also accounted for prior treatment history. KEY FINDINGS AND LIMITATIONS Thirteen RCTs (16 292 patients) were included. For mHSPC, androgen deprivation therapy (ADT) plus docetaxel (DTX) plus abiraterone acetate (AA) with prednisone (P) demonstrated a significant increase in hypertension and arrhythmias versus ADT + DTX (risk ratio [RR] 2.85, 95% confidence interval [CI] 1.67-4.89, and RR 2.01, 95% CI 1.17-3.44, respectively); however, no corresponding differences were observed between ADT + DTX plus darolutamide (DAR) and ADT + DTX (RR 1.55, 95% CI 0.73-3.30, and RR 0.94, 95% CI 0.63-1.40, respectively). For mCRPC assuming a history of mHSPC treatment, ADT + AA + P plus olaparib (OLA) demonstrated a statistically significant decrease in hypertension versus ADT + AA + P (RR 0.20, 95% CI 0.16-0.26). M0CRPC results were unremarkable. CONCLUSIONS AND CLINICAL IMPLICATIONS For mHSPC, ADT + DTX + DAR demonstrates less cardiotoxicity than ADT + DTX + AA + P due to a lower risk of hypertension and arrhythmias from decreased mineralocorticoid excess. In addition, OLA counterintuitively offers decreased hypertension when superimposed on ADT + AA + P for mCRPC treatment after prior androgen deprivation from mHSPC therapy.
Collapse
Affiliation(s)
- Moez Karim Aziz
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA; Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Donald Molony
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dominique Monlezun
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Travis Holder
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Oliver Brunckhorst
- MRC Centre for Transplantation, Guy's Hospital Campus, King's College London, King's Health Partners, London, UK
| | - Noel Higgason
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jerry Roland
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Resa Magill
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Mariya Fatakdawala
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexander Iacobucci
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Neal Mody-Bailey
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Chris Owen
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Andrew Zarker
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Emma Thames
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Justin Swaby
- Department of Internal Medicine, University of Georgia, Augusta, GA, USA
| | - Daniel Xiao
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lily Choi
- Department of Internal Medicine, University of the Incarnate Word, San Antonio, TX, USA
| | - Shubh Desai
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jacob Galan
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Brett Deng
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Taylor Hartshorne
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Alexis Nichols
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Allan Zhang
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jared Imber
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jeffrey Song
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - William Jones
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alexis Rivas
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Darren Sanchez
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Maya Guhan
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Shreyas Ranganath
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jerril Jacob
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Skyler Howell
- McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Juan Plana
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Roderick van den Bergh
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Matthew Roberts
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Silke Gillessen Sommer
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Jan Oldenburg
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Guillaume Ploussard
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Derya Tilki
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Ivo Schoots
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Erik Briers
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Johan Stranne
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Olivier Rouviere
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Inge van Oort
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Daniela Oprea-Lager
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Maria De Santis
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| | - Philip Cornford
- Prostate Cancer Guidelines Panel, European Association of Urology, Arnhem, The Netherlands
| |
Collapse
|
3
|
Jalink EA, Schonk AW, Boon RA, Juni RP. Non-coding RNAs in the pathophysiology of heart failure with preserved ejection fraction. Front Cardiovasc Med 2024; 10:1300375. [PMID: 38259314 PMCID: PMC10800550 DOI: 10.3389/fcvm.2023.1300375] [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: 09/23/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is the largest unmet clinical need in cardiovascular medicine. Despite decades of research, the treatment option for HFpEF is still limited, indicating our ongoing incomplete understanding on the underlying molecular mechanisms. Non-coding RNAs, comprising of microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are non-protein coding RNA transcripts, which are implicated in various cardiovascular diseases. However, their role in the pathogenesis of HFpEF is unknown. Here, we discuss the role of miRNAs, lncRNAs and circRNAs that are involved in the pathophysiology of HFpEF, namely microvascular dysfunction, inflammation, diastolic dysfunction and cardiac fibrosis. We interrogated clinical evidence and dissected the molecular mechanisms of the ncRNAs by looking at the relevant in vivo and in vitro models that mimic the co-morbidities in patients with HFpEF. Finally, we discuss the potential of ncRNAs as biomarkers and potential novel therapeutic targets for future HFpEF treatment.
Collapse
Affiliation(s)
- Elisabeth A. Jalink
- Department of Physiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, Netherlands
| | - Amber W. Schonk
- Department of Physiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, Netherlands
| | - Reinier A. Boon
- Department of Physiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, Netherlands
- Institute for Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt am Main, Frankfurt am Main, Germany
- German Centre for Cardiovascular Research, Partner Site Frankfurt Rhein/Main, Frankfurt, Germany
| | - Rio P. Juni
- Department of Physiology, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, Netherlands
| |
Collapse
|
4
|
Allbritton-King JD, García-Cardeña G. Endothelial cell dysfunction in cardiac disease: driver or consequence? Front Cell Dev Biol 2023; 11:1278166. [PMID: 37965580 PMCID: PMC10642230 DOI: 10.3389/fcell.2023.1278166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
The vascular endothelium is a multifunctional cellular system which directly influences blood components and cells within the vessel wall in a given tissue. Importantly, this cellular interface undergoes critical phenotypic changes in response to various biochemical and hemodynamic stimuli, driving several developmental and pathophysiological processes. Multiple studies have indicated a central role of the endothelium in the initiation, progression, and clinical outcomes of cardiac disease. In this review we synthesize the current understanding of endothelial function and dysfunction as mediators of the cardiomyocyte phenotype in the setting of distinct cardiac pathologies; outline existing in vivo and in vitro models where key features of endothelial cell dysfunction can be recapitulated; and discuss future directions for development of endothelium-targeted therapeutics for cardiac diseases with limited existing treatment options.
Collapse
Affiliation(s)
- Jules D. Allbritton-King
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Guillermo García-Cardeña
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
5
|
Qiu Z, Fan Y, Wang Z, Huang F, Li Z, Sun Z, Hua S, Jin W, Chen Y. Catestatin Protects Against Diastolic Dysfunction by Attenuating Mitochondrial Reactive Oxygen Species Generation. J Am Heart Assoc 2023; 12:e029470. [PMID: 37119063 PMCID: PMC10227223 DOI: 10.1161/jaha.123.029470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/30/2023] [Indexed: 04/30/2023]
Abstract
Background Catestatin has been reported as a pleiotropic cardioprotective peptide. Heart failure with preserved ejection fraction (HFpEF) was considered a heterogeneous syndrome with a complex cause. We sought to investigate the role of catestatin in HFpEF and diastolic dysfunction. METHODS AND RESULTS Administration of recombinant catestatin (1.5 mg/kg/d) improved diastolic dysfunction and left ventricular chamber stiffness in transverse aortic constriction mice with deoxycorticosterone acetate pellet implantation, as reflected by Doppler tissue imaging and pressure-volume loop catheter. Less cardiac hypertrophy and myocardial fibrosis was observed, and transcriptomic analysis revealed downregulation of mitochondrial electron transport chain components after catestatin treatment. Catestatin reversed mitochondrial structural and respiratory chain component abnormality, decreased mitochondrial proton leak, and reactive oxygen species generation in myocardium. Excessive oxidative stress induced by Ru360 abolished catestatin treatment effects on HFpEF-like cardiomyocytes in vitro, indicating the beneficial role of catestatin in HFpEF as a mitochondrial ETC modulator. The serum concentration of catestatin was tested among 81 patients with HFpEF and 76 non-heart failure controls. Compared with control subjects, serum catestatin concentration was higher in patients with HFpEF and positively correlated with E velocity to mitral annular e' velocity ratio, indicating a feedback compensation role of catestatin in HFpEF. Conclusions Catestatin protects against diastolic dysfunction in HFpEF through attenuating mitochondrial electron transport chain-derived reactive oxygen species generation. Serum catestatin concentration is elevated in patients with HFpEF, probably as a relatively insufficient but self-compensatory mechanism.
Collapse
Affiliation(s)
- Zeping Qiu
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
| | - Yingze Fan
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
| | - Zhiyan Wang
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
| | - Fanyi Huang
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
| | - Zhuojin Li
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
| | - Zhihong Sun
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
| | - Sha Hua
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Heart Failure Center, Ruijin Hospital, & Lu Wan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Jin
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Heart Failure Center, Ruijin Hospital, & Lu Wan BranchShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yanjia Chen
- Department of Cardiovascular Medicine, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
- Institute of Cardiovascular DiseasesShanghai Jiao Tong University School of MedicineShanghaiPeople’s Republic of China
| |
Collapse
|
6
|
Wang M, Yang Y, Xu Y. Brain nuclear receptors and cardiovascular function. Cell Biosci 2023; 13:14. [PMID: 36670468 PMCID: PMC9854230 DOI: 10.1186/s13578-023-00962-3] [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: 12/21/2022] [Accepted: 01/12/2023] [Indexed: 01/22/2023] Open
Abstract
Brain-heart interaction has raised up increasing attentions. Nuclear receptors (NRs) are abundantly expressed in the brain, and emerging evidence indicates that a number of these brain NRs regulate multiple aspects of cardiovascular diseases (CVDs), including hypertension, heart failure, atherosclerosis, etc. In this review, we will elaborate recent findings that have established the physiological relevance of brain NRs in the context of cardiovascular function. In addition, we will discuss the currently available evidence regarding the distinct neuronal populations that respond to brain NRs in the cardiovascular control. These findings suggest connections between cardiac control and brain dynamics through NR signaling, which may lead to novel tools for the treatment of pathological changes in the CVDs.
Collapse
Affiliation(s)
- Mengjie Wang
- grid.508989.50000 0004 6410 7501Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yongjie Yang
- grid.508989.50000 0004 6410 7501Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA
| | - Yong Xu
- grid.508989.50000 0004 6410 7501Department of Pediatrics, USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX USA ,grid.39382.330000 0001 2160 926XDepartment of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
7
|
Holder ER, Alibhai FJ, Caudle SL, McDermott JC, Tobin SW. The importance of biological sex in cardiac cachexia. Am J Physiol Heart Circ Physiol 2022; 323:H609-H627. [PMID: 35960634 DOI: 10.1152/ajpheart.00187.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiac cachexia is a catabolic muscle wasting syndrome observed in approximately 1 in 10 heart failure patients. Increased skeletal muscle atrophy leads to frailty and limits mobility which impacts quality of life, exacerbates clinical care, and is associated with higher rates of mortality. Heart failure is known to exhibit a wide range of prevalence and severity when examined across individuals of different ages and with co-morbidities related to diabetes, renal failure and pulmonary dysfunction. It is also recognized that men and women exhibit striking differences in the pathophysiology of heart failure as well as skeletal muscle homeostasis. Given that both skeletal muscle and heart failure physiology are in-part sex dependent, the diagnosis and treatment of cachexia in heart failure patients may depend on a comprehensive examination of how these organs interact. In this review we explore the potential for sex-specific differences in cardiac cachexia. We summarize advantages and disadvantages of clinical methods used to measure muscle mass and function and provide alternative measurements that should be considered in preclinical studies. Additionally, we summarize sex-dependent effects on muscle wasting in preclinical models of heart failure, disuse, and cancer. Lastly, we discuss the endocrine function of the heart and outline unanswered questions that could directly impact patient care.
Collapse
|
8
|
Cornuault L, Rouault P, Duplàa C, Couffinhal T, Renault MA. Endothelial Dysfunction in Heart Failure With Preserved Ejection Fraction: What are the Experimental Proofs? Front Physiol 2022; 13:906272. [PMID: 35874523 PMCID: PMC9304560 DOI: 10.3389/fphys.2022.906272] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) has been recognized as the greatest single unmet need in cardiovascular medicine. Indeed, the morbi-mortality of HFpEF is high and as the population ages and the comorbidities increase, so considerably does the prevalence of HFpEF. However, HFpEF pathophysiology is still poorly understood and therapeutic targets are missing. An unifying, but untested, theory of the pathophysiology of HFpEF, proposed in 2013, suggests that cardiovascular risk factors lead to a systemic inflammation, which triggers endothelial cells (EC) and coronary microvascular dysfunction. This cardiac small vessel disease is proposed to be responsible for cardiac wall stiffening and diastolic dysfunction. This paradigm is based on the fact that microvascular dysfunction is highly prevalent in HFpEF patients. More specifically, HFpEF patients have been shown to have decreased cardiac microvascular density, systemic endothelial dysfunction and a lower mean coronary flow reserve. Importantly, impaired coronary microvascular function has been associated with the severity of HF. This review discusses evidence supporting the causal role of endothelial dysfunction in the pathophysiology of HFpEF in human and experimental models.
