51
|
Hulot JS, Trochu JN, Donal E, Galinier M, Logeart D, De Groote P, Juillière Y. Vericiguat for the treatment of heart failure: mechanism of action and pharmacological properties compared with other emerging therapeutic options. Expert Opin Pharmacother 2021; 22:1847-1855. [PMID: 34074190 DOI: 10.1080/14656566.2021.1937121] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
INTRODUCTION The significant morbidity and mortality in patients with heart failure (HF), notably in the most advanced forms of the disease, justify the need for novel therapeutic options. In the last year, the soluble guanylate cyclase (sGC) stimulator, vericiguat, has drawn the attention of the medical community following the report of reduced clinical outcomes in patients with worsening chronic HF (WCHF). AREAS COVERED The authors review the available data on the mechanism of action of vericiguat (cyclic guanosine monophosphate (cGMP) pathway), its clinical development program, its role in HF management, and its future positioning in the therapeutic recommendations. EXPERT OPINION cGMP deficiency has deleterious effects on the heart and contributes to the progression of HF. Different molecules, including nitric oxide (NO) donors, phosphodiesterase inhibitors, and natriuretic peptides analogues, target the NO-sCG-cGMP pathway but have yielded conflicting results in HF patients. Vericiguat acts as a sGC stimulator thus targeting the NO-sGC-cGMP pathway by a different mechanism that complements the current pharmacotherapy for HF. Vericiguat has shown an additional statistical add-on therapy efficacy by reducing morbi-mortality in patients with WCHF. A better evaluation of HF severity might be an important determinant to guide the use of vericiguat among the available therapies.
Collapse
Affiliation(s)
- Jean-Sébastien Hulot
- Université de Paris, INSERM, PARCC, Paris, France.,CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, Paris, France
| | - Jean-Noël Trochu
- Université de Nantes, CHU Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France
| | - Erwan Donal
- LTSI INSERM U1099, Service de Cardiologie et Maladies Vasculaires, Centre Hospitalier Universitaire de Rennes, Université Rennes, Rennes, France
| | - Michel Galinier
- Department of Cardiology, Toulouse-Rangueil University Hospital, Toulouse University School of Medicine, Toulouse, France
| | - Damien Logeart
- Department of Cardiology, Lariboisière Hospital, Paris, France.,Université de Paris, Paris, France
| | - Pascal De Groote
- CHU Lille, Service de Cardiologie, France.,INSERM U1167, Institut Pasteur de Lille, Lille, France
| | - Yves Juillière
- Centre Hospitalier Universitaire de Nancy Brabois, Vandoeuvre Lès Nancy, France
| |
Collapse
|
52
|
Complex functionality of protein phosphatase 1 isoforms in the heart. Cell Signal 2021; 85:110059. [PMID: 34062239 DOI: 10.1016/j.cellsig.2021.110059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/21/2021] [Accepted: 05/28/2021] [Indexed: 02/04/2023]
Abstract
Protein phosphatase 1(PP1) is a key regulator of cardiac function through dephosphorylating serine/threonine residues within target proteins to oppose the function of protein kinases. Studies from failing hearts of animal models and human patients have demonstrated significant increase of PP1 activity in myocardium, while elevated PP1 activity in transgenic mice leads to cardiac dysfunction, suggesting that PP1 might be a therapeutic target to ameliorate cardiac dysfunction in failing hearts. In fact, cardiac overexpression of inhibitor 1, the endogenous inhibitor of PP1, increases cardiac contractility and suppresses heart failure progression. However, this notion of PP1 inhibition for heart failure treatment has been challenged by recent studies on the isoform-specific roles of PP1 in the heart. PP1 is a holoenzyme composed of catalytic subunits (PP1α, PP1β, or PP1γ) and regulatory proteins that target them to distinct subcellular locations for functional specificity. This review will summarize how PP1 regulates phosphorylation of some of the key cardiac proteins involved in Ca2+ handling and cardiac contraction, and the potential role of PP1 isoforms in controlling cardiac physiology and pathophysiology.
Collapse
|
53
|
Paulus WJ, Zile MR. From Systemic Inflammation to Myocardial Fibrosis: The Heart Failure With Preserved Ejection Fraction Paradigm Revisited. Circ Res 2021; 128:1451-1467. [PMID: 33983831 PMCID: PMC8351796 DOI: 10.1161/circresaha.121.318159] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In accordance with the comorbidity-inflammation paradigm, comorbidities and especially metabolic comorbidities are presumed to drive development and severity of heart failure with preserved ejection fraction through a cascade of events ranging from systemic inflammation to myocardial fibrosis. Recently, novel experimental and clinical evidence emerged, which strengthens the validity of the inflammatory/profibrotic paradigm. This evidence consists among others of (1) myocardial infiltration by immunocompetent cells not only because of an obesity-induced metabolic load but also because of an arterial hypertension-induced hemodynamic load. The latter is sensed by components of the extracellular matrix like basal laminin, which also interact with cardiomyocyte titin; (2) expression in cardiomyocytes of inducible nitric oxide synthase because of circulating proinflammatory cytokines. This results in myocardial accumulation of degraded proteins because of a failing unfolded protein response; (3) definition by machine learning algorithms of phenogroups of patients with heart failure with preserved ejection fraction with a distinct inflammatory/profibrotic signature; (4) direct coupling in mediation analysis between comorbidities, inflammatory biomarkers, and deranged myocardial structure/function with endothelial expression of adhesion molecules already apparent in early preclinical heart failure with preserved ejection fraction (HF stage A, B). This new evidence paves the road for future heart failure with preserved ejection fraction treatments such as biologicals directed against inflammatory cytokines, stimulation of protein ubiquitylation with phosphodiesterase 1 inhibitors, correction of titin stiffness through natriuretic peptide-particulate guanylyl cyclase-PDE9 (phosphodiesterase 9) signaling and molecular/cellular regulatory mechanisms that control myocardial fibrosis.
Collapse
Affiliation(s)
- Walter J Paulus
- Amsterdam University Medical Centers, The Netherlands (W.J.P.)
| | - Michael R Zile
- RHJ Department of Veterans Affairs Medical Center, Medical University of South Carolina, Charleston (M.R.Z.)
| |
Collapse
|
54
|
Chen J, Norling LV, Cooper D. Cardiac Dysfunction in Rheumatoid Arthritis: The Role of Inflammation. Cells 2021; 10:881. [PMID: 33924323 PMCID: PMC8070480 DOI: 10.3390/cells10040881] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/08/2021] [Accepted: 04/10/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatoid arthritis is a chronic, systemic inflammatory disease that carries an increased risk of mortality due to cardiovascular disease. The link between inflammation and atherosclerotic disease is clear; however, recent evidence suggests that inflammation may also play a role in the development of nonischemic heart disease in rheumatoid arthritis (RA) patients. We consider here the link between inflammation and cardiovascular disease in the RA community with a focus on heart failure with preserved ejection fraction. The effect of current anti-inflammatory therapeutics, used to treat RA patients, on cardiovascular disease are discussed as well as whether targeting resolution of inflammation might offer an alternative strategy for tempering inflammation and subsequent inflammation-driven comorbidities in RA.
Collapse
Affiliation(s)
- Jianmin Chen
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.C.); (L.V.N.)
| | - Lucy V. Norling
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.C.); (L.V.N.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| | - Dianne Cooper
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (J.C.); (L.V.N.)