Collapse
|
9
|
Roh J, Hill JA, Singh A, Valero-Muñoz M, Sam F. Heart Failure With Preserved Ejection Fraction: Heterogeneous Syndrome, Diverse Preclinical Models. Circ Res 2022; 130:1906-1925. [PMID: 35679364 PMCID: PMC10035274 DOI: 10.1161/circresaha.122.320257] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents one of the greatest challenges facing cardiovascular medicine today. Despite being the most common form of heart failure worldwide, there has been limited success in developing therapeutics for this syndrome. This is largely due to our incomplete understanding of the biology driving its systemic pathophysiology and the heterogeneity of clinical phenotypes, which are increasingly being recognized as distinct HFpEF phenogroups. Development of efficacious therapeutics fundamentally relies on robust preclinical models that not only faithfully recapitulate key features of the clinical syndrome but also enable rigorous investigation of putative mechanisms of disease in the context of clinically relevant phenotypes. In this review, we propose a preclinical research strategy that is conceptually grounded in model diversification and aims to better align with our evolving understanding of the heterogeneity of clinical HFpEF. Although heterogeneity is often viewed as a major obstacle in preclinical HFpEF research, we challenge this notion and argue that embracing it may be the key to demystifying its pathobiology. Here, we first provide an overarching guideline for developing HFpEF models through a stepwise approach of comprehensive cardiac and extra-cardiac phenotyping. We then present an overview of currently available models, focused on the 3 leading phenogroups, which are primarily based on aging, cardiometabolic stress, and chronic hypertension. We discuss how well these models reflect their clinically relevant phenogroup and highlight some of the more recent mechanistic insights they are providing into the complex pathophysiology underlying HFpEF.
Collapse
Affiliation(s)
- Jason Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (J.R., A.S.)
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology) (J.A.H.), University of Texas Southwestern Medical Center, Dallas
- Department of Molecular Biology (J.A.H.), University of Texas Southwestern Medical Center, Dallas
| | - Abhilasha Singh
- Cardiovascular Research Center, Massachusetts General Hospital, Boston (J.R., A.S.)
| | - María Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (M.V.-M., F.S.)
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, MA (M.V.-M., F.S.)
| |
Collapse
|
10
|
Ivabradine Ameliorates Cardiac Function in Heart Failure with Preserved and Reduced Ejection Fraction via Upregulation of miR-133a. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1257283. [PMID: 34630844 PMCID: PMC8494584 DOI: 10.1155/2021/1257283] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/04/2021] [Indexed: 11/21/2022]
Abstract
Heart failure (HF) is a clinical syndrome caused by impairment of ventricular filling, ejection of blood, or both and is categorized as HF with reduced ejection fraction (HFrEF) or HF with preserved ejection fraction (HFpEF) based on left ventricular function. Cardiac fibrosis contributes to left ventricular dysfunction and leads to the development of HF. Ivabradine, an If current selective specific inhibitor, has been shown to improve the prognosis of patients with HF. However, the effects of ivabradine on cardiac function and fibrosis in HFpEF and HFrEF and the underlying mechanism remain unclear. In the present study, we utilized mouse models to mimic HFpEF and HFrEF and evaluated the therapeutic effects of ivabradine. By treating mice with different doses (10 mg/kg/d and 20 mg/kg/d) of ivabradine for 4 or 8 weeks, we found that a high dose of ivabradine improved cardiac diastolic function in HFpEF mice and ameliorated cardiac diastolic and systolic function and ventricular tachycardia incidence in HFrEF mice. Moreover, ivabradine significantly reduced the activation of cardiac fibroblasts and myocardial fibrosis in mice. Mechanistically, microRNA-133a, which was upregulated by ivabradine, targeted connective tissue growth factor and collagen 1 in cardiac fibroblasts and might contribute to the protective role of ivabradine. Together, our work utilized mouse models to study HFpEF and HFrEF, demonstrated the protective role of ivabradine in HFpEF and HFrEF, and elucidated the potential underlying mechanism, which provides an effective strategy for related diseases.
Collapse
|
11
|
Withaar C, Lam CSP, Schiattarella GG, de Boer RA, Meems LMG. Heart failure with preserved ejection fraction in humans and mice: embracing clinical complexity in mouse models. Eur Heart J 2021; 42:4420-4430. [PMID: 34414416 PMCID: PMC8599003 DOI: 10.1093/eurheartj/ehab389] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/15/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is a multifactorial disease accounting for a large and increasing proportion of all clinical HF presentations. As a clinical syndrome, HFpEF is characterized by typical signs and symptoms of HF, a distinct cardiac phenotype and raised natriuretic peptides. Non-cardiac comorbidities frequently co-exist and contribute to the pathophysiology of HFpEF. To date, no therapy has proven to improve outcomes in HFpEF, with drug development hampered, at least partly, by lack of consensus on appropriate standards for pre-clinical HFpEF models. Recently, two clinical algorithms (HFA-PEFF and H2FPEF scores) have been developed to improve and standardize the diagnosis of HFpEF. In this review, we evaluate the translational utility of HFpEF mouse models in the context of these HFpEF scores. We systematically recorded evidence of symptoms and signs of HF or clinical HFpEF features and included several cardiac and extra-cardiac parameters as well as age and sex for each HFpEF mouse model. We found that most of the pre-clinical HFpEF models do not meet the HFpEF clinical criteria, although some multifactorial models resemble human HFpEF to a reasonable extent. We therefore conclude that to optimize the translational value of mouse models to human HFpEF, a novel approach for the development of pre-clinical HFpEF models is needed, taking into account the complex HFpEF pathophysiology in humans.
Collapse
Affiliation(s)
- Coenraad Withaar
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Carolyn S P Lam
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands.,National University Heart Centre, Singapore and Duke-National University of Singapore
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Department of Cardiology, Center for Cardiovascular Research (CCR), Charité - Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.,Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy.,Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - Laura M G Meems
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| |
Collapse
|
12
|
Fusco-Allison G, Li DK, Hunter B, Jackson D, Bannon PG, Lal S, O'Sullivan JF. Optimizing the discovery and assessment of therapeutic targets in heart failure with preserved ejection fraction. ESC Heart Fail 2021; 8:3643-3655. [PMID: 34342166 PMCID: PMC8497375 DOI: 10.1002/ehf2.13504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 06/02/2021] [Accepted: 06/21/2021] [Indexed: 01/09/2023] Open
Abstract
There is an urgent need for models that faithfully replicate heart failure with preserved ejection fraction (HFpEF), now recognized as the most common form of heart failure in the world. In vitro approaches have several shortcomings, most notably the immature nature of stem cell‐derived human cardiomyocytes [induced pluripotent stem cells (iPSC)] and the relatively short lifespan of primary cardiomyocytes. Three‐dimensional ‘organoids’ incorporating mature iPSCs with other cell types such as endothelial cells and fibroblasts are a significant advance, but lack the complexity of true myocardium. Animal models can replicate many features of human HFpEF, and rodent models are the most common, and recent attempts to incorporate haemodynamic, metabolic, and ageing contributions are encouraging. Differences relating to species, physiology, heart rate, and heart size are major limitations for rodent models. Porcine models mitigate many of these shortcomings and approximate human physiology more closely, but cost and time considerations limit their potential for widespread use. Ex vivo analysis of failing hearts from animal models offer intriguing possibilities regarding cardiac substrate utilisation, but are ultimately subject to the same constrains as the animal models from which the hearts are obtained. Ex vivo approaches using human myocardial biopsies can uncover new insights into pathobiology leveraging myocardial energetics, substrate turnover, molecular changes, and systolic/diastolic function. In collaboration with a skilled cardiothoracic surgeon, left ventricular endomyocardial biopsies can be obtained at the time of valvular surgery in HFpEF patients. Critically, these tissues maintain their disease phenotype, preserving inter‐relationship of myocardial cells and extracellular matrix. This review highlights a novel approach, where ultra‐thin myocardial tissue slices from human HFpEF hearts can be used to assess changes in myocardial structure and function. We discuss current approaches to modelling HFpEF, describe in detail the novel tissue slice model, expand on exciting opportunities this model provides, and outline ways to improve this model further.
Collapse
Affiliation(s)
- Gabrielle Fusco-Allison
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Desmond K Li
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Benjamin Hunter
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Dan Jackson
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Paul G Bannon
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Discipline of Surgery, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Sean Lal
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - John F O'Sullivan
- Precision Cardiovascular Laboratory, The University of Sydney, Sydney, New South Wales, Australia.,Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia.,Heart Research Institute, Newtown, Sydney, New South Wales, Australia.,Central Clinical School, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia.,Faculty of Medicine, TU Dresden, Dresden, Germany
| |
Collapse
|
13
|
Travers JG, Wennersten SA, Peña B, Bagchi RA, Smith HE, Hirsch RA, Vanderlinden LA, Lin YH, Dobrinskikh E, Demos-Davies KM, Cavasin MA, Mestroni L, Steinkühler C, Lin CY, Houser SR, Woulfe KC, Lam MPY, McKinsey TA. HDAC Inhibition Reverses Preexisting Diastolic Dysfunction and Blocks Covert Extracellular Matrix Remodeling. Circulation 2021; 143:1874-1890. [PMID: 33682427 DOI: 10.1161/circulationaha.120.046462] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Diastolic dysfunction (DD) is associated with the development of heart failure and contributes to the pathogenesis of other cardiac maladies, including atrial fibrillation. Inhibition of histone deacetylases (HDACs) has been shown to prevent DD by enhancing myofibril relaxation. We addressed the therapeutic potential of HDAC inhibition in a model of established DD with preserved ejection fraction. METHODS Four weeks after uninephrectomy and implantation with deoxycorticosterone acetate pellets, when DD was clearly evident, 1 cohort of mice was administered the clinical-stage HDAC inhibitor ITF2357/Givinostat. Echocardiography, blood pressure measurements, and end point invasive hemodynamic analyses were performed. Myofibril mechanics and intact cardiomyocyte relaxation were assessed ex vivo. Cardiac fibrosis was evaluated by picrosirius red staining and second harmonic generation microscopy of left ventricle (LV) sections, RNA sequencing of LV mRNA, mass spectrometry-based evaluation of decellularized LV biopsies, and atomic force microscopy determination of LV stiffness. Mechanistic studies were performed with primary rat and human cardiac fibroblasts. RESULTS HDAC inhibition normalized DD without lowering blood pressure in this model of systemic hypertension. In contrast to previous models, myofibril relaxation was unimpaired in uninephrectomy/deoxycorticosterone acetate mice. Furthermore, cardiac fibrosis was not evident in any mouse cohort on the basis of picrosirius red staining or second harmonic generation microscopy. However, mass spectrometry revealed induction in the expression of >100 extracellular matrix proteins in LVs of uninephrectomy/deoxycorticosterone acetate mice, which correlated with profound tissue stiffening based on atomic force microscopy. ITF2357/Givinostat treatment blocked extracellular matrix expansion and LV stiffening. The HDAC inhibitor was subsequently shown to suppress cardiac fibroblast activation, at least in part, by blunting recruitment of the profibrotic chromatin reader protein BRD4 (bromodomain-containing protein 4) to key gene regulatory elements. CONCLUSIONS These findings demonstrate the potential of HDAC inhibition as a therapeutic intervention to reverse existing DD and establish blockade of extracellular matrix remodeling as a second mechanism by which HDAC inhibitors improve ventricular filling. Our data reveal the existence of pathophysiologically relevant covert or hidden cardiac fibrosis that is below the limit of detection of histochemical stains such as picrosirius red, highlighting the need to evaluate fibrosis of the heart using diverse methodologies.
Collapse
Affiliation(s)
- Joshua G Travers
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Sara A Wennersten
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Brisa Peña
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Rushita A Bagchi
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Harrison E Smith
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (H.E.S., R.A.H., C.Y.L.).,Department of Biostatistics and Informatics (H.E.S., L.A.V.), Colorado School of Public Health, Aurora
| | - Rachel A Hirsch
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (H.E.S., R.A.H., C.Y.L.)
| | - Lauren A Vanderlinden
- Department of Biostatistics and Informatics (H.E.S., L.A.V.), Colorado School of Public Health, Aurora
| | - Ying-Hsi Lin
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Evgenia Dobrinskikh
- Department of Medicine, Division of Pulmonary Sciences & Critical Care (E.D.), University of Colorado Anschutz Medical Campus, Aurora
| | - Kimberly M Demos-Davies
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Maria A Cavasin
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Luisa Mestroni
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | | | - Charles Y Lin
- Molecular and Human Genetics, Baylor College of Medicine, Houston, TX (H.E.S., R.A.H., C.Y.L.).,now with Kronos Bio, Cambridge, MA (C.Y.L.)
| | - Steven R Houser
- Cardiovascular Research Center (S.R.H.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Kathleen C Woulfe
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Maggie P Y Lam
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., K.M.D.-D., M.A.C., L.M., K.C.W., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora.,Consortium for Fibrosis Research & Translation (J.G.T., S.A.W., B.P., R.A.B., Y.-H.L., M.A.C., M.P.Y.L., T.A.M.), University of Colorado Anschutz Medical Campus, Aurora
| |
Collapse
|
14
|
Abstract
Diabetic heart disease is a growing and important public health risk. Apart from the risk of coronary artery disease or hypertension, diabetes mellitus (DM) is a well-known risk factor for heart failure in the form of diabetic cardiomyopathy (DiaCM). Currently, DiaCM is defined as myocardial dysfunction in patients with DM in the absence of coronary artery disease and hypertension. The underlying pathomechanism of DiaCM is partially understood, but accumulating evidence suggests that metabolic derangements, oxidative stress, increased myocardial fibrosis and hypertrophy, inflammation, enhanced apoptosis, impaired intracellular calcium handling, activation of the renin-angiotensin-aldosterone system, mitochondrial dysfunction, and dysregulation of microRNAs, among other factors, are involved. Numerous animal models have been used to investigate the pathomechanisms of DiaCM. Despite some limitations, animal models for DiaCM have greatly advanced our understanding of pathomechanisms and have helped in the development of successful disease management strategies. In this review, we summarize the current pathomechanisms of DiaCM and provide animal models for DiaCM according to its pathomechanisms, which may contribute to broadening our understanding of the underlying mechanisms and facilitating the identification of possible new therapeutic targets.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Corresponding authors: Wang-Soo Lee https://orcid.org/0000-0002-8264-0866 Division of Cardiology, Department of Internal Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul 06973, Korea E-mail:
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
- Corresponding authors: Wang-Soo Lee https://orcid.org/0000-0002-8264-0866 Division of Cardiology, Department of Internal Medicine, Chung-Ang University Hospital, 102 Heukseok-ro, Dongjak-gu, Seoul 06973, Korea E-mail:
| |
Collapse
|
15
|
Methawasin M, Strom J, Borkowski T, Hourani Z, Runyan R, Smith JE, Granzier H. Phosphodiesterase 9a Inhibition in Mouse Models of Diastolic Dysfunction. Circ Heart Fail 2020; 13:e006609. [PMID: 32418479 DOI: 10.1161/circheartfailure.119.006609] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Low myocardial cGMP-PKG (cyclic guanosine monophosphate-protein kinase G) activity has been associated with increased cardiomyocyte diastolic stiffness in heart failure with preserved ejection fraction. Cyclic guanosine monophosphate is mainly hydrolyzed by PDE (phosphodiesterases) 5a and 9a. Importantly, PDE9a expression has been reported to be upregulated in human heart failure with preserved ejection fraction myocardium and chronic administration of a PDE9a inhibitor reverses preestablished cardiac hypertrophy and systolic dysfunction in mice subjected to transverse aortic constriction (TAC). We hypothesized that inhibiting PDE9a activity ameliorates diastolic dysfunction. METHODS To examine the effect of chronic PDE9a inhibition, 2 diastolic dysfunction mouse models were studied: (1) TAC-deoxycorticosterone acetate and (2) Leprdb/db. PDE9a inhibitor (5 and 8 mg/kg per day) was administered to the mice via subcutaneously implanted osmotic minipumps for 28 days. The effect of acute PDE9a inhibition was investigated in intact cardiomyocytes isolated from TAC-deoxycorticosterone acetate mice. Atrial natriuretic peptide together with PDE9a inhibitor were administered to the isolated intact cardiomyocytes through the cell perfusate. RESULTS For acute inhibition, no cellular stiffness reduction was found, whereas chronic PDE9a inhibition resulted in reduced left ventricular chamber stiffness in TAC-deoxycorticosterone acetate, but not in Leprdb/db mice. Passive cardiomyocyte stiffness was reduced by chronic PDE9a inhibition, with no differences in myocardial fibrosis or cardiac morphometry. PDE9a inhibition increased the ventricular-arterial coupling ratio, reflecting impaired systolic function. CONCLUSIONS Chronic PDE9a inhibition lowers left ventricular chamber stiffness in TAC-deoxycorticosterone acetate mice. However, the usefulness of PDE9a inhibition to treat high-diastolic stiffness may be limited as the required PDE9a inhibitor dose also impairs systolic function, observed as a decline in ventricular-arterial coordination, in this model.
Collapse
Affiliation(s)
- Mei Methawasin
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ
| | - Joshua Strom
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ
| | - Tomasz Borkowski
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ
| | - Ray Runyan
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ
| | - John E Smith
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ
| | - Henk Granzier
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ
| |
Collapse
|
16
|
Bretagne M, Lebrun-Vignes B, Pariente A, Shaffer CM, Malouf GG, Dureau P, Potey C, Funck-Brentano C, Roden DM, Moslehi JJ, Salem JE. Heart failure and atrial tachyarrhythmia on abiraterone: A pharmacovigilance study. Arch Cardiovasc Dis 2020; 113:9-21. [DOI: 10.1016/j.acvd.2019.09.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 01/07/2023]
|
17
|
Heart failure with preserved ejection fraction: present status and future directions. Exp Mol Med 2019; 51:1-9. [PMID: 31857581 PMCID: PMC6923411 DOI: 10.1038/s12276-019-0323-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/01/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022] Open
Abstract
The clinical importance of heart failure with preserved ejection fraction (HFpEF) has recently become apparent. HFpEF refers to heart failure (HF) symptoms with normal or near-normal cardiac function on echocardiography. Common clinical features of HFpEF include diastolic dysfunction, reduced compliance, and ventricular hypokinesia. HFpEF differs from the better-known HF with reduced ejection fraction (HFrEF). Despite having a "preserved ejection fraction," patients with HFpEF have symptoms such as shortness of breath, excessive tiredness, and limited exercise capability. Furthermore, the mortality rate and cumulative survival rate are as severe in HFpEF as they are in HFrEF. While beta-blockers and renin-angiotensin-aldosterone system modulators can improve the survival rate in HFrEF, no known therapeutic agents show similar effectiveness in HFpEF. Researchers have examined molecular events in the development of HFpEF using small and middle-sized animal models. This review discusses HFpEF with regard to etiology and clinical features and introduces the use of mouse and other animal models of human HFpEF.
Collapse
|
18
|
Abstract
STUDY OBJECTIVE Computed tomography (CT) is an important imaging modality in diagnosing a variety of disorders. Although systolic heart failure is a well-known risk factor for postcontrast acute kidney injury (PC-AKI), few studies have evaluated the association between diastolic dysfunction and PC-AKI. Therefore, the aim of our study was to investigate whether PC-AKI occurs more likely in patients with diastolic dysfuction. METHODS This retrospective study was conducted by collecting the data of patients who visited an emergency medical center between January 2008 and December 2014. Patients who underwent contrast-enhanced CT (CECT) in the emergency department and had undergone echocardiography within 1 month of CECT were included. We defined PC-AKI as an elevation in the serum creatinine level of ≥0.5 mg/dL or ≥25% within 72 hours after CECT. RESULTS We included 327 patients, aged 18 years and older, who had a CECT scan and underwent an echocardiography within 1 month of the CECT scan at our institute over 20 years. The mean value of estimated glomerular filtration rate and E/E (early left ventricular filling velocity to early diastolic mitral annular velocity ratio) was 51.55 ± 7.66 mL·min·1.73 m and 11.56 ± 5.33, respectively. A total of 32 patients (9.79%) developed PC-AKI. The prevalence of diabetes mellitus and chronic kidney disease was significantly higher in the PC-AKI group than in the non-PC-AKI group. Echocardiographic findings revealed that E/E was significantly increased in patients with PC-AKI. The logistic regression analysis showed that a higher E/E value (odds ratio [OR] 5.39, 95% confidence interval [CI] 1.51-25.23, P = .015) was a significant risk factor for PC-AKI. CONCLUSION This study demonstrated that, among the echocardiographic variables, E/E was an independent predictor of PC- AKI. This, in turn, suggests that diastolic dysfunction may be a useful parameter in PC-AKI risk stratification.
Collapse
Affiliation(s)
| | | | - Hyuk-Hoon Kim
- Department of Emergency Medicine, Ajou University School of Medicine, 16499, Suwon, Republic of Korea
| |
Collapse
|
19
|
Inflammation and fibrosis in murine models of heart failure. Basic Res Cardiol 2019; 114:19. [PMID: 30887214 DOI: 10.1007/s00395-019-0722-5] [Citation(s) in RCA: 238] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 02/12/2019] [Indexed: 02/07/2023]
Abstract
Heart failure is a consequence of various cardiovascular diseases and associated with poor prognosis. Despite progress in the treatment of heart failure in the past decades, prevalence and hospitalisation rates are still increasing. Heart failure is typically associated with cardiac remodelling. Here, inflammation and fibrosis are thought to play crucial roles. During cardiac inflammation, immune cells invade the cardiac tissue and modulate tissue-damaging responses. Cardiac fibrosis, however, is characterised by an increased amount and a disrupted composition of extracellular matrix proteins. As evidence exists that cardiac inflammation and fibrosis are potentially reversible in experimental and clinical set ups, they are interesting targets for innovative heart failure treatments. In this context, animal models are important as they mimic clinical conditions of heart failure patients. The advantages of mice in this respect are short generation times and genetic modifications. As numerous murine models of heart failure exist, the selection of a proper disease model for a distinct research question is demanding. To facilitate this selection, this review aims to provide an overview about the current understanding of the pathogenesis of cardiac inflammation and fibrosis in six frequently used murine models of heart failure. Hence, it compares the models of myocardial infarction with or without reperfusion, transverse aortic constriction, chronic subjection to angiotensin II or deoxycorticosterone acetate, and coxsackievirus B3-induced viral myocarditis in this context. It furthermore provides information about the clinical relevance and the limitations of each model, and, if applicable, about the recent advancements in their methodological proceedings.
Collapse
|
20
|
Slater RE, Strom JG, Methawasin M, Liss M, Gotthardt M, Sweitzer N, Granzier HL. Metformin improves diastolic function in an HFpEF-like mouse model by increasing titin compliance. J Gen Physiol 2018; 151:42-52. [PMID: 30567709 PMCID: PMC6314384 DOI: 10.1085/jgp.201812259] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a syndrome characterized by increased diastolic stiffness, for which effective therapies are lacking. Slater et al. show that metformin lowers titin-based passive stiffness in an HFpEF mouse model and may therefore be of therapeutic benefit. Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome characterized by a preserved ejection fraction but increased diastolic stiffness and abnormalities of filling. Although the prevalence of HFpEF is high and continues to rise, no effective therapies exist; however, the diabetic drug metformin has been associated with improved diastolic function in diabetic patients. Here we determine the therapeutic potential of metformin for improving diastolic function in a mouse model with HFpEF-like symptoms. We combine transverse aortic constriction (TAC) surgery with deoxycorticosterone acetate (DOCA) supplementation to obtain a mouse model with increased diastolic stiffness and exercise intolerance. Echocardiography and pressure–volume analysis reveal that providing metformin to TAC/DOCA mice improves diastolic function in the left ventricular (LV) chamber. Muscle mechanics show that metformin lowers passive stiffness of the LV wall muscle. Concomitant with this improvement in diastolic function, metformin-treated TAC/DOCA mice also demonstrate preserved exercise capacity. No metformin effects are seen in sham operated mice. Extraction experiments on skinned ventricular muscle strips show that the metformin-induced reduction of passive stiffness in TAC/DOCA mice is due to an increase in titin compliance. Using phospho-site-specific antibodies, we assay the phosphorylation of titin’s PEVK and N2B spring elements. Metformin-treated mice have unaltered PEVK phosphorylation but increased phosphorylation of PKA sites in the N2B element, a change which has previously been shown to lower titin’s stiffness. Consistent with this result, experiments with a mouse model deficient in the N2B element reveal that the beneficial effect of metformin on LV chamber and muscle stiffness requires the presence of the N2B element. We conclude that metformin offers therapeutic benefit during HFpEF by lowering titin-based passive stiffness.