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London EC1M 6BQ, UK
| |
Collapse
|
55
|
Liu Y, Chen J, Fontes SK, Bautista EN, Cheng Z. Physiological And Pathological Roles Of Protein Kinase A In The Heart. Cardiovasc Res 2021; 118:386-398. [PMID: 33483740 DOI: 10.1093/cvr/cvab008] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/30/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Protein kinase A (PKA) is a central regulator of cardiac performance and morphology. Myocardial PKA activation is induced by a variety of hormones, neurotransmitters and stress signals, most notably catecholamines secreted by the sympathetic nervous system. Catecholamines bind β-adrenergic receptors to stimulate cAMP-dependent PKA activation in cardiomyocytes. Elevated PKA activity enhances Ca2+ cycling and increases cardiac muscle contractility. Dynamic control of PKA is essential for cardiac homeostasis, as dysregulation of PKA signaling is associated with a broad range of heart diseases. Specifically, abnormal PKA activation or inactivation contributes to the pathogenesis of myocardial ischemia, hypertrophy, heart failure, as well as diabetic, takotsubo, or anthracycline cardiomyopathies. PKA may also determine sex-dependent differences in contractile function and heart disease predisposition. Here, we describe the recent advances regarding the roles of PKA in cardiac physiology and pathology, highlighting previous study limitations and future research directions. Moreover, we discuss the therapeutic strategies and molecular mechanisms associated with cardiac PKA biology. In summary, PKA could serve as a promising drug target for cardioprotection. Depending on disease types and mechanisms, therapeutic intervention may require either inhibition or activation of PKA. Therefore, specific PKA inhibitors or activators may represent valuable drug candidates for the treatment of heart diseases.
Collapse
Affiliation(s)
- Yuening Liu
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Jingrui Chen
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Shayne K Fontes
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Erika N Bautista
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, PBS 423, 412 E. Spokane Falls Blvd, ., Spokane, WA, 99202-2131, USA
| |
Collapse
|
56
|
Clinical Implications of Uric Acid in Heart Failure: A Comprehensive Review. Life (Basel) 2021; 11:life11010053. [PMID: 33466609 PMCID: PMC7828696 DOI: 10.3390/life11010053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
Affecting more than 26 million people worldwide and with rising prevalence, heart failure (HF) represents a major global health problem. Hence, further research is needed in order to abate poor HF outcomes and mitigate significant expenses that burden health care systems. Based on available data, experts agree that there is an urgent need for a cost-effective prognostic biomarker in HF. Although a significant number of biomarkers have already been investigated in this setting, the clinical utility of adding biomarker evaluation to routine HF care still remains ambiguous. Specifically, in this review we focused on uric acid (UA), a purine metabolism detriment whose role as cardiovascular risk factor has been exhaustingly debated for decades. Multiple large population studies indicate that UA is an independent predictor of mortality in acute and chronic HF, making it a significant prognostic factor in both settings. High serum levels have been also associated with an increased incidence of HF, thus expanding the clinical utility of UA. Importantly, emerging data suggests that UA is also implicated in the pathogenesis of HF, which sheds light on UA as a feasible therapeutic target. Although to date clinical studies have not been able to prove the benefits of xanthine oxidase in HF patients, we discuss the putative role of UA and xanthine oxidase in the pathophysiology of HF as a therapeutic target.
Collapse
|
57
|
Abstract
The 3',5'-cyclic guanosine monophosphate (cGMP)-dependent protein kinase type I (cGKI aka PKGI) is a major cardiac effector acting downstream of nitric oxide (NO)-sensitive soluble guanylyl cyclase and natriuretic peptides (NPs), which signal through transmembrane guanylyl cyclases. Consistent with the wide distribution of the cGMP-generating guanylyl cyclases, cGKI, which usually elicits its cellular effects by direct phosphorylation of its targets, is present in multiple cardiac cell types including cardiomyocytes (CMs). Although numerous targets of cGMP/cGKI in heart were identified in the past, neither their exact patho-/physiological functions nor cell-type specific roles are clear. Herein, we inform about the current knowledge on the signal transduction downstream of CM cGKI. We believe that better insights into the specific actions of cGMP and cGKI in these cells will help to guide future studies in the search for predictive biomarkers for the response to pharmacological cGMP pathway modulation. In addition, targets downstream of cGMP/cGKI may be exploited for refined and optimized diagnostic and therapeutic strategies in different types of heart disease and their causes. Importantly, key functions of these proteins and particularly sites of regulatory phosphorylation by cGKI should, at least in principle, remain intact, although upstream signaling through the second messenger cGMP is impaired or dysregulated in a stressed or diseased heart state.
Collapse
|
58
|
Cuijpers I, Papageorgiou A, Carai P, Herwig M, Mügge A, Klein T, Hamdani N, Jones EAV, Heymans S. Linagliptin prevents left ventricular stiffening by reducing titin cleavage and hypophosphorylation. J Cell Mol Med 2021; 25:729-741. [PMID: 33295687 PMCID: PMC7812306 DOI: 10.1111/jcmm.16122] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 11/08/2020] [Indexed: 12/25/2022] Open
Abstract
The metabolic syndrome (MetS) is an escalating problem worldwide, causing left ventricular stiffening, an early characteristic of diastolic dysfunction for which no treatment exists. As diastolic dysfunction and stiffening in MetS patients are associated with increased circulating dipeptidyl peptidase-4 (DPP-4) levels, we investigated whether the clinically approved DPP-4 inhibitor linagliptin reduces left ventricular stiffness in MetS-induced cardiac disease. Sixteen-week-old obese ZSF1 rats, displaying the MetS and left ventricular stiffness, received linagliptin-supplemented or placebo diet for four weeks. Linagliptin significantly reduced obesity, hyperlipidaemia, and hyperglycaemia and improved left ventricular relaxation. This improved relaxation was related to decreased cardiac fibrosis and cardiomyocyte passive stiffness (Fpassive ). The reduced Fpassive was the result of titin isoform switching from the stiff N2B to the more flexible N2BA and increased phosphorylation of total titin and specifically its N2Bus region (S4080 and S3391). Importantly, DPP-4 directly cleaved titin in vitro, resulting in an increased Fpassive , which was prevented by simultaneous administration of linagliptin. In conclusion, linagliptin improves left ventricular stiffness in obese ZSF1 rats by preventing direct DPP4-mediated titin cleavage, as well as by modulating both titin isoform levels and phosphorylation. Reducing left ventricular stiffness by administering linagliptin might prevent MetS-induced early diastolic dysfunction in human.