Collapse
Affiliation(s)
- Rebecca E Slater
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Joshua G Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ
| | - Martin Liss
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.,German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Michael Gotthardt
- Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.,German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Nancy Sweitzer
- Sarver Heart Center, College of Medicine, University of Arizona, Tucson, AZ
| | - Henk L Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ .,Sarver Heart Center, College of Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
21
|
Barandiarán Aizpurua A, Schroen B, van Bilsen M, van Empel V. Targeted HFpEF therapy based on matchmaking of human and animal models. Am J Physiol Heart Circ Physiol 2018; 315:H1670-H1683. [PMID: 30239232 DOI: 10.1152/ajpheart.00024.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The diversity in clinical phenotypes and poor understanding of the underlying pathophysiology of heart failure with preserved ejection fraction (HFpEF) is the main reason why no effective treatments have been found yet. Targeted, instead of one size fits all, treatment seems the only promising approach for treating HFpEF. To be able to design a targeted, phenotype-specific HFpEF treatment, the matrix relating clinical phenotypes and underlying pathophysiological mechanisms has to be clarified. This review discusses the opportunities for additional evaluation of the underlying pathophysiological processes, e.g., to evaluate biological phenotypes on top of clinical routine, to guide us toward a phenotype-specific HFpEF treatment. Moreover, a translational approach with matchmaking of animal models to biological HFpEF phenotypes will be a valuable step to test the effectiveness of novel, targeted interventions in HFpEF. Listen to this article's corresponding podcast at https://ajpheart.podbean.com/e/personalized-medicine-in-hfpef/ .
Collapse
Affiliation(s)
- Arantxa Barandiarán Aizpurua
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Blanche Schroen
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Marc van Bilsen
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Department of Physiology, Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases, Maastricht University , Maastricht , The Netherlands
| | - Vanessa van Empel
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre , Maastricht , The Netherlands
| |
Collapse
|
22
|
MicroRNA-101a suppresses fibrotic programming in isolated cardiac fibroblasts and in vivo fibrosis following trans-aortic constriction. J Mol Cell Cardiol 2018; 121:266-276. [PMID: 30053527 DOI: 10.1016/j.yjmcc.2018.07.251] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/19/2018] [Accepted: 07/23/2018] [Indexed: 01/17/2023]
Abstract
AIMS MiR-101a is reported to reduce post-infarction myocardial fibrosis through targeting c-FOS and TGFbr1. However the actions of miR-101a within the TGF-β signaling pathway are largely unknown. We demonstrate the mechanisms underlying mutual inhibition between miR-101a and TGF-β signaling and explore the therapeutic potential of miR-101a in suppressing pressure overload-induced cardiac fibrosis. METHODS AND RESULTS The effects of miR-101a on fibroblast proliferation, myofibroblast transdifferentiation, collagen synthesis, apoptosis, and autophagy were assessed in isolated rat cardiac fibroblasts (cFB). MiR-101a effects upon cFB TGF-β signaling were assessed by qPCR, Western blotting, 3'UTR luciferase reporter assay and promoter activity assessments. Rats subjected to transverse aortic constriction (TAC) were treated with PBS or 1 mg/kg of a miR-101a mimic i.v. at 1, 3, and 7-day post-surgery. Left ventricular (LV) function (echocardiography; LV catheterization) and LV fibrosis (picro-sirius red staining, qPCR and WB) were assessed at 2-day and 2 & 10-week post-surgery. MiR-101a inhibited cFB proliferation by inducing cell apoptosis rather than through cell cycle inhibition; and reduced collagen synthesis by downregulation of collagen gene expression and increased autophagy. MiR-101a inhibited TGF-β signaling pathways by directly targeting TGFbr1, reducing Smad3 phosphorylation and inhibiting Tab3 promoter activity. Conversely TGF-β inhibited promoter activity for both miR-101a and b. In vivo endogenous miR-101a expression was downregulated 2-day post-TAC returning to baseline by 14-day. MiR-101a mimic treatment inhibited myocardial TGF-β signaling and collagen gene up-regulation at 2-day post-TAC. MiR-101a reduced fibrosis, improved +dP/dt and lowered end diastolic pressure (EDP) at 2-week post-TAC. Treatment also attenuated adverse LV remodeling and preserved cardiac ejection fraction at 10-week post-TAC. CONCLUSIONS MiR-101a and TGF-β are mutually inhibitory and co-direct the activation, proliferation, and collagen synthesis of cFBs. The predominance of TGF-β signaling over reduced miR-101a expression early post-TAC is associated with cardiac fibrosis and dysfunction. Treatment with miR-101a, introduced early after imposition of abnormal pressure loading, inhibits TGF-β signaling, reduces cardiac fibrosis and preserves LV function.
Collapse
|
23
|
Marques FZ, Chu PY, Ziemann M, Kaspi A, Kiriazis H, Du XJ, El-Osta A, Kaye DM. Age-Related Differential Structural and Transcriptomic Responses in the Hypertensive Heart. Front Physiol 2018; 9:817. [PMID: 30038575 PMCID: PMC6046461 DOI: 10.3389/fphys.2018.00817] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/11/2018] [Indexed: 01/20/2023] Open
Abstract
While aging is a critical risk factor for heart failure, it remains uncertain whether the aging heart responds differentially to a hypertensive stimuli. Here we investigated phenotypic and transcriptomic differences between the young and aging heart using a mineralocorticoid-excess model of hypertension. Ten-week (“young”) and 36-week (“aging”) mice underwent a unilateral uninephrectomy with deoxycorticosterone acetate (DOCA) pellet implantation (n = 6–8/group) and were followed for 6 weeks. Cardiac structure and function, blood pressure (BP) and the cardiac transcriptome were subsequently examined. Young and aging DOCA mice had high BP, increased cardiac mass, cardiac hypertrophy, and fibrosis. Left ventricular end-diastolic pressure increased in aging DOCA-treated mice in contrast to young DOCA mice. Interstitial and perivascular fibrosis occurred in response to DOCA, but perivascular fibrosis was greater in aging mice. Transcriptomic analysis showed that young mice had features of higher oxidative stress, likely due to activation of the respiratory electron transport chain. In contrast, aging mice showed up-regulation of collagen formation in association with activation of innate immunity together with markers of inflammation including cytokine and platelet signaling. In comparison to younger mice, aging mice demonstrated different phenotypic and molecular responses to hypertensive stress. These findings have potential implications for the pathogenesis of age-related forms of cardiovascular disease, particularly heart failure.
Collapse
Affiliation(s)
- Francine Z Marques
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Pharmacology, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Po-Yin Chu
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Mark Ziemann
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Antony Kaspi
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia
| | - Helen Kiriazis
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Xiao-Jun Du
- Experimental Cardiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Assam El-Osta
- Epigenetics in Human Health and Disease, Department of Diabetes, Monash University, Melbourne, VIC, Australia.,Hong Kong Institute of Diabetes and Obesity, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong.,Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Heart Centre, Alfred Hospital, Melbourne, VIC, Australia
| |
Collapse
|
24
|
Valero-Muñoz M, Backman W, Sam F. Murine Models of Heart Failure with Preserved Ejection Fraction: a "Fishing Expedition". JACC Basic Transl Sci 2017; 2:770-789. [PMID: 29333506 PMCID: PMC5764178 DOI: 10.1016/j.jacbts.2017.07.013] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/25/2017] [Accepted: 07/27/2017] [Indexed: 12/28/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is characterized by signs and symptoms of HF in the presence of a normal left ventricular (LV) ejection fraction (EF). Despite accounting for up to 50% of all clinical presentations of HF, the mechanisms implicated in HFpEF are poorly understood, thus precluding effective therapy. The pathophysiological heterogeneity in the HFpEF phenotype also contributes to this disease and likely to the absence of evidence-based therapies. Limited access to human samples and imperfect animal models that completely recapitulate the human HFpEF phenotype have impeded our understanding of the mechanistic underpinnings that exist in this disease. Aging and comorbidities such as atrial fibrillation, hypertension, diabetes and obesity, pulmonary hypertension and renal dysfunction are highly associated with HFpEF. Yet, the relationship and contribution between them remains ill-defined. This review discusses some of the distinctive clinical features of HFpEF in association with these comorbidities and highlights the advantages and disadvantage of commonly used murine models, used to study the HFpEF phenotype.
Collapse
Affiliation(s)
- Maria Valero-Muñoz
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Warren Backman
- Evans Department of Internal Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
- Evans Department of Internal Medicine, Boston University School of Medicine, Boston, Massachusetts
- Cardiovascular Section, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
25
|
Slater RE, Strom JG, Granzier H. Effect of exercise on passive myocardial stiffness in mice with diastolic dysfunction. J Mol Cell Cardiol 2017; 108:24-33. [PMID: 28476659 DOI: 10.1016/j.yjmcc.2017.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 12/20/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex syndrome, characterized by increased diastolic stiffness and a preserved ejection fraction, with no effective treatment options. Here we studied the therapeutic potential of exercise for improving diastolic function in a mouse model with HFpEF-like symptoms, the TtnΔIAjxn mouse model. TtnΔIAjxn mice have increased diastolic stiffness and reduced exercise tolerance, mimicking aspects of HFpEF observed in patients. We investigated the effect of free-wheel running exercise on diastolic function. Mechanical studies on cardiac muscle strips from the LV free wall revealed that both TtnΔIAjxn and wildtype (WT) exercised mice had a reduction in passive stiffness, relative to sedentary controls. In both genotypes, this reduction is due to an increase in the compliance of titin whereas ECM-based stiffness was unaffected. Phosphorylation of titin's PEVK and N2B spring elements were assayed with phospho-site specific antibodies. Exercised mice had decreased PEVK phosphorylation and increased N2B phosphorylation both of which are predicted to contribute to the increased compliance of titin. Since exercise lowers the heart rate we examined whether reduction in heart rate per se can improve passive stiffness by administering the heart-rate-lowering drug ivabradine. Ivabradine lowered heart rate in our study but it did not affect passive tension, in neither WT nor TtnΔIAjxn mice. We conclude that exercise is beneficial for decreasing passive stiffness and that it involves beneficial alterations in titin phosphorylation.
Collapse
Affiliation(s)
- Rebecca E Slater
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States
| | - Joshua G Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, United States; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, United States.
| |
Collapse
|
26
|
Zakeri R, Moulay G, Chai Q, Ogut O, Hussain S, Takahama H, Lu T, Wang XL, Linke WA, Lee HC, Redfield MM. Left Atrial Remodeling and Atrioventricular Coupling in a Canine Model of Early Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2016; 9:CIRCHEARTFAILURE.115.003238. [PMID: 27758811 DOI: 10.1161/circheartfailure.115.003238] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/16/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND Left atrial (LA) compliance and contractility influence left ventricular stroke volume. We hypothesized that diminished LA compliance and contractile function occur early during the development of heart failure with preserved ejection fraction (HFpEF) and impair overall cardiac performance. METHODS AND RESULTS Cardiac magnetic resonance imaging, echocardiography, left ventricular and LA pressure-volume studies, and tissue analyses were performed in a model of early HFpEF (elderly dogs, renal wrap-induced hypertension, exogenous aldosterone; n=9) and young control dogs (sham surgery; n=13). Early HFpEF was associated with LA enlargement, cardiomyocyte hypertrophy, and enhanced LA contractile function (median active emptying fraction 16% [95% confidence interval, 13-24]% versus 12 [10-14]%, P=0.008; end-systolic pressure-volume relationship slope 2.4 [1.9-3.2]mm Hg/mL HFpEF versus 1.5 [1.2-2.2]mm Hg/mL controls, P=0.01). However, atrioventricular coupling was impaired and the curvilinear LA end-reservoir pressure-volume relationship was shifted upward/leftward in HFpEF (LA stiffness constant [βLA] 0.16 [0.11-0.18]mm Hg/mL versus 0.06 [0.04-0.10]mm Hg/mL controls; P=0.002), indicating reduced LA compliance. Impaired atrioventricular coupling and lower LA compliance correlated with lower left ventricular stroke volume. Total fibrosis and titin isoform composition were similar between groups; however, titin was hyperphosphorylated in HFpEF and correlated with βLA. LA microvascular reactivity was diminished in HFpEF versus controls. LA microvascular density tended to be lower in HFpEF and inversely correlated with βLA. CONCLUSIONS In early-stage hypertensive HFpEF, LA cardiomyocyte hypertrophy, titin hyperphosphorylation, and microvascular dysfunction occur in association with increased systolic and diastolic LA chamber stiffness, impaired atrioventricular coupling, and decreased left ventricular stroke volume. These data indicate that maladaptive LA remodeling occurs early during HFpEF development, supporting a concept of global myocardial remodeling.
Collapse
Affiliation(s)
- Rosita Zakeri
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L).
| | - Gilles Moulay
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Qiang Chai
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Ozgur Ogut
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Saad Hussain
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Hiroyuki Takahama
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Tong Lu
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Xiao-Li Wang
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Wolfgang A Linke
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Hon-Chi Lee
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| | - Margaret M Redfield
- From the Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (R.Z., G.M., Q.C., O.O., S.H., H.T., T.L., X.-L.W., H.-C.L., M.M.R.); and Department of Cardiovascular Physiology, Ruhr University Bochum, Germany (WA.L)
| |
Collapse
|
27
|
Methawasin M, Strom JG, Slater RE, Fernandez V, Saripalli C, Granzier H. Experimentally Increasing the Compliance of Titin Through RNA Binding Motif-20 (RBM20) Inhibition Improves Diastolic Function In a Mouse Model of Heart Failure With Preserved Ejection Fraction. Circulation 2016; 134:1085-1099. [PMID: 27630136 DOI: 10.1161/circulationaha.116.023003] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/05/2016] [Indexed: 01/05/2023]
Abstract
BACKGROUND Left ventricular (LV) stiffening contributes to heart failure with preserved ejection fraction (HFpEF), a syndrome with no effective treatment options. Increasing the compliance of titin in the heart has become possible recently through inhibition of the splicing factor RNA binding motif-20. Here, we investigated the effects of increasing the compliance of titin in mice with diastolic dysfunction. METHODS Mice in which the RNA recognition motif (RRM) of one of the RNA binding motif-20 alleles was floxed and that expressed the MerCreMer transgene under control of the αMHC promoter (referred to as cRbm20ΔRRM mice) were used. Mice underwent transverse aortic constriction (TAC) surgery and deoxycorticosterone acetate (DOCA) pellet implantation. RRM deletion in adult mice was triggered by injecting raloxifene (cRbm20ΔRRM-raloxifene), with dimethyl sulfoxide (DMSO)-injected mice (cRbm20ΔRRM-DMSO) as the control. Diastolic function was investigated with echocardiography and pressure-volume analysis; passive stiffness was studied in LV muscle strips and isolated cardiac myocytes before and after elimination of titin-based stiffness. Treadmill exercise performance was also studied. Titin isoform expression was evaluated with agarose gels. RESULTS cRbm20ΔRRM-raloxifene mice expressed large titins in the hearts, called supercompliant titin (N2BAsc), which, within 3 weeks after raloxifene injection, made up ≈45% of total titin. TAC/DOCA cRbm20ΔRRM-DMSO mice developed LV hypertrophy and a marked increase in LV chamber stiffness as shown by both pressure-volume analysis and echocardiography. LV chamber stiffness was normalized in TAC/DOCA cRbm20ΔRRM-raloxifene mice that expressed N2BAsc. Passive stiffness measurements on muscle strips isolated from the LV free wall revealed that extracellular matrix stiffness was equally increased in both groups of TAC/DOCA mice (cRbm20ΔRRM-DMSO and cRbm20ΔRRM-raloxifene). However, titin-based muscle stiffness was reduced in the mice that expressed N2BAsc (TAC/DOCAcRbm20ΔRRM-raloxifene). Exercise testing demonstrated significant improvement in exercise tolerance in TAC/DOCA mice that expressed N2BAsc. CONCLUSIONS Inhibition of the RNA binding motif-20-based titin splicing system upregulates compliant titins, which improves diastolic function and exercise tolerance in the TAC/DOCA model. Titin holds promise as a therapeutic target for heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Mei Methawasin
- From Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson
| | - Joshua G Strom
- From Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson
| | - Rebecca E Slater
- From Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson
| | - Vanessa Fernandez
- From Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson
| | - Chandra Saripalli
- From Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson
| | - Henk Granzier
- From Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson.
| |
Collapse
|
28
|
Zempo H, Suzuki JI, Ogawa M, Watanabe R, Fujiu K, Manabe I, Conway SJ, Taniyama Y, Morishita R, Hirata Y, Isobe M, Nagai R. Influence of periostin-positive cell-specific Klf5 deletion on aortic thickening in DOCA-salt hypertensive mice. Hypertens Res 2016; 39:764-768. [PMID: 27334059 DOI: 10.1038/hr.2016.65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 04/30/2016] [Accepted: 05/05/2016] [Indexed: 12/30/2022]
Abstract
Chronic hypertension causes vascular remodeling that is associated with an increase in periostin- (postn) positive cells, including fibroblasts and smooth muscle cells. Krüppel-like factor (KLF) 5, a transcription factor, is also observed in vascular remodeling; however, it is unknown what role KLF5 plays in postn-positive cells during vascular remodeling induced by deoxycorticosterone-acetate (DOCA) salt. We used postn-positive cell-specific Klf5-deficient mice (Klf5PostnKO: Klf5flox/flox; PostnCre/-) and wild-type mice (WT: Klf5flox/flox; Postn-/-). We implanted a DOCA pellet and provided drinking water containing 0.9% NaCl for 8 weeks. The DOCA-salt treatment induced hypertension in both genotypes, as observed by increases in systolic blood pressure. In WT animals, DOCA-salt treatment increased the aortic medial area compared with the non-treated controls. Similarly, Tgfb1 was overexpressed in the aortas of the DOCA-salt treated WT mice compared with the controls. Immunofluorescence staining revealed that fibroblast-specific protein 1 (FSP1)+-α smooth muscle actin (αSMA)+ myofibroblasts exist in the medial area of the WT aortas after DOCA-salt intervention. Importantly, these changes were not observed in the Klf5PostnKO animals. In conclusion, the results of this study suggest that the presence of KLF5 in postn-positive cells contributes to the pathogenesis of aortic thickening induced by DOCA-salt hypertension.
Collapse
Affiliation(s)
- Hirofumi Zempo
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jun-Ichi Suzuki
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Tokyo, Japan
| | - Masahito Ogawa
- Westmead Millennium Institute for Medical Research, Westmead, New South Wales, Australia
| | - Ryo Watanabe
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuhito Fujiu
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Ichiro Manabe
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Simon J Conway
- Developmental Biology and Neonatal Medicine Program, Herman B Wells Center for Pediatric Research, Indiana University of Medicine, Indianapolis, IN, USA
| | | | | | - Yasunobu Hirata
- Department of Advanced Clinical Science and Therapeutics, The University of Tokyo, Tokyo, Japan
| | - Mitsuaki Isobe
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke City, Tochigi, Japan
| |
Collapse
|
29
|
Conceição G, Heinonen I, Lourenço AP, Duncker DJ, Falcão-Pires I. Animal models of heart failure with preserved ejection fraction. Neth Heart J 2016; 24:275-86. [PMID: 26936157 PMCID: PMC4796054 DOI: 10.1007/s12471-016-0815-9] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) constitutes a clinical syndrome in which the diagnostic criteria of heart failure are not accompanied by gross disturbances of systolic function, as assessed by ejection fraction. In turn, under most circumstances, diastolic function is impaired. Although it now represents over 50 % of all patients with heart failure, the mechanisms of HFpEF remain understood, precluding effective therapy. Understanding the pathophysiology of HFpEF has been restricted by both limited access to human myocardial biopsies and by the lack of animal models that fully mimic human pathology. Animal models are valuable research tools to clarify subcellular and molecular mechanisms under conditions where the comorbidities and other confounding factors can be precisely controlled. Although most of the heart failure animal models currently available represent heart failure with reduced ejection fraction, several HFpEF animal models have been proposed. However, few of these fulfil all the features present in human disease. In this review we will provide an overview of the currently available models to study HFpEF from rodents to large animals as well as present advantages and disadvantages of these models.
Collapse
Affiliation(s)
- G Conceição
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - I Heinonen
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Turku PET Centre, University of Turku, Turku, Finland
| | - A P Lourenço
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Anesthesiology, Centro Hospitalar de São João, Porto, Portugal
| | - D J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - I Falcão-Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal.
| |
Collapse
|
30
|
Mohamed IA, Mraiche F. Targeting osteopontin, the silent partner of Na+/H+ exchanger isoform 1 in cardiac remodeling. J Cell Physiol 2015; 230:2006-18. [PMID: 25677682 DOI: 10.1002/jcp.24958] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/06/2015] [Indexed: 12/11/2022]
Abstract
Cardiac hypertrophy (CH), characterized by the enlargement of cardiomyocytes, fibrosis and apoptosis, contributes to cardiac remodeling, which if left unresolved results in heart failure. Understanding the signaling pathways underlying CH is necessary to identify potential therapeutic targets. The Na(+) /H(+) -exchanger isoform I (NHE1), a ubiquitously expressed glycoprotein and cardiac specific isoform, regulates intracellular pH. Recent studies have demonstrated that enhanced expression/activity of NHE1 contributes to cardiac remodeling and CH. Inhibition of NHE1 in both in vitro and in vivo models have suggested that inhibition of NHE1 protects against hypertrophy. However, clinical trials using NHE1 inhibitors have proven to be unsuccessful, suggesting that additional factors maybe contributing to cardiac remodeling. Recent studies have indicated that the upregulation of NHE1 is associated with enhanced levels of osteopontin (OPN) in the setting of CH. OPN has been demonstrated to be upregulated in left ventricular hypertrophy, dilated cardiomyopathy and in diabetic cardiomyopathy. The cellular interplay between OPN and NHE1 in the setting of CH remains unknown. This review focuses on the role of NHE1 and OPN in cardiac remodeling and emphasizes the signaling pathways implicating OPN in the NHE1-induced hypertrophic response.
Collapse
|
31
|
Abstract
Neurohormonal activation with attendant aldosteronism contributes to the clinical appearance of congestive heart failure (CHF). Aldosteronism is intrinsically coupled to Zn and Ca dyshomeostasis, in which consequent hypozincemia compromises Zn homeostasis and Zn-based antioxidant defenses that contribute to the CHF prooxidant phenotype. Ionized hypocalcemia leads to secondary hyperparathyroidism with parathyroid hormone-mediated Ca overloading of diverse cells, including cardiomyocytes. When mitochondrial Ca overload exceeds a threshold, myocyte necrosis follows. The reciprocal regulation involving cytosolic free [Zn]i as antioxidant and [Ca]i as prooxidant can be uncoupled in favor of Zn-based antioxidant defenses. Increased [Zn]i acts as a multifaceted antioxidant by: (1) inhibiting Ca entry through L-type channels and hence cardioprotectant from the Ca-driven mitochondriocentric signal-transducer effector pathway to nonischemic necrosis, (2) serving as catalytic regulator of Cu/Zn-superoxide dismutase, and (3) activating its cytosolic sensor, metal-responsive transcription factor that regulates the expression of relevant antioxidant defense genes. Albeit present in subnanomolar range, increased cytosolic free [Zn]i enhances antioxidant capacity that confers cardioprotection. It can be achieved exogenously by ZnSO4 supplementation or endogenously using a β3-receptor agonist (eg, nebivolol) that enhances NO generation to release inactive cytosolic Zn bound to metallothionein. By recognizing the pathophysiologic relevance of Zn dyshomeostasis in the prooxidant CHF phenotype and by exploiting the pharmacophysiologic potential of [Zn]i as antioxidant, vulnerable cardiomyocytes under assault from neurohormonal activation can be protected and the myocardium spared from adverse structural remodeling.
Collapse
|
32
|
Degen CV, Bishu K, Zakeri R, Ogut O, Redfield MM, Brozovich FV. The emperor's new clothes: PDE5 and the heart. PLoS One 2015; 10:e0118664. [PMID: 25747598 PMCID: PMC4351884 DOI: 10.1371/journal.pone.0118664] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 01/21/2015] [Indexed: 02/07/2023] Open
Abstract
Phosphodiesterase-5 (PDE5) is highly expressed in the pulmonary vasculature, but its expression in the myocardium is controversial. Cyclic guanosine monophosphate (cGMP) activates protein kinase G (PKG), which has been hypothesized to blunt cardiac hypertrophy and negative remodeling in heart failure. Although PDE5 has been suggested to play a significant role in the breakdown of cGMP in cardiomyocytes and hence PKG regulation in the myocardium, the RELAX trial, which tested effect of PDE5 inhibition on exercise capacity in patients with heart failure with preserved ejection fraction (HFpEF) failed to show a beneficial effect. These results highlight the controversy regarding the role and expression of PDE5 in the healthy and failing heart. This study used one- and two-dimensional electrophoresis and Western blotting to examine PDE5 expression in mouse (before and after trans-aortic constriction), dog (control and HFpEF) as well as human (healthy and failing) heart. We were unable to detect PDE5 in any cardiac tissue lysate, whereas PDE5 was present in the murine and bovine lung samples used as positive controls. These results indicate that if PDE5 is expressed in cardiac tissue, it is present in very low quantities, as PDE5 was not detected in either humans or any model of heart failure examined. Therefore in cardiac muscle, it is unlikely that PDE5 is involved the regulation of cGMP-PKG signaling, and hence PDE5 does not represent a suitable drug target for the treatment of cardiac hypertrophy. These results highlight the importance of rigorous investigation prior to clinical trial design.