Collapse
Affiliation(s)
- Ilona Cuijpers
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Anna‐Pia Papageorgiou
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Paolo Carai
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
| | - Melissa Herwig
- Molecular Cardiology and Experimental CardiologyRuhr University BochumBochumGermany
- Department of CardiologySt. Josef‐HospitalRuhr University BochumBochumGermany
- Institute of PhysiologyRuhr University BochumBochumGermany
| | - Andreas Mügge
- Molecular Cardiology and Experimental CardiologyRuhr University BochumBochumGermany
- Department of CardiologySt. Josef‐HospitalRuhr University BochumBochumGermany
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KGBiberachGermany
| | - Nazha Hamdani
- Molecular Cardiology and Experimental CardiologyRuhr University BochumBochumGermany
- Department of CardiologySt. Josef‐HospitalRuhr University BochumBochumGermany
- Institute of PhysiologyRuhr University BochumBochumGermany
- Department of Clinical PharmacologyRuhr University BochumBochumGermany
| | - Elizabeth A. V. Jones
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Stephane Heymans
- Center for Molecular and Vascular BiologyKU LeuvenLeuvenBelgium
- Department of CardiologyCARIM School for Cardiovascular DiseasesMaastricht University Medical CenterMaastrichtThe Netherlands
- Holland Heart HouseICIN‐Netherlands Heart InstituteUtrechtThe Netherlands
| |
Collapse
|
59
|
Shchendrigina AA, Zhbanov KA, Privalova EV, Iusupova AO, Bytdaeva AH, Danilogorskaya YA, Zheleznykh EA, Suvorov AY, Zektser VY, Mnatsakanyan MG, Lyapidevskaya OV, Khabarova NV, Naymann YI, Belenkov YN, Starostina ES. [Circulating Neuregulin-1 and Chronic Heart Failure with Preserved Ejection]. ACTA ACUST UNITED AC 2020; 60:1222. [PMID: 33487159 DOI: 10.18087/cardio.2020.11.n1222] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 10/08/2020] [Indexed: 11/18/2022]
Abstract
Chronic heart failure (CHF) with preserved ejection fraction (CHFpEF) is an unsolved, socially relevant challenge since it is associated with a high level of morbidity and mortality. Early markers for this pathology are unavailable, and therapeutic approaches are undeveloped. This necessitates extensive studying the mechanisms of CHFpEF to identify therapeutic targets. According to current notions, systemic inflammation and endothelial dysfunction play an important role in the pathogenesis of CHFpEF. These processes induce the development of myocardial fibrosis and impairment of cardiomyocyte relaxation, thereby resulting in diastolic dysfunction and increased left ventricular (LV) filling pressure. Neuregulin-1 (NRG-1) is a paracrine growth factor and a natural agonist of ErbB receptor family synthesized in the endothelium of coronary microvessels. The NRG-1 / ErbB4 system of the heart is activated at early stages of CHFpEF to enhance the cardiomyocyte resistance to oxidative stress. Preclinical and clinical (phases II and III) studies have shown that the recombinant NRG-1 therapy results in improvement of myocardial contractility and in LV reverse remodeling. Results of recent studies suggest possible anti-inflammatory and antifibrotic effects of NRG-1, which warrants studying the activity of this system in patients with CHFpEF.
Collapse
Affiliation(s)
- A A Shchendrigina
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - K A Zhbanov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - E V Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - A O Iusupova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - A H Bytdaeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu A Danilogorskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - E A Zheleznykh
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | | | - V Yu Zektser
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - M G Mnatsakanyan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - O V Lyapidevskaya
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - N V Khabarova
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu I Naymann
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | - Yu N Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow
| | | |
Collapse
|
60
|
Mannozzi J, Massoud L, Kaur J, Coutsos M, O'Leary DS. Ventricular contraction and relaxation rates during muscle metaboreflex activation in heart failure: are they coupled? Exp Physiol 2020; 106:401-411. [PMID: 33226720 DOI: 10.1113/ep089053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/18/2020] [Indexed: 01/08/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does the muscle metaboreflex affect the ratio of left ventricular contraction/relaxation rates and does heart failure impact this relationship. What is the main finding and its importance? The effect of muscle metaboreflex activation on the ventricular relaxation rate was significantly attenuated in heart failure. Heart failure attenuates the exercise and muscle metaboreflex-induced changes in the contraction/relaxation ratio. In heart failure, the reduced ability to raise cardiac output during muscle metaboreflex activation may not solely be due to attenuation of ventricular contraction but also alterations in ventricular relaxation and diastolic function. ABSTRACT The relationship between contraction and relaxation rates of the left ventricle varies with exercise. In in vitro models, this ratio was shown to be relatively unaltered by changes in sarcomere length, frequency of stimulation, and β-adrenergic stimulation. We investigated whether the ratio of contraction to relaxation rate is maintained in the whole heart during exercise and muscle metaboreflex activation and whether heart failure alters these relationships. We observed that in healthy subjects the ratio of contraction to relaxation increases from rest to exercise as a result of a higher increase in contraction relative to relaxation. During muscle metaboreflex activation the ratio of contraction to relaxation is significantly reduced towards 1.0 due to a large increase in relaxation rate matching contraction rate. In heart failure, contraction and relaxation rates are significantly reduced, and increases during exercise are attenuated. A significant increase in the ratio was observed from rest to exercise although baseline ratio values were significantly reduced close to 1.0 when compared to healthy subjects. There was no significant change observed between exercise and muscle metaboreflex activation nor was the ratio during muscle metaboreflex activation significantly different between heart failure and control. We conclude that heart failure reduces the muscle metaboreflex gain and contraction and relaxation rates. Furthermore, we observed that the ratio of the contraction and relaxation rates during muscle metaboreflex activation is not significantly different between control and heart failure, but significant changes in the ratio in healthy subjects due to increased relaxation rate were abolished in heart failure.
Collapse
Affiliation(s)
- Joseph Mannozzi
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Louis Massoud
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jasdeep Kaur
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Matthew Coutsos
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Donal S O'Leary
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
61
|
Michel K, Herwig M, Werner F, Špiranec Spes K, Abeßer M, Schuh K, Dabral S, Mügge A, Baba HA, Skryabin BV, Hamdani N, Kuhn M. C-type natriuretic peptide moderates titin-based cardiomyocyte stiffness. JCI Insight 2020; 5:139910. [PMID: 33055420 PMCID: PMC7710274 DOI: 10.1172/jci.insight.139910] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Heart failure is often accompanied by titin-dependent myocardial stiffness. Phosphorylation of titin by cGMP-dependent protein kinase I (PKGI) increases cardiomyocyte distensibility. The upstream pathways stimulating PKGI-mediated titin phosphorylation are unclear. We studied whether C-type natriuretic peptide (CNP), via its guanylyl cyclase-B (GC-B) receptor and cGMP/PKGI signaling, modulates titin-based ventricular compliance. To dissect GC-B–mediated effects of endogenous CNP in cardiomyocytes, we generated mice with cardiomyocyte-restricted GC-B deletion (CM GC-B–KO mice). The impact on heart morphology and function, myocyte passive tension, and titin isoform expression and phosphorylation was studied at baseline and after increased afterload induced by transverse aortic constriction (TAC). Pressure overload increased left ventricular endothelial CNP expression, with an early peak after 3 days. Concomitantly, titin phosphorylation at Ser4080, the site phosphorylated by PKGI, was augmented. Notably, in CM GC-B–KO mice this titin response was abolished. TAC-induced hypertrophy and fibrosis were not different between genotypes. However, the KO mice presented mild systolic and diastolic dysfunction together with myocyte stiffness, which were not observed in control littermates. In vitro, recombinant PKGI rescued reduced titin-Ser4080 phosphorylation and reverted passive stiffness of GC-B–deficient cardiomyocytes. CNP-induced activation of GC-B/cGMP/PKGI signaling in cardiomyocytes provides a protecting regulatory circuit preventing titin-based myocyte stiffening during early phases of pressure overload. C-type natriuretic peptide via GC-B/cGMP/PKGI signalling in cardiomyocytes attenuates titin-based cardiomyocyte stiffening during early phases of pressure-overload.