Collapse
Affiliation(s)
- Chantal V. Degen
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Kalkidan Bishu
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Rosita Zakeri
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Ozgur Ogut
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Margaret M. Redfield
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
| | - Frank V. Brozovich
- Mayo Medical School, Department of Cardiovascular Diseases, Rochester, MN, 55905, United States of America
- * E-mail:
| |
Collapse
|
33
|
Catena C, Colussi G, Sechi LA. Treatment of Primary Aldosteronism and Organ Protection. Int J Endocrinol 2015; 2015:597247. [PMID: 26074961 PMCID: PMC4449945 DOI: 10.1155/2015/597247] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/31/2015] [Indexed: 12/25/2022] Open
Abstract
Primary aldosteronism is a frequent form of secondary hypertension that had long been considered relatively benign. Experimental and clinical evidence collected in the last two decades, however, has clearly demonstrated that this endocrine disorder is associated with excess cardiovascular and renal complications as compared to essential hypertension. These complications reflect the ability of inappropriate elevation of plasma aldosterone to cause tissue damage beyond that induced by high blood pressure itself, thereby setting the stage for major cardiovascular and renal disease. Because of the impact of elevated aldosterone on organ damage, goals of treatment in patients with primary aldosteronism should not be limited to normalization of blood pressure, and prevention or correction of organ complications is mandatory. Treatment with mineralocorticoid receptor antagonists or unilateral adrenalectomy is the respective options for treatment of idiopathic adrenal hyperplasia or aldosterone-producing adenoma. Last years have witnessed a rapid growth in knowledge concerning the effects of these treatments on cardiovascular and renal protection. This paper is an overview of the cardiovascular and renal complications that occur in patients with primary aldosteronism and a summary of the results that have been obtained in the long term on cardiovascular and renal outcomes with either medical or surgical treatment.
Collapse
Affiliation(s)
- Cristiana Catena
- Hypertension Unit, Internal Medicine, Department of Experimental and Clinical Medical Sciences, University of Udine, 33100 Udine, Italy
- *Cristiana Catena:
| | - GianLuca Colussi
- Hypertension Unit, Internal Medicine, Department of Experimental and Clinical Medical Sciences, University of Udine, 33100 Udine, Italy
| | - Leonardo A. Sechi
- Hypertension Unit, Internal Medicine, Department of Experimental and Clinical Medical Sciences, University of Udine, 33100 Udine, Italy
| |
Collapse
|
34
|
Acikel S, Akdemir R, Kilic H, Cagirci G, Dogan M, Yesilay AB, Yeter E. Diastolic dysfunction and contrast-induced nephropathy in patients undergoing coronary angiography. Herz 2014; 40 Suppl 3:254-9. [PMID: 25432103 DOI: 10.1007/s00059-014-4173-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 10/02/2014] [Accepted: 10/14/2014] [Indexed: 01/20/2023]
Abstract
OBJECTIVES It has been demonstrated that decreased left ventricular ejection fraction (LVEF) is associated with an increased risk of contrast-induced nephropathy (CIN). In this study, we aimed to assess whether there is a relationship between left ventricular (LV) diastolic dysfunction and renal function decline after coronary angiography (CAG). PATIENTS AND METHODS The study consisted of two groups: group I, patients with normal diastolic function; group II, patients with cardiac symptoms and abnormal diastolic function. Serum creatinine (Crea) and glomerular filtration rates (GFR) were measured before and after 48 h of CAG. RESULTS After the procedure, serum Crea values were higher in group II compared with group I (p = 0.051). Postprocedural 48-h GFR values determined by Cockcroft-Gault and Modification of Diet in Renal Disease (MDRD) equations were lower in group II compared with group I (p = 0.016 and p = 0.003, respectively). Delta (Δ) ΔCrea and ΔGFR determined by the Cockcroft-Gault and MDRD equations were statistically higher in group II than in group I (p = 0.005, p = 0.052, p = 0.030). The presence of higher age (p = 0.025), E/E' lateral ratio (p = 0.030), and left atrial volume index (p = 0.05) were independent predictors of worsening renal function. CONCLUSION The presence of diastolic dysfunction may play a role in determining the risk of CIN in patients with normal LVEF.
Collapse
Affiliation(s)
- S Acikel
- Department of Cardiology, Ministry of Health Dışkapı Yıldırım Beyazıt Research and Educational Hospital, 06110, Ankara, Turkey,
| | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Classical effects of mineralocorticoids include stimulation of Na(+) reabsorption and K(+) secretion in the kidney and other epithelia including colon and several glands. Moreover, mineralocorticoids enhance the excretion of Mg(2+) and renal tubular H(+) secretion. The renal salt retention following mineralocorticoid excess leads to extracellular volume expansion and hypertension. The increase of blood pressure following mineralocorticoid excess is, however, not only the result of volume expansion but may result from stiff endothelial cell syndrome impairing the release of vasodilating nitric oxide. Beyond that, mineralocorticoids are involved in the regulation of a wide variety of further functions, including cardiac fibrosis, platelet activation, neuronal function and survival, inflammation as well as vascular and tissue fibrosis and calcification. Those functions are briefly discussed in this short introduction to the special issue. Beyond that, further contributions of this special issue amplify on mineralocorticoid-induced sodium appetite and renal salt retention, the role of mineralocorticoids in the regulation of acid-base balance, the involvement of aldosterone and its receptors in major depression, the mineralocorticoid stimulation of inflammation and tissue fibrosis and the effect of aldosterone on osteoinductive signaling and vascular calcification. Clearly, still much is to be learned about the various ramifications of mineralocorticoid-sensitive physiology and pathophysiology.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
36
|
|
37
|
Nakagawa H, Oberwinkler H, Nikolaev VO, Gaßner B, Umbenhauer S, Wagner H, Saito Y, Baba HA, Frantz S, Kuhn M. Atrial Natriuretic Peptide Locally Counteracts the Deleterious Effects of Cardiomyocyte Mineralocorticoid Receptor Activation. Circ Heart Fail 2014; 7:814-21. [DOI: 10.1161/circheartfailure.113.000885] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background—
The endocrine balance between atrial natriuretic peptide (ANP) and the renin–angiotensin–aldosterone system is critical for the maintenance of arterial blood pressure and volume homeostasis. This study investigated whether a cardiac imbalance between ANP and aldosterone, toward increased mineralocorticoid receptor (MR) signaling, contributes to adverse left ventricular remodeling in response to pressure overload.
Methods and Results—
We used the MR-selective antagonist eplerenone to test the role of MRs in mediating pressure overload–induced dilatative cardiomyopathy of mice with abolished local, cardiac ANP activity. In response to 21 days of transverse aortic constriction, mice with cardiomyocyte-restricted inactivation (knockout) of the ANP receptor (guanylyl cyclase [GC]-A) or the downstream cGMP-dependent protein kinase I developed enhanced left ventricular hypertrophy and fibrosis together with contractile dysfunction. Treatment with eplerenone (100 mg/kg/d) attenuated left ventricular hypertrophy and fully prevented fibrosis, dilatation, and failure. Transverse aortic constriction induced the cardiac expression of profibrotic connective tissue growth factor and attenuated the expression of SERCA2a (sarcoplasmic reticulum Ca
2+
-ATPase) in knockout mice, but not in controls. These genotype-dependent molecular changes were similarly prevented by eplerenone. ANP attenuated the aldosterone-induced nuclear translocation of MRs via GC-A/cGMP-dependent protein kinase I in transfected HEK 293 (human embryonic kidney) cells. Coimmunoprecipitation and fluorescence resonance energy transfer experiments demonstrated that a population of MRs were membrane associated in close interaction with GC-A and cGMP-dependent protein kinase I and, moreover, that aldosterone caused a conformational change of this membrane MR/GC-A protein complex which was prevented by ANP.
Conclusions—
ANP counter-regulates cardiac MR activation in hypertensive heart disease. An imbalance in cardiac ANP/GC-A (inhibition) and aldosterone/MR signaling (augmentation) favors adverse cardiac remodeling in chronic pressure overload.
Collapse
Affiliation(s)
- Hitoshi Nakagawa
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Heike Oberwinkler
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Viacheslav O. Nikolaev
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Birgit Gaßner
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Sandra Umbenhauer
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Helga Wagner
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Yoshihiko Saito
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Hideo A. Baba
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Stefan Frantz
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| | - Michaela Kuhn
- From the Institute of Physiology (H.N., H.O., B.G., M.K.) and Comprehensive Heart Failure Center (H.N., S.F., M.K.), University Würzburg, Würzburg, Germany; Emmy Noether Group of the Deutsche Forschungsgemeinschaft, Department of Cardiology and Pneumology, University Göttingen, Göttingen, Germany (V.O.N.); Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany (S.U., H.W, S.F.); First Department of Internal Medicine, Nara Medical University, Kashihara, Japan (Y.S.); and
| |
Collapse
|
38
|
Tanaka K, Wilson RM, Essick EE, Duffen JL, Scherer PE, Ouchi N, Sam F. Effects of adiponectin on calcium-handling proteins in heart failure with preserved ejection fraction. Circ Heart Fail 2014; 7:976-85. [PMID: 25149095 DOI: 10.1161/circheartfailure.114.001279] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Despite the increasing prevalence of heart failure with preserved ejection fraction (HFpEF) in humans, there remains no therapeutic options for HFpEF. Adiponectin, an adipocyte-derived cytokine, exerts cardioprotective actions, and its deficiency is implicated in the development of hypertension and HF with reduced ejection fraction. Similarly, adiponectin deficiency in HFpEF exacerbates left ventricular hypertrophy, diastolic dysfunction, and HF. However, the therapeutic effects of adiponectin in HFpEF remain unknown. We sought to test the hypothesis that chronic adiponectin overexpression protects against the progression of HF in a murine model of HFpEF. METHODS AND RESULTS Adiponectin transgenic and wild-type mice underwent uninephrectomy, a continuous saline or d-aldosterone infusion and given 1.0% sodium chloride drinking water for 4 weeks. Aldosterone-infused wild-type mice developed HFpEF with hypertension, left ventricular hypertrophy, and diastolic dysfunction. Aldosterone infusion increased myocardial oxidative stress and decreased sarcoplasmic reticulum Ca(2+)-ATPase protein expression in HFpEF. Although total phospholamban protein expression was unchanged, there was a decreased expression of protein kinase A-dependent phospholamban phosphorylation at Ser16 and CaMKII (Ca(2+)/calmodulin-dependent protein kinase II)-dependent phospholamban phosphorylation at Thr17. Adiponectin overexpression in aldosterone-infused mice ameliorated left ventricular hypertrophy, diastolic dysfunction, lung congestion, and myocardial oxidative stress without affecting blood pressure and left ventricular EF. This improvement in diastolic dysfunction parameters in aldosterone-infused adiponectin transgenic mice was accompanied by the preserved protein expression of protein kinase A-dependent phosphorylation of phospholamban at Ser16. Adiponectin replacement prevented the progression of aldosterone-induced HFpEF, independent of blood pressure, by improving diastolic dysfunction and by modulating cardiac hypertrophy. CONCLUSIONS These findings suggest that adiponectin may have therapeutic effects in patients with HFpEF.
Collapse
Affiliation(s)
- Komei Tanaka
- From the Whitaker Cardiovascular Institute (K.T., R.M.W., E.E.E., J.L.D., N.O., F.S.) and Cardiovascular Section and Evans Department of Medicine (F.S.), Boston University School of Medicine, MA; and Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (P.E.S.)
| | - Richard M Wilson
- From the Whitaker Cardiovascular Institute (K.T., R.M.W., E.E.E., J.L.D., N.O., F.S.) and Cardiovascular Section and Evans Department of Medicine (F.S.), Boston University School of Medicine, MA; and Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (P.E.S.)
| | - Eric E Essick
- From the Whitaker Cardiovascular Institute (K.T., R.M.W., E.E.E., J.L.D., N.O., F.S.) and Cardiovascular Section and Evans Department of Medicine (F.S.), Boston University School of Medicine, MA; and Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (P.E.S.)
| | - Jennifer L Duffen
- From the Whitaker Cardiovascular Institute (K.T., R.M.W., E.E.E., J.L.D., N.O., F.S.) and Cardiovascular Section and Evans Department of Medicine (F.S.), Boston University School of Medicine, MA; and Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (P.E.S.)
| | - Philipp E Scherer
- From the Whitaker Cardiovascular Institute (K.T., R.M.W., E.E.E., J.L.D., N.O., F.S.) and Cardiovascular Section and Evans Department of Medicine (F.S.), Boston University School of Medicine, MA; and Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (P.E.S.)
| | - Noriyuki Ouchi
- From the Whitaker Cardiovascular Institute (K.T., R.M.W., E.E.E., J.L.D., N.O., F.S.) and Cardiovascular Section and Evans Department of Medicine (F.S.), Boston University School of Medicine, MA; and Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (P.E.S.)
| | - Flora Sam
- From the Whitaker Cardiovascular Institute (K.T., R.M.W., E.E.E., J.L.D., N.O., F.S.) and Cardiovascular Section and Evans Department of Medicine (F.S.), Boston University School of Medicine, MA; and Touchstone Diabetes Center, Departments of Internal Medicine and Cell Biology, University of Texas Southwestern Medical Center, Dallas (P.E.S.).
| |
Collapse
|
39
|
Vizzardi E, Regazzoni V, Caretta G, Gavazzoni M, Sciatti E, Bonadei I, Trichaki E, Raddino R, Metra M. Mineralocorticoid receptor antagonist in heart failure: Past, present and future perspectives. INTERNATIONAL JOURNAL OF CARDIOLOGY. HEART & VESSELS 2014; 3:6-14. [PMID: 29450163 PMCID: PMC5801434 DOI: 10.1016/j.ijchv.2014.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 03/08/2014] [Indexed: 12/24/2022]
Abstract
Aldosterone is involved in various deleterious effects on the cardiovascular system, including sodium and fluid retention, myocardial fibrosis, vascular stiffening, endothelial dysfunction, catecholamine release and stimulation of cardiac arrhythmias. Therefore, aldosterone receptor blockade may have several potential benefits in patients with cardiovascular disease. Mineralocorticoid receptor antagonists (MRAs) have been shown to prevent many of the maladaptive effects of aldosterone, in particular among patients with heart failure (HF). Randomized controlled trials have demonstrated efficacy of MRA in heart failure with reduced ejection fraction, both in patients with NYHA functional classes III and IV and in asymptomatic and mildly symptomatic patients (NYHA classes I and II). Recent data in patients with heart failure with preserved ejection fraction are encouraging. MRA could also have anti-arrhythmic effects on atrial and ventricular arrhythmias and may be helpful in patient ischemic heart disease through prevention of myocardial fibrosis and vascular damage. This article aims to discuss the pathophysiological effects of aldosterone in patients with cardiovascular disease and to review the current data that support the use of MRA in heart failure.
Collapse
Affiliation(s)
- Enrico Vizzardi
- Corresponding author. Tel.: + 39 303995679; fax: + 39 303996801Corresponding author.Tel.: + 39 303995679; fax: + 39 303996801
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Changes in cardiopulmonary reserve and peripheral arterial function concomitantly with subclinical inflammation and oxidative stress in patients with heart failure with preserved ejection fraction. Int J Vasc Med 2014; 2014:917271. [PMID: 24719767 PMCID: PMC3955597 DOI: 10.1155/2014/917271] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 01/14/2014] [Indexed: 11/24/2022] Open
Abstract
Background. Changes in cardiopulmonary reserve and biomarkers related to wall stress, inflammation, and oxidative stress concomitantly with the evaluation of peripheral arterial blood flow have not been investigated in patients with heart failure with preserved ejection fraction (HFpEF) compared with healthy subjects (CTL). Methods and Results. Eighteen HFpEF patients and 14 CTL were recruited. Plasma levels of inflammatory and oxidative stress biomarkers were measured at rest. Brain natriuretic peptide (BNP) was measured at rest and peak exercise. Cardiopulmonary reserve was assessed using an exercise protocol with gas exchange analyses. Peripheral arterial blood flow was determined by strain gauge plethysmography. Peak VO2 (12.0 ± 0.4 versus 19.1 ± 1.1 mL/min/kg, P < 0.001) and oxygen uptake efficiency slope (1.55 ± 0.12 versus 2.06 ± 0.14, P < 0.05) were significantly decreased in HFpEF patients compared with CTL. BNP at rest and following stress, C-reactive-protein, interleukin-6, and TBARS were significantly elevated in HFpEF. Both basal and posthyperemic arterial blood flow were not significantly different between the HFpEF patients and CTL. Conclusions. HFpEF exhibits a severe reduction in cardiopulmonary reserve and oxygen uptake efficiency concomitantly with an elevation in a broad spectrum of biomarkers confirming an inflammatory and prooxidative status in patients with HFpEF.
Collapse
|
41
|
Liu G, Liu G, Alzoubi K, Umbach AT, Pelzl L, Borst O, Gawaz M, Lang F. Upregulation of store operated Ca channel Orai1, stimulation of Ca(2+) entry and triggering of cell membrane scrambling in platelets by mineralocorticoid DOCA. Kidney Blood Press Res 2014; 38:21-30. [PMID: 24525794 DOI: 10.1159/000355750] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Mineralocorticoid excess leads to vascular injury, which is partially due to hypertension but in addition involves increased concentration of cytosolic Ca(2+) concentration in platelets, key players in the pathophysiology of occlusive vascular disease. Mineralocorticoids are in part effective by rapid nongenomic mechanisms including phosphatidylinositide-3-kinase (PI3K) signaling, which involves activation of the serum & glucocorticoid inducible kinase (SGK) isoforms. SGK1 has in turn been shown to participate in the regulation of the pore forming Ca(2+) channel protein Orai1 in platelets. Orai1 accomplishes entry of Ca(2+), which is in turn known to trigger cell membrane scrambling. Platelets lack nuclei but are able to express protein by translation, which is stimulated by PI3K signaling. The present study explored whether the mineralocorticoid desoxycorticosterone acetate (DOCA) influences platelet Orai1 protein abundance, cytosolic Ca(2+)-activity ([Ca(2+)]i), phosphatidylserine abundance at the cell surface and/or cell volume. METHODS Orai1 protein abundance was estimated utilizing CF™488A conjugated antibodies, [Ca(2+)]i utilizing Fluo3-fluorescence, phosphatidylserine abundance utilizing FITC-labelled annexin V, and cell volume utilizing forward scatter in flow cytometry. RESULTS DOCA (10 µg/ml) treatment of murine platelets was followed by a significant increase of Orai1 protein abundance, [Ca(2+)]i, percentage of phosphatidylserine exposing platelets and platelet swelling. The effect on [Ca(2+)]i, phosphatidylserine abundance and cell volume were completely abrogated by addition of the specific SGK inhibitor EMD638683 (50 µM) CONCLUSIONS: The mineralocorticoid DOCA upregulates Orai1 protein abundance in the cell membrane, thus increasing [Ca(2+)]i and triggering phosphatidylserine abundance, effects paralleled by platelet swelling.
Collapse
Affiliation(s)
- Guoxing Liu
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Catena C, Colussi G, Nait F, Martinis F, Pezzutto F, Sechi LA. Aldosterone and the heart: still an unresolved issue? Front Endocrinol (Lausanne) 2014; 5:168. [PMID: 25352832 PMCID: PMC4196571 DOI: 10.3389/fendo.2014.00168] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 09/26/2014] [Indexed: 11/21/2022] Open
Abstract
Receptors for mineralocorticoid hormones are expressed in myocardial cells and evidence obtained in animal studies suggests that activation of these receptors causes cardiac damage independent from blood pressure levels. In the last years, many of the issues related to the effects of aldosterone on the heart have received convincing answers and clinical investigation has focused on a variety of conditions including systolic and diastolic heart failure, arrhythmia, primary hypertension, and primary aldosteronism. Some issues, however, await clarification in order to obtain better understanding of what could be the role of aldosterone blockade in prevention and treatment of cardiovascular diseases. In this article, we overview the most recent findings of animal studies that have examined the contribution of aldosterone to cardiac function and clinical studies that have investigated the influence of aldosterone on left ventricular structure and function in the setting of primary hypertension and primary aldosteronism.
Collapse
Affiliation(s)
- Cristiana Catena
- Hypertension Unit, Department of Experimental and Clinical Medical Sciences, Clinica Medica, University Hospital, University of Udine, Udine, Italy
| | - GianLuca Colussi
- Hypertension Unit, Department of Experimental and Clinical Medical Sciences, Clinica Medica, University Hospital, University of Udine, Udine, Italy
| | - Francesca Nait
- Hypertension Unit, Department of Experimental and Clinical Medical Sciences, Clinica Medica, University Hospital, University of Udine, Udine, Italy
| | - Flavia Martinis
- Hypertension Unit, Department of Experimental and Clinical Medical Sciences, Clinica Medica, University Hospital, University of Udine, Udine, Italy
| | - Francesca Pezzutto
- Hypertension Unit, Department of Experimental and Clinical Medical Sciences, Clinica Medica, University Hospital, University of Udine, Udine, Italy
| | - Leonardo A. Sechi
- Hypertension Unit, Department of Experimental and Clinical Medical Sciences, Clinica Medica, University Hospital, University of Udine, Udine, Italy
- *Correspondence: Leonardo A. Sechi, Hypertension Unit, Department of Experimental and Clinical Medical Sciences, Clinica Medica, University Hospital, University of Udine, Building 8, Udine 33100, Italy e-mail:
| |
Collapse
|
43
|
Bishu K, Ogut O, Kushwaha S, Mohammed SF, Ohtani T, Xu X, Brozovich FV, Redfield MM. Anti-remodeling effects of rapamycin in experimental heart failure: dose response and interaction with angiotensin receptor blockade. PLoS One 2013; 8:e81325. [PMID: 24312548 PMCID: PMC3849273 DOI: 10.1371/journal.pone.0081325] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/11/2013] [Indexed: 01/11/2023] Open
Abstract
While neurohumoral antagonists improve outcomes in heart failure (HF), cardiac remodeling and dysfunction progress and outcomes remain poor. Therapies superior or additive to standard HF therapy are needed. Pharmacologic mTOR inhibition by rapamycin attenuated adverse cardiac remodeling and dysfunction in experimental heart failure (HF). However, these studies used rapamycin doses that produced blood drug levels targeted for primary immunosuppression in human transplantation and therefore the immunosuppressive effects may limit clinical translation. Further, the relative or incremental effect of rapamycin combined with standard HF therapies targeting upstream regulators of cardiac remodeling (neurohumoral antagonists) has not been defined. Our objectives were to determine if anti-remodeling effects of rapamycin were preserved at lower doses and whether rapamycin effects were similar or additive to a standard HF therapy (angiotensin receptor blocker (losartan)). Experimental murine HF was produced by transverse aortic constriction (TAC). At three weeks post-TAC, male mice with established HF were treated with placebo, rapamycin at a dose producing immunosuppressive drug levels (target dose), low dose (50% target dose) rapamycin, losartan or rapamycin + losartan for six weeks. Cardiac structure and function (echocardiography, catheterization, pathology, hypertrophic and fibrotic gene expression profiles) were assessed. Downstream mTOR signaling pathways regulating protein synthesis (S6K1 and S6) and autophagy (LC3B-II) were characterized. TAC-HF mice displayed eccentric hypertrophy, systolic dysfunction and pulmonary congestion. These perturbations were attenuated to a similar degree by oral rapamycin doses achieving target (13.3±2.1 ng/dL) or low (6.7±2.5 ng/dL) blood levels. Rapamycin treatment decreased mTOR mediated regulators of protein synthesis and increased mTOR mediated regulators of autophagy. Losartan monotherapy did not attenuate remodeling, whereas Losartan added to rapamycin provided no incremental benefit over rapamycin alone. These data lend support to investigation of low dose rapamycin as a novel therapy in human HF.