Collapse
Affiliation(s)
- Konstanze Michel
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| | - Melissa Herwig
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Franziska Werner
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | | | - Marco Abeßer
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Kai Schuh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Swati Dabral
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Andreas Mügge
- Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Hideo A Baba
- Institute of Pathology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Boris V Skryabin
- Medical Faculty, Core Facility TRAnsgenic animal and genetic engineering Models (TRAM), University of Münster, Münster, Germany
| | - Nazha Hamdani
- Institute of Physiology and.,Department of Cardiology, St-Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany.,Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
62
|
Berezin AE, Berezin AA, Lichtenauer M. Emerging Role of Adipocyte Dysfunction in Inducing Heart Failure Among Obese Patients With Prediabetes and Known Diabetes Mellitus. Front Cardiovasc Med 2020; 7:583175. [PMID: 33240938 PMCID: PMC7667132 DOI: 10.3389/fcvm.2020.583175] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue dysfunction is a predictor for cardiovascular (CV) events and heart failure (HF) in patient population with obesity, metabolic syndrome, and known type 2 diabetes mellitus. Previous preclinical and clinical studies have yielded controversial findings regarding the role of accumulation of adipose tissue various types in CV risk and HF-related clinical outcomes in obese patients. There is evidence for direct impact of infiltration of epicardial adipocytes into the underlying myocardium to induce adverse cardiac remodeling and mediate HF development and atrial fibrillation. Additionally, perivascular adipocytes accumulation is responsible for release of proinflammatory adipocytokines (adiponectin, leptin, resistin), stimulation of oxidative stress, macrophage phenotype switching, and worsening vascular reparation, which all lead to microvascular inflammation, endothelial dysfunction, atherosclerosis acceleration, and finally to increase in CV mortality. However, systemic effects of white and brown adipose tissue can be different, and adipogenesis including browning of adipose tissue and deficiency of anti-inflammatory adipocytokines (visfatin, omentin, zinc-α2-glycoprotein, glypican-4) was frequently associated with adipose triglyceride lipase augmentation, altered glucose homeostasis, resistance to insulin of skeletal muscles, increased cardiomyocyte apoptosis, lowered survival, and weak function of progenitor endothelial cells, which could significantly influence on HF development, as well as end-organ fibrosis and multiple comorbidities. The exact underlying mechanisms for these effects are not fully understood, while they are essential to help develop improved treatment strategies. The aim of the review is to summarize the evidence showing that adipocyte dysfunction may induce the onset of HF and support advance of HF through different biological mechanisms involving inflammation, pericardial, and perivascular adipose tissue accumulation, adverse and electrical cardiac remodeling, and skeletal muscle dysfunction. The unbalancing effects of natriuretic peptides, neprilysin, and components of renin–angiotensin system, as exacerbating cause of altered adipocytokine signaling on myocardium and vasculature, in obesity patients at high risk of HF are disputed. The profile of proinflammatory and anti-inflammatory adipocytokines as promising biomarker for HF risk stratification is discussed in the review.
Collapse
Affiliation(s)
- Alexander E Berezin
- Internal Medicine Department, State Medical University, Ministry of Health of Ukraine, Zaporozhye, Ukraine
| | - Alexander A Berezin
- Internal Medicine Department, Medical Academy of Post-Graduate Education, Ministry of Health of Ukraine, Zaporozhye, Ukraine
| | - Michael Lichtenauer
- Division of Cardiology, Department of Internal Medicine II, Paracelsus Medical University Salzburg, Salzburg, Austria
| |
Collapse
|
63
|
Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, Garcia-Ropero A, Ishikawa K, Watanabe S, Picatoste B, Vargas-Delgado AP, Flores-Umanzor EJ, Sanz J, Fuster V, Badimon JJ. Empagliflozin Ameliorates Diastolic Dysfunction and Left Ventricular Fibrosis/Stiffness in Nondiabetic Heart Failure: A Multimodality Study. JACC Cardiovasc Imaging 2020; 14:393-407. [PMID: 33129742 DOI: 10.1016/j.jcmg.2020.07.042] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 07/08/2020] [Accepted: 07/31/2020] [Indexed: 01/09/2023]
Abstract
OBJECTIVES The purpose of this study was to investigate the effect of empagliflozin on diastolic function in a nondiabetic heart failure with reduced ejection fraction (HFrEF) scenario and on the pathways causing diastolic dysfunction. BACKGROUND This group demonstrated that empagliflozin ameliorates adverse cardiac remodeling, enhances myocardial energetics, and improves left ventricular systolic function in a nondiabetic porcine model of HF. Whether empagliflozin also improves diastolic function remains unknown. Hypothetically, empagliflozin would improve diastolic function in HF mediated both by a reduction in interstitial myocardial fibrosis and an improvement in cardiomyocyte stiffness (titin phosphorylation). METHODS HF was induced in nondiabetic pigs by 2-h balloon occlusion of proximal left anterior descending artery. Animals were randomized to empagliflozin or placebo for 2 months. Cardiac function was evaluated with cardiac magnetic resonance (CMR), 3-dimensional echocardiography, and invasive hemodynamics. In vitro relaxation of cardiomyocytes was studied in primary culture. Myocardial samples were obtained for histological and molecular evaluation. Myocardial metabolite consumption was analyzed by simultaneous blood sampling from coronary artery and coronary sinus. RESULTS Despite similar initial ischemic myocardial injury, the empagliflozin group showed significantly improved diastolic function at 2 months, assessed by conventional echocardiography (higher e' and color M-mode propagation velocity, lower E/e' ratio, myocardial performance Tei index, isovolumic relaxation time, and left atrial size), echocardiography-derived strain imaging (strain imaging diastolic index, strain rate at isovolumic relaxation time and during early diastole, and untwisting), and CMR (higher peak filling rate, larger first filling volume). Invasive hemodynamics confirmed improved diastolic function with empagliflozin (better peak LV pressure rate of decay (-dP/dt), shorter Tau, lower end-diastolic pressure-volume relationship (EDPVR), and reduced filling pressures). Empagliflozin reduced interstitial myocardial fibrosis at the imaging, histological and molecular level. Empagliflozin improved nitric oxide signaling (endothelial nitric oxide synthetase [eNOS] activity, nitric oxide [NO] availability, cyclic guanosine monophosphate (cGMP) content, protein kinase G [PKG] signaling) and enhanced titin phosphorylation (which is responsible for cardiomyocyte stiffness). Indeed, isolated cardiomyocytes exhibited better relaxation in empagliflozin-treated animals. Myocardial consumption of glucose and ketone bodies negatively and positively correlated with diastolic function, respectively. CONCLUSIONS Empagliflozin ameliorates diastolic function in a nondiabetic HF porcine model, mitigates histological and molecular remodeling, and reduces both left ventricle and cardiomyocyte stiffness.
Collapse
Affiliation(s)
| | | | - Rodolfo San Antonio
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA
| | - Alvaro Garcia-Ropero
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA
| | - Kiyotake Ishikawa
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA
| | - Shin Watanabe
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA
| | - Belen Picatoste
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | - Javier Sanz
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA
| | - Valentin Fuster
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA
| | - Juan J Badimon
- Department of Cardiology, Mount Sinai School of Medicine, New York, New York, USA.
| |
Collapse
|
64
|
Scheffer M, Driessen-Waaijer A, Hamdani N, Landzaat JWD, Jonkman NH, Paulus WJ, van Heerebeek L. Stratified Treatment of Heart Failure with preserved Ejection Fraction: rationale and design of the STADIA-HFpEF trial. ESC Heart Fail 2020; 7:4478-4487. [PMID: 33073523 PMCID: PMC7754753 DOI: 10.1002/ehf2.13055] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/28/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022] Open
Abstract
AIMS High myocardial stiffness in heart failure with preserved ejection fraction (HFpEF) is attributed to comorbidity-induced structural and functional remodelling through inflammation and oxidative stress affecting coronary microvascular endothelial cells and cardiomyocytes, which augments interstitial fibrosis and cardiomyocyte stiffness. In murine and human HFpEF myocardium, sodium glucose co-transporter 2 (SGLT2) inhibition ameliorates cardiac microvascular endothelial cell and cardiomyocyte oxidative stress, while enhancing myocardial protein kinase G activity and lowering titin-based cardiomyocyte stiffness. Failure of previous HFpEF outcome trials refocuses attention to improving pathophysiological insight and trial design with better phenotyping of patients and matching of therapeutic targets to prevailing pathogenetic mechanisms. SGLT2 inhibition could represent a viable therapeutic option especially in HFpEF patients in whom high diastolic left ventricular (LV) stiffness is predominantly caused by elevated cardiomyocyte stiffness and associated endothelial dysfunction, whereas HFpEF patients with extensive myocardial fibrosis might be less responsive. This study aims to investigate a stratified treatment approach, using dapagliflozin in heart failure patients with preserved ejection fraction without evidence of significant myocardial fibrosis. METHODS AND RESULTS The Stratified Treatment to Ameliorate DIAstolic left ventricular stiffness in early Heart Failure with preserved Ejection Fraction (STADIA-HFpEF) is a Phase II, randomized, 2 × 2 crossover trial, evaluating the efficacy of 13 weeks of treatment with dapagliflozin 10 mg od in 26 patients with HFpEF, with normal cardiac magnetic resonance imaging-derived extracellular volume. The co-primary endpoint is echocardiographically derived change in E/e'/LV end-diastolic volume index and change in mean LV e'. CONCLUSIONS The STADIA-HFpEF trial will be the first study to evaluate the direct effects of dapagliflozin on amelioration of LV stiffness, using histological phenotyping to discern early HFpEF.
Collapse
Affiliation(s)
- Mariëlle Scheffer
- Department of Cardiology, OLVG, Oosterpark 9, Amsterdam, 1091 AC, The Netherlands
| | | | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St-Joseph Hospital, Ruhr University Bochum, Bochum, Germany
| | - Jochem W D Landzaat
- Department of Cardiology, OLVG, Oosterpark 9, Amsterdam, 1091 AC, The Netherlands
| | - Nini H Jonkman
- Department of Research and Epidemiology, OLVG, Amsterdam, The Netherlands
| | - Walter J Paulus
- Department of Physiology, Institute for Cardiovascular Research VU, VU University Medical Center Amsterdam, Amsterdam, The Netherlands
| | - Loek van Heerebeek
- Department of Cardiology, OLVG, Oosterpark 9, Amsterdam, 1091 AC, The Netherlands
| |
Collapse
|
65
|
Popova S, Ulanova A, Gritsyna Y, Salmov N, Rogachevsky V, Mikhailova G, Bobylev A, Bobyleva L, Yutskevich Y, Morenkov O, Zakharova N, Vikhlyantsev I. Predominant synthesis of giant myofibrillar proteins in striated muscles of the long-tailed ground squirrel Urocitellus undulatus during interbout arousal. Sci Rep 2020; 10:15185. [PMID: 32938992 PMCID: PMC7495002 DOI: 10.1038/s41598-020-72127-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/24/2020] [Indexed: 12/11/2022] Open
Abstract
Molecular mechanisms underlying muscle-mass retention during hibernation have been extensively discussed in recent years. This work tested the assumption that protein synthesis hyperactivation during interbout arousal of the long-tailed ground squirrel Urocitellus undulatus should be accompanied by increased calpain-1 activity in striated muscles. Calpain-1 is known to be autolysed and activated in parallel. Western blotting detected increased amounts of autolysed calpain-1 fragments in the heart (1.54-fold, p < 0.05) and m. longissimus dorsi (1.8-fold, p < 0.01) of ground squirrels during interbout arousal. The total protein synthesis rate determined by SUnSET declined 3.67-fold in the heart (p < 0.01) and 2.96-fold in m. longissimus dorsi (p < 0.01) during interbout arousal. The synthesis rates of calpain-1 substrates nebulin and titin in muscles did not differ during interbout arousal from those in active summer animals. A recovery of the volume of m. longissimus dorsi muscle fibres, a trend towards a heart-muscle mass increase and a restoration of the normal titin content (reduced in the muscles during hibernation) were observed. The results indicate that hyperactivation of calpain-1 in striated muscles of long-tailed ground squirrels during interbout arousal is accompanied by predominant synthesis of giant sarcomeric cytoskeleton proteins. These changes may contribute to muscle mass retention during hibernation.
Collapse
Affiliation(s)
- Svetlana Popova
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Anna Ulanova
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Yulia Gritsyna
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Nikolay Salmov
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Vadim Rogachevsky
- Laboratory of Signal Perception Mechanisms, Institute of Cell Biophysics, FRC PSCBR, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Gulnara Mikhailova
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Alexander Bobylev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Liya Bobyleva
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Yana Yutskevich
- Kuban State University, Krasnodar, Krasnodar Krai, 350040, Russia
| | - Oleg Morenkov
- Laboratory of Cell Culture and Tissue Engineering, Institute of Cell Biophysics, FRC PSCBR, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Nadezda Zakharova
- Laboratory of Natural and Artificial Hypobiosis Mechanisms, Institute of Cell Biophysics, FRC PSCBR, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Ivan Vikhlyantsev
- Laboratory of the Structure and Functions of Muscle Proteins, Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| |
Collapse
|
66
|
Change the Laminin, Change the Cardiomyocyte: Improve Untreatable Heart Failure. Int J Mol Sci 2020; 21:ijms21176013. [PMID: 32825544 PMCID: PMC7504464 DOI: 10.3390/ijms21176013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
No effective medical treatment exists for heart failure with preserved ejection fraction (HFpEF), accounting for approximately half of all heart failure cases. The elevated passive myocardial stiffness in HFpEF is attributed to a combination of alterations in the extracellular matrix (ECM) collagen content and modifications in the sarcomeric protein titin. Here, we propose polylaminin, a biomimetic polymer of laminin, as a promising approach for manipulating the titin isoform shift and phosphorylation in cardiomyocytes. Exploring the pleiotropic effects of polylaminin may be a novel strategy for alleviating symptoms in HFpEF's multifactorial pathophysiology.
Collapse
|
67
|
Nagueh SF. Heart failure with preserved ejection fraction: insights into diagnosis and pathophysiology. Cardiovasc Res 2020; 117:999-1014. [PMID: 32717061 DOI: 10.1093/cvr/cvaa228] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/15/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for at least half the cases of heart failure, currently diagnosed. There are several cardiac and non-cardiac manifestations of the syndrome. Structure and function abnormalities can include all four cardiac chambers. The left ventricle has abnormal systolic and diastolic functions which can be examined by invasive and non-invasive measurements. In addition, the left atrium enlarges with abnormal left atrial function, pulmonary hypertension occurs, and the right ventricle can develop hypertrophy, enlargement, and systolic dysfunction. There are a paucity of data on calcium handling in HFpEF patients. Growing literature supports the presence of abnormalities in titin and its phosphorylation, and increased interstitial fibrosis contributing to increased chamber stiffness. A systemic inflammatory state causing reduced myocardial cyclic guanosine monophosphate along with defects in the unfolded protein response have been recently reported. Diagnosis relies on signs and symptoms of heart failure, preserved ejection fraction, and detection of diastolic function abnormalities based on echocardiographic findings and abnormally elevated natriuretic peptide levels or invasive measurements of wedge pressure at rest or with exercise. There are currently two diagnostic algorithms: H2FPEF, and HFA-PEFF with limited data comparing their performance head to head in the same patient population. Despite the growing understanding of the syndrome's pathophysiology, there have been little success in developing specific treatment for patients with HFpEF.
Collapse
Affiliation(s)
- Sherif F Nagueh
- Methodist DeBakey Heart and Vascular Center, 6550 Fannin, SM-1801, Houston, TX 77030, USA
| |
Collapse
|
68
|
The molecular mechanisms associated with the physiological responses to inflammation and oxidative stress in cardiovascular diseases. Biophys Rev 2020; 12:947-968. [PMID: 32691301 PMCID: PMC7429613 DOI: 10.1007/s12551-020-00742-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
The complex physiological signal transduction networks that respond to the dual challenges of inflammatory and oxidative stress are major factors that promote the development of cardiovascular pathologies. These signaling networks contribute to the development of age-related diseases, suggesting crosstalk between the development of aging and cardiovascular disease. Inhibition and/or attenuation of these signaling networks also delays the onset of disease. Therefore, a concept of targeting the signaling networks that are involved in inflammation and oxidative stress may represent a novel treatment paradigm for many types of heart disease. In this review, we discuss the molecular mechanisms associated with the physiological responses to inflammation and oxidative stress especially in heart failure with preserved ejection fraction and emphasize the nature of the crosstalk of these signaling processes as well as possible therapeutic implications for cardiovascular medicine.
Collapse
|
69
|
Pagel PS, Tawil JN, Boettcher BT, Izquierdo DA, Lazicki TJ, Crystal GJ, Freed JK. Heart Failure With Preserved Ejection Fraction: A Comprehensive Review and Update of Diagnosis, Pathophysiology, Treatment, and Perioperative Implications. J Cardiothorac Vasc Anesth 2020; 35:1839-1859. [PMID: 32747202 DOI: 10.1053/j.jvca.2020.07.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023]
Abstract
Almost three-quarters of all heart failure patients who are older than 65 have heart failure with preserved ejection fraction (HFpEF). The proportion and hospitalization rate of patients with HFpEF are increasing steadily relative to patients in whom heart failure occurs as result of reduced ejection fraction. The predominance of the HFpEF phenotype most likely is explained by the prevalence of medical conditions associated with an aging population. A multitude of age-related, medical, and lifestyle risk factors for HFpEF have been identified as potential causes for the sustained low-grade proinflammatory state that accelerates disease progression. Profound left ventricular (LV) systolic and diastolic stiffening, elevated LV filling pressures, reduced arterial compliance, left atrial hypertension, pulmonary venous congestion, and microvascular dysfunction characterize HFpEF, but pulmonary arterial hypertension, right ventricular dilation and dysfunction, and atrial fibrillation also frequently occur. These cardiovascular features make patients with HFpEF exquisitely sensitive to the development of hypotension in response to acute declines in LV preload or afterload that may occur during or after surgery. With the exception of symptom mitigation, lifestyle modifications, and rigorous control of comorbid conditions, few long-term treatment options exist for these unfortunate individuals. Patients with HFpEF present for surgery on a regular basis, and anesthesiologists need to be familiar with this heterogeneous and complex clinical syndrome to provide successful care. In this article, the authors review the diagnosis, pathophysiology, and treatment of HFpEF and also discuss its perioperative implications.
Collapse
Affiliation(s)
- Paul S Pagel
- Anesthesia Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, WI.
| | - Justin N Tawil
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Brent T Boettcher
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - David A Izquierdo
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - Timothy J Lazicki
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| | - George J Crystal
- Department of Anesthesiology, University of Illinois College of Medicine, Chicago, IL
| | - Julie K Freed
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
70
|
Endothelial Dysfunction: A Contributor to Adverse Cardiovascular Remodeling and Heart Failure Development in Type 2 Diabetes beyond Accelerated Atherogenesis. J Clin Med 2020; 9:jcm9072090. [PMID: 32635218 PMCID: PMC7408687 DOI: 10.3390/jcm9072090] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction, associated with depressed nitric oxide (NO) bioavailability, is awell-recognized contributor to both accelerated atherogenesis and microvascular complications intype 2 diabetes (DM). However, growing evidence points to the comorbidities-driven endothelialdysfunction within coronary microvessels as a key player responsible for left ventricular (LV)diastolic dysfunction, restrictive LV remodeling and heart failure with preserved ejection fraction(HFpEF), the most common form of heart failure in DM. In this review we have described: (1)multiple cellular pathways which may link depressed NO bioavailability to LV diastolicdysfunction and hypertrophy; (2) hemodynamic consequences and prognostic effects of restrictiveLV remodeling and combined diastolic and mild systolic LV dysfunction on cardiovascularoutcomes in DM and HFpEF, with a focus on the clinical relevance of endothelial dysfunction; (3)novel therapeutic strategies to improve endothelial function in DM. In summary, beyondassociations with accelerated atherogenesis and microvascular complications, endothelialdysfunction supplements the multiple interwoven pathways affecting cardiomyocytes, endothelialcells and the extracellular matrix with consequent LV dysfunction in DM patients. The associationamongst impaired endothelial function, reduced coronary flow reserve, combined LV diastolic anddiscrete systolic dysfunction, and low LV stroke volume and preload reserve-all of which areadverse outcome predictors-is a dangerous constellation of inter-related abnormalities, underlyingthe development of heart failure. Nevertheless, the relevance of endothelial effects of novel drugsin terms of their ability to attenuate cardiovascular remodeling and delay heart failure onset in DMpatients remains to be investigated.
Collapse
|
71
|
A directed network analysis of the cardiome identifies molecular pathways contributing to the development of HFpEF. J Mol Cell Cardiol 2020; 144:66-75. [DOI: 10.1016/j.yjmcc.2020.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/08/2020] [Indexed: 01/06/2023]
|
72
|
Microvascular and lymphatic dysfunction in HFpEF and its associated comorbidities. Basic Res Cardiol 2020; 115:39. [PMID: 32451732 PMCID: PMC7248044 DOI: 10.1007/s00395-020-0798-y] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/13/2020] [Indexed: 02/07/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex heterogeneous disease for which our pathophysiological understanding is still limited and specific prevention and treatment strategies are lacking. HFpEF is characterised by diastolic dysfunction and cardiac remodelling (fibrosis, inflammation, and hypertrophy). Recently, microvascular dysfunction and chronic low-grade inflammation have been proposed to participate in HFpEF development. Furthermore, several recent studies demonstrated the occurrence of generalized lymphatic dysfunction in experimental models of risk factors for HFpEF, including obesity, hypercholesterolaemia, type 2 diabetes mellitus (T2DM), hypertension, and aging. Here, we review the evidence for a combined role of coronary (micro)vascular dysfunction and lymphatic vessel alterations in mediating key pathological steps in HFpEF, including reduced cardiac perfusion, chronic low-grade inflammation, and myocardial oedema, and their impact on cardiac metabolic alterations (oxygen and nutrient supply/demand imbalance), fibrosis, and cardiomyocyte stiffness. We focus primarily on HFpEF caused by metabolic risk factors, such as obesity, T2DM, hypertension, and aging.
Collapse
|
73
|
Kolijn D, Kovács Á, Herwig M, Lódi M, Sieme M, Alhaj A, Sandner P, Papp Z, Reusch PH, Haldenwang P, Falcão-Pires I, Linke WA, Jaquet K, Van Linthout S, Mügge A, Tschöpe C, Hamdani N. Enhanced Cardiomyocyte Function in Hypertensive Rats With Diastolic Dysfunction and Human Heart Failure Patients After Acute Treatment With Soluble Guanylyl Cyclase (sGC) Activator. Front Physiol 2020; 11:345. [PMID: 32523538 PMCID: PMC7261855 DOI: 10.3389/fphys.2020.00345] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/26/2020] [Indexed: 01/09/2023] Open
Abstract
Aims Our aim was to investigate the effect of nitric oxide (NO)-independent activation of soluble guanylyl cyclase (sGC) on cardiomyocyte function in a hypertensive animal model with diastolic dysfunction and in biopsies from human heart failure with preserved ejection fraction (HFpEF). Methods Dahl salt-sensitive (DSS) rats and control rats were fed a high-salt diet for 10 weeks and then acutely treated in vivo with the sGC activator BAY 58-2667 (cinaciguat) for 30 min. Single skinned cardiomyocyte passive stiffness (Fpassive) was determined in rats and human myocardium biopsies before and after acute treatment. Titin phosphorylation, activation of the NO/sGC/cyclic guanosine monophosphate (cGMP)/protein kinase G (PKG) cascade, as well as hypertrophic pathways including NO/sGC/cGMP/PKG, PKA, calcium–calmodulin kinase II (CaMKII), extracellular signal-regulated kinase 2 (ERK2), and PKC were assessed. In addition, we explored the contribution of pro-inflammatory cytokines and oxidative stress levels to the modulation of cardiomyocyte function. Immunohistochemistry and electron microscopy were used to assess the translocation of sGC and connexin 43 proteins in the rat model before and after treatment. Results High cardiomyocyte Fpassive was found in rats and human myocardial biopsies compared to control groups, which was attributed to hypophosphorylation of total titin and to deranged site-specific phosphorylation of elastic titin regions. This was accompanied by lower levels of PKG and PKA activity, along with dysregulation of hypertrophic pathway markers such as CaMKII, PKC, and ERK2. Furthermore, DSS rats and human myocardium biopsies showed higher pro-inflammatory cytokines and oxidative stress compared to controls. DSS animals benefited from treatment with the sGC activator, as Fpassive, titin phosphorylation, PKG and the hypertrophic pathway kinases, pro-inflammatory cytokines, and oxidative stress markers all significantly improved to the level observed in controls. Immunohistochemistry and electron microscopy revealed a translocation of sGC protein toward the intercalated disc and t-tubuli following treatment in both control and DSS samples. This translocation was confirmed by staining for the gap junction protein connexin 43 at the intercalated disk. DSS rats showed a disrupted connexin 43 pattern, and sGC activator was able to partially reduce disruption and increase expression of connexin 43. In human HFpEF biopsies, the high Fpassive, reduced titin phosphorylation, dysregulation of the NO–sGC–cGMP–PKG pathway and PKA activity level, and activity of kinases involved in hypertrophic pathways CaMKII, PKC, and ERK2 were all significantly improved by sGC treatment and accompanied by a reduction in pro-inflammatory cytokines and oxidative stress markers. Conclusion Our data show that sGC activator improves cardiomyocyte function, reduces inflammation and oxidative stress, improves sGC–PKG signaling, and normalizes hypertrophic kinases, indicating that it is a potential treatment option for HFpEF patients and perhaps also for cases with increased hypertrophic signaling.
Collapse
Affiliation(s)
- Detmar Kolijn
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Árpád Kovács
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Melissa Herwig
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Mária Lódi
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany.,Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,University of Debrecen, Kálmán Laki Doctoral School, Debrecen, Hungary
| | - Marcel Sieme
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Abdulatif Alhaj
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany
| | - Peter Sandner
- Bayer AG, Drug Discovery Cardiology, Wuppertal, Germany
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter H Reusch
- Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Peter Haldenwang
- Department of Cardiothoracic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Ines Falcão-Pires
- Department of Surgery and Physiology and Cardiovascular Research Centre, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, University of Münster, Münster, Germany
| | - Kornelia Jaquet
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany
| | - Sophie Van Linthout
- Department of Medicine and Cardiology (CVK), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Mügge
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany
| | - Carsten Tschöpe
- Department of Medicine and Cardiology (CVK), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
74
|
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
|
75
|
Herwig M, Kolijn D, Lódi M, Hölper S, Kovács Á, Papp Z, Jaquet K, Haldenwang P, Dos Remedios C, Reusch PH, Mügge A, Krüger M, Fielitz J, Linke WA, Hamdani N. Modulation of Titin-Based Stiffness in Hypertrophic Cardiomyopathy via Protein Kinase D. Front Physiol 2020; 11:240. [PMID: 32351396 PMCID: PMC7174613 DOI: 10.3389/fphys.2020.00240] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
The giant protein titin performs structure-preserving functions in the sarcomere and is important for the passive stiffness (Fpassive) of cardiomyocytes. Protein kinase D (PKD) enzymes play crucial roles in regulating myocardial contraction, hypertrophy, and remodeling. PKD phosphorylates myofilament proteins, but it is not known whether the giant protein titin is also a PKD substrate. Here, we aimed to determine whether PKD phosphorylates titin and thereby modulates cardiomyocyte Fpassive in normal and failing myocardium. The phosphorylation of titin was assessed in cardiomyocyte-specific PKD knock-out mice (cKO) and human hearts using immunoblotting with a phosphoserine/threonine and a phosphosite-specific titin antibody. PKD-dependent site-specific titin phosphorylation in vivo was quantified by mass spectrometry using stable isotope labeling by amino acids in cell culture (SILAC) of SILAC-labeled mouse heart protein lysates that were mixed with lysates isolated from hearts of either wild-type control (WT) or cKO mice. Fpassive of single permeabilized cardiomyocytes was recorded before and after PKD and HSP27 administration. All-titin phosphorylation was reduced in cKO compared to WT hearts. Multiple conserved PKD-dependent phosphosites were identified within the Z-disk, A-band and M-band regions of titin by quantitative mass spectrometry, and many PKD-dependent phosphosites detected in the elastic titin I-band region were significantly decreased in cKO. Analysis of titin site-specific phosphorylation showed unaltered or upregulated phosphorylation in cKO compared to matched WT hearts. Fpassive was elevated in cKO compared to WT cardiomyocytes and PKD administration lowered Fpassive of WT and cKO cardiomyocytes. Cardiomyocytes from hypertrophic cardiomyopathy (HCM) patients showed higher Fpassive compared to control hearts and significantly lower Fpassive after PKD treatment. In addition, we found higher phosphorylation at CaMKII-dependent titin sites in HCM compared to control hearts. Expression and phosphorylation of HSP27, a substrate of PKD, were elevated in HCM hearts, which was associated with increased PKD expression and phosphorylation. The relocalization of HSP27 in HCM away from the sarcomeric Z-disk and I-band suggested that HSP27 failed to exert its protective action on titin extensibility. This protection could, however, be restored by administration of HSP27, which significantly reduced Fpassive in HCM cardiomyocytes. These findings establish a previously unknown role for PKDin regulating diastolic passive properties of healthy and diseased hearts.
Collapse
Affiliation(s)
- Melissa Herwig
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Detmar Kolijn
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| | - Mária Lódi
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hungary
| | - Soraya Hölper
- Sanofi-Aventis Deutschland GmbH Industriepark Höchst, Frankfurt, Germany
| | - Árpád Kovács
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Zoltán Papp
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kornelia Jaquet
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Peter Haldenwang
- Department of Cardiothoracic Surgery, University Hospital Bergmannsheil Bochum, Bochum, Germany
| | - Cris Dos Remedios
- School of Medical Sciences, Bosch Institute, University of Sydney, Camperdown, NSW, Australia
| | - Peter H Reusch
- Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Andreas Mügge
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany
| | - Marcus Krüger
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Jens Fielitz
- Department of Internal Medicine B, Cardiology, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, University of Münster, Münster, Germany
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochums, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
76
|
Yousefi K, Irion CI, Takeuchi LM, Ding W, Lambert G, Eisenberg T, Sukkar S, Granzier HL, Methawasin M, Lee DI, Hahn VS, Kass DA, Hatzistergos KE, Hare JM, Webster KA, Shehadeh LA. Osteopontin Promotes Left Ventricular Diastolic Dysfunction Through a Mitochondrial Pathway. J Am Coll Cardiol 2020; 73:2705-2718. [PMID: 31146816 DOI: 10.1016/j.jacc.2019.02.074] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/25/2019] [Accepted: 02/26/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Patients with chronic kidney disease (CKD) and coincident heart failure with preserved ejection fraction (HFpEF) may constitute a distinct HFpEF phenotype. Osteopontin (OPN) is a biomarker of HFpEF and predictive of disease outcome. We recently reported that OPN blockade reversed hypertension, mitochondrial dysfunction, and kidney failure in Col4a3-/- mice, a model of human Alport syndrome. OBJECTIVES The purpose of this study was to identify potential OPN targets in biopsies of HF patients, healthy control subjects, and human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs), and to characterize the cardiac phenotype of Col4a3-/- mice, relate this to HFpEF, and investigate possible causative roles for OPN in driving the cardiomyopathy. METHODS OGDHL mRNA and protein were quantified in myocardial samples from patients with HFpEF, heart failure with reduced ejection fraction, and donor control subjects. OGDHL expression was quantified in hiPS-CMs treated with or without anti-OPN antibody. Cardiac parameters were evaluated in Col4a3-/- mice with and without global OPN knockout or AAV9-mediated delivery of 2-oxoglutarate dehydrogenase-like (Ogdhl) to the heart. RESULTS OGDHL mRNA and protein displayed abnormal abundances in cardiac biopsies of HFpEF (n = 17) compared with donor control subjects (n = 12; p < 0.01) or heart failure with reduced ejection fraction patients (n = 12; p < 0.05). Blockade of OPN in hiPS-CMs conferred increased OGDHL expression. Col4a3-/- mice demonstrated cardiomyopathy with similarities to HFpEF, including diastolic dysfunction, cardiac hypertrophy and fibrosis, pulmonary edema, and impaired mitochondrial function. The cardiomyopathy was ameliorated by Opn-/- coincident with improved renal function and increased expression of Ogdhl. Heart-specific overexpression of Ogdhl in Col4a3-/- mice also improved cardiac function and cardiomyocyte energy state. CONCLUSIONS Col4a3-/- mice present a model of HFpEF secondary to CKD wherein OPN and OGDHL are intermediates, and possibly therapeutic targets.
Collapse
Affiliation(s)
- Keyvan Yousefi
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Camila I Irion
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Wen Ding
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Guerline Lambert
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Trevor Eisenberg
- Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Sarah Sukkar
- Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Henk L Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Dong I Lee
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Konstantinos E Hatzistergos
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Department of Cell Biology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Keith A Webster
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Vascular Biology Institute and Peggy and Harold Katz Family Drug Discovery Center, University of Miami Leonard M. Miller School of Medicine, Miami, Florida
| | - Lina A Shehadeh
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Department of Medicine, Division of Cardiology, University of Miami Leonard M. Miller School of Medicine, Miami, Florida; Vascular Biology Institute and Peggy and Harold Katz Family Drug Discovery Center, University of Miami Leonard M. Miller School of Medicine, Miami, Florida.
| |
Collapse
|
77
|
Blair CA, Pruitt BL. Mechanobiology Assays with Applications in Cardiomyocyte Biology and Cardiotoxicity. Adv Healthc Mater 2020; 9:e1901656. [PMID: 32270928 PMCID: PMC7480481 DOI: 10.1002/adhm.201901656] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/19/2022]
Abstract
Cardiomyocytes are the motor units that drive the contraction and relaxation of the heart. Traditionally, testing of drugs for cardiotoxic effects has relied on primary cardiomyocytes from animal models and focused on short-term, electrophysiological, and arrhythmogenic effects. However, primary cardiomyocytes present challenges arising from their limited viability in culture, and tissue from animal models suffers from a mismatch in their physiology to that of human heart muscle. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can address these challenges. They also offer the potential to study not only electrophysiological effects but also changes in cardiomyocyte contractile and mechanical function in response to cardiotoxic drugs. With growing recognition of the long-term cardiotoxic effects of some drugs on subcellular structure and function, there is increasing interest in using hiPSC-CMs for in vitro cardiotoxicity studies. This review provides a brief overview of techniques that can be used to quantify changes in the active force that cardiomyocytes generate and variations in their inherent stiffness in response to cardiotoxic drugs. It concludes by discussing the application of these tools in understanding how cardiotoxic drugs directly impact the mechanobiology of cardiomyocytes and how cardiomyocytes sense and respond to mechanical load at the cellular level.
Collapse
Affiliation(s)
- Cheavar A. Blair
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Beth L. Pruitt
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
78
|
Ward M, Iskratsch T. Mix and (mis-)match - The mechanosensing machinery in the changing environment of the developing, healthy adult and diseased heart. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118436. [PMID: 30742931 PMCID: PMC7042712 DOI: 10.1016/j.bbamcr.2019.01.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
The composition and the stiffness of cardiac microenvironment change during development and/or in heart disease. Cardiomyocytes (CMs) and their progenitors sense these changes, which decides over the cell fate and can trigger CM (progenitor) proliferation, differentiation, de-differentiation or death. The field of mechanobiology has seen a constant increase in output that also includes a wealth of new studies specific to cardiac or cardiomyocyte mechanosensing. As a result, mechanosensing and transduction in the heart is increasingly being recognised as a main driver of regulating the heart formation and function. Recent work has for instance focused on measuring the molecular, physical and mechanical changes of the cellular environment - as well as intracellular contributors to the passive stiffness of the heart. On the other hand, a variety of new studies shed light into the molecular machinery that allow the cardiomyocytes to sense these properties. Here we want to discuss the recent work on this topic, but also specifically focus on how the different components are regulated at various stages during development, in health or disease in order to highlight changes that might contribute to disease progression and heart failure.
Collapse
Key Words
- cm, cardiomyocytes
- hcm, hypertrophic cardiomyopathy
- dcm, dilated cardiomyopathy
- icm, idiopathic cardiomyopathy
- myh, myosin heavy chain
- tnnt, troponin t
- tnni, troponin i
- afm, atomic force microscope
- mre, magnetic resonance elastography
- swe, ultrasound cardiac shear-wave elastography
- lv, left ventricle
- lox, lysyl oxidase
- loxl, lysyl oxidase like protein
- lh, lysyl hydroxylase
- lys, lysin
- lccs, lysald-derived collagen crosslinks
- hlccs, hylald-derived collagen crosslinks
- pka, protein kinase a
- pkc, protein kinase c
- vash1, vasohibin-1
- svbp, small vasohibin binding protein
- tcp, tubulin carboxypeptidase
- ttl, tubulin tyrosine ligase
- mrtf, myocardin-related transcription factor
- gap, gtpase activating protein
- gef, guanine nucleotide exchange factor
Collapse
Affiliation(s)
- Matthew Ward
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom.
| |
Collapse
|
79
|
Pitoulis FG, Terracciano CM. Heart Plasticity in Response to Pressure- and Volume-Overload: A Review of Findings in Compensated and Decompensated Phenotypes. Front Physiol 2020; 11:92. [PMID: 32116796 PMCID: PMC7031419 DOI: 10.3389/fphys.2020.00092] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
The adult human heart has an exceptional ability to alter its phenotype to adapt to changes in environmental demand. This response involves metabolic, mechanical, electrical, and structural alterations, and is known as cardiac plasticity. Understanding the drivers of cardiac plasticity is essential for development of therapeutic agents. This is particularly important in contemporary cardiology, which uses treatments with peripheral effects (e.g., on kidneys, adrenal glands). This review focuses on the effects of different hemodynamic loads on myocardial phenotype. We examine