Collapse
Affiliation(s)
- Kalkidan Bishu
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Ozgur Ogut
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Sudhir Kushwaha
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Selma F. Mohammed
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tomohito Ohtani
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Xiaolei Xu
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Frank V. Brozovich
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Margaret M. Redfield
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
44
|
Coelho-Filho OR, Shah RV, Mitchell R, Neilan TG, Moreno H, Simonson B, Kwong R, Rosenzweig A, Das S, Jerosch-Herold M. Quantification of cardiomyocyte hypertrophy by cardiac magnetic resonance: implications for early cardiac remodeling. Circulation 2013; 128:1225-33. [PMID: 23912910 PMCID: PMC5308548 DOI: 10.1161/circulationaha.112.000438] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 07/24/2013] [Indexed: 12/29/2022]
Abstract
BACKGROUND Cardiomyocyte hypertrophy is a critical precursor to the development of heart failure. Methods to phenotype cellular hypertrophy noninvasively are limited. The goal was to validate a cardiac magnetic resonance-based approach for the combined assessment of extracellular matrix expansion and cardiomyocyte hypertrophy. METHODS AND RESULTS Two murine models of hypertension (n=18, with n=15 controls) induced by l-N(G)-nitroarginine methyl ester (L-NAME) and pressure overload (n=11) from transaortic constriction (TAC) were imaged by cardiac magnetic resonance at baseline and 7 weeks after L-NAME treatment or up to 7 weeks after TAC. T1 relaxation times were measured before and after gadolinium contrast. The intracellular lifetime of water (τic), a cell size-dependent parameter, and extracellular volume fraction, a marker of interstitial fibrosis, were determined with a model for transcytolemmal water exchange. Cardiomyocyte diameter and length were measured on FITC-wheat germ agglutinin-stained sections. The τic correlated strongly with histological cardiomyocyte volume-to-surface ratio (r=0.78, P<0.001) and cell volume (r=0.75, P<0.001). Histological cardiomyocyte diameters and cell volumes were higher in mice treated with L-NAME compared with controls (P<0.001). In the TAC model, cardiac magnetic resonance and histology showed cell hypertrophy at 2 weeks after TAC without significant fibrosis at this early time point. Mice exposed to TAC demonstrated a significant, longitudinal, and parallel increase in histological cell volume, volume-to-surface ratio, and τic between 2 and 7 weeks after TAC. CONCLUSION The τic measured by contrast-enhanced cardiac magnetic resonance provides a noninvasive measure of cardiomyocyte hypertrophy. Extracellular volume fraction and τic can track myocardial tissue remodeling from pressure overload.
Collapse
Affiliation(s)
- Otavio R Coelho-Filho
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02114
- Department of Internal Medicine, State University of Campinas (UNICAMP), Boston, MA
| | - Ravi V Shah
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02114
| | - Richard Mitchell
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Tomas G Neilan
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02114
| | - Heitor Moreno
- Department of Internal Medicine, State University of Campinas (UNICAMP), Boston, MA
| | - Bridget Simonson
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Raymond Kwong
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02114
| | - Anthony Rosenzweig
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Saumya Das
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | | |
Collapse
|
45
|
Essick EE, Wilson RM, Pimentel DR, Shimano M, Baid S, Ouchi N, Sam F. Adiponectin modulates oxidative stress-induced autophagy in cardiomyocytes. PLoS One 2013; 8:e68697. [PMID: 23894332 PMCID: PMC3716763 DOI: 10.1371/journal.pone.0068697] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/02/2013] [Indexed: 11/18/2022] Open
Abstract
Diastolic heart failure (HF) i.e., "HF with preserved ejection fraction" (HF-preserved EF) accounts for up to 50% of all HF presentations; however there have been no therapeutic advances. This stems in part from an incomplete understanding about HF-preserved EF. Hypertension is the major cause of HF-preserved EF whilst HF-preserved EF is also highly associated with obesity. Similarly, excessive reactive oxygen species (ROS), i.e., oxidative stress occurs in hypertension and obesity, sensitizing the heart to the renin-angiotensin-aldosterone system, inducing autophagic type-II programmed cell death and accelerating the propensity to adverse cardiac remodeling, diastolic dysfunction and HF. Adiponectin (APN), an adipokine, mediates cardioprotective actions but it is unknown if APN modulates cardiomyocyte autophagy. We tested the hypothesis that APN ameliorates oxidative stress-induced autophagy in cardiomyocytes. Isolated adult rat ventricular myocytes were pretreated with recombinant APN (30 µg/mL) followed by 1mM hydrogen peroxide (H2O2) exposure. Wild type (WT) and APN-deficient (APN-KO) mice were infused with angiotensin (Ang)-II (3.2 mg/kg/d) for 14 days to induced oxidative stress. Autophagy-related proteins, mTOR, AMPK and ERK expression were measured. H2O2 induced LC3I to LC3II conversion by a factor of 3.4±1.0 which was abrogated by pre-treatment with APN by 44.5±10%. However, neither H2O2 nor APN affected ATG5, ATG7, or Beclin-1 expression. H2O2 increased phospho-AMPK by 49±6.0%, whilst pretreatment with APN decreased phospho-AMPK by 26±4%. H2O2 decreased phospho-mTOR by 36±13%, which was restored by APN. ERK inhibition demonstrated that the ERK-mTOR pathway is involved in H2O2-induced autophagy. Chronic Ang-II infusion significantly increased myocardial LC3II/I protein expression ratio in APN-KO vs. WT mice. These data suggest that excessive ROS caused cardiomyocyte autophagy which was ameliorated by APN by inhibiting an H2O2-induced AMPK/mTOR/ERK-dependent mechanism. These findings demonstrate the anti-oxidant potential of APN in oxidative stress-associated cardiovascular diseases, such as hypertension-induced HF-preserved EF.
Collapse
Affiliation(s)
- Eric E. Essick
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Richard M. Wilson
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - David R. Pimentel
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Cardiovascular Section and Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Masayuki Shimano
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Simoni Baid
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Noriyuki Ouchi
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Flora Sam
- Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Cardiovascular Section and Evans Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
46
|
Guichard JL, Clark D, Calhoun DA, Ahmed MI. Aldosterone receptor antagonists: current perspectives and therapies. Vasc Health Risk Manag 2013; 9:321-31. [PMID: 23836977 PMCID: PMC3699348 DOI: 10.2147/vhrm.s33759] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aldosterone is a downstream effector of angiotensin II in the renin-angiotensin-aldosterone system and binds to the mineralocorticoid receptor. The classical view of aldosterone primarily acting at the level of the kidneys to regulate plasma potassium and intravascular volume status is being supplemented by evidence of new "off-target" effects of aldosterone in other organ systems. The genomic effects of aldosterone are well known, but there is also evidence for non-genomic effects and these recently identified effects of aldosterone have required a revision in the traditional view of aldosterone's role in human health and disease. The aim of this article is to review the biological action of aldosterone and the mineralocorticoid receptor leading to subsequent physiologic and pathophysiologic effects involving the vasculature, central nervous system, heart, and kidneys. Furthermore, we outline current evidence evaluating the use of mineralocorticoid receptor antagonists in the treatment of primary aldosteronism, primary hypertension, resistant hypertension, obstructive sleep apnea, heart failure, and chronic kidney disease.
Collapse
Affiliation(s)
- Jason L Guichard
- University of Alabama at Birmingham, Department of Medicine, Division of Cardiovascular Disease, Birmingham, AL 35294-2041 , USA
| | | | | | | |
Collapse
|
47
|
Chung CS, Hutchinson KR, Methawasin M, Saripalli C, Smith JE, Hidalgo CG, Luo X, Labeit S, Guo C, Granzier HL. Shortening of the elastic tandem immunoglobulin segment of titin leads to diastolic dysfunction. Circulation 2013; 128:19-28. [PMID: 23709671 DOI: 10.1161/circulationaha.112.001268] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Diastolic dysfunction is a poorly understood but clinically pervasive syndrome that is characterized by increased diastolic stiffness. Titin is the main determinant of cellular passive stiffness. However, the physiological role that the tandem immunoglobulin (Ig) segment of titin plays in stiffness generation and whether shortening this segment is sufficient to cause diastolic dysfunction need to be established. METHODS AND RESULTS We generated a mouse model in which 9 Ig-like domains (Ig3-Ig11) were deleted from the proximal tandem Ig segment of the spring region of titin (IG KO). Exon microarray analysis revealed no adaptations in titin splicing, whereas novel phospho-specific antibodies did not detect changes in titin phosphorylation. Passive myocyte stiffness was increased in the IG KO, and immunoelectron microscopy revealed increased extension of the remaining titin spring segments as the sole likely underlying mechanism. Diastolic stiffness was increased at the tissue and organ levels, with no consistent changes in extracellular matrix composition or extracellular matrix-based passive stiffness, supporting a titin-based mechanism for in vivo diastolic dysfunction. Additionally, IG KO mice have a reduced exercise tolerance, a phenotype often associated with diastolic dysfunction. CONCLUSIONS Increased titin-based passive stiffness is sufficient to cause diastolic dysfunction with exercise intolerance.
Collapse
Affiliation(s)
- Charles S Chung
- Department of Physiology, University of Arizona, PO Box245051, Tucson AZ 85724, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Decreased Nox4 levels in the myocardium of patients with aortic valve stenosis. Clin Sci (Lond) 2013; 125:291-300. [DOI: 10.1042/cs20120612] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The NADPH oxidases are a key family of ROS (reactive oxygen species)-producing enzymes which may differentially contribute to cardiac pathophysiology. Animal studies show uncertain results regarding the regulation of cardiac Nox4 by pressure overload and no data are available on human myocardial Nox4. In the present study, we evaluated Nox4 expression and its relationship with myocardial remodelling and LV (left ventricular) function in patients with severe AS (aortic valve stenosis). Endomyocardial biopsies from 34 patients with AS were obtained during aortic valve replacement surgery. LV morphology and function were assessed by echocardiography. Myocardial samples from subjects deceased of non-CVDs (cardiovascular diseases) were analysed as controls. Nox4 localization was evaluated by immunohistochemistry and quantified by Western blot. Myocardial capillary density, fibrosis and cardiomyocyte dimensions and apoptosis were assessed histologically to evaluate myocardial remodelling. Nox4 was present in samples from all subjects and expressed in cardiomyocytes, VSMCs (vascular smooth muscle cells), endothelium and fibroblasts. Nox4 levels were reduced 5-fold in AS patients compared with controls (P<0.01). Nox4 levels directly correlated with cardiomyocyte cross-sectional area (r=0.299, P<0.05) and diameter (r=0.406, P<0.05) and capillary density (r=0.389, P<0.05), and inversely with cardiomyocyte apoptosis (r=−0.316, P<0.05) in AS patients. In addition, Nox4 levels correlated with echocardiographic parameters (LV ejection fraction: r=0.353, P<0.05; midwall fractional shortening: r=0.355, P<0.05; deceleration time: r=−0.345, P<0.05) in AS patients. Nox4 is expressed in human myocardium and reduced in AS patients. The observed associations of Nox4 with cardiomyocyte parameters and capillary density in AS patients suggest a potential role of Nox4 deficiency in the myocardial remodelling present in the human pressure-overloaded heart.
Collapse
|
49
|
Rugale C, Oudot C, Desmetz C, Guzman C, Lajoix A, Jover B. [Sodium restriction prevents cardiovascular remodeling associated with insulin-resistance in the rat]. Ann Cardiol Angeiol (Paris) 2013; 62:139-43. [PMID: 23601354 DOI: 10.1016/j.ancard.2013.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 03/08/2013] [Indexed: 11/19/2022]
Abstract
AIM OF THE STUDY In the present work, the objective was to evaluate the influence of a dietary sodium restriction on cardiovascular morphology changes associated with insulin-resistance. ANIMALS AND PROTOCOL At 8 weeks of age, rats were fed for 12 weeks a 60%-fructose diet containing a regular sodium content (0.64%) or totally lacking in sodium chloride (<0.01%). A group of rats fed a wheat starch-based diet with regular sodium content served as control group. RESULTS Elevated HOMA index and plasma insulin confirm the presence of insulin-resistance in fructose-fed rats. Concomitantly, an increase in cardiac mass and in cardiac collagen (Sirius red staining) was detected without obvious change in arterial pressure or cardiac aldosterone synthase mRNA expression. In addition, cross-sectional area of the carotid artery was higher in fructose-fed rats. Production of superoxide anion, equated with dihydroethidium (DHE) staining, was enhanced in cardiac tissue of rats with insulin-resistance. Withdrawal of sodium from the fructose diet prevented all the cardiovascular effects of fructose consumption, including DHE staining. CONCLUSION These results are in favor of the participation of oxidative stress normalization in the beneficial influence of dietary sodium deprivation on cardiovascular remodeling in this model of insulin-resistance in rats.
Collapse
Affiliation(s)
- C Rugale
- EA7288, groupe rein et hypertension, PRES Sud de France, institut universitaire de recherche clinique, 641, avenue du Doyen-Gaston-Giraud, 34093 Montpellier cedex 5, France
| | | | | | | | | | | |
Collapse
|
50
|
Perlini S, Chung ES, Aurigemma GP, Meyer TE. Alterations in Early Filling Dynamics Predict the Progression of Compensated Pressure Overload Hypertrophy to Heart Failure Better than Abnormalities in Midwall Systolic Shortening. Clin Exp Hypertens 2012; 35:401-11. [DOI: 10.3109/10641963.2012.739235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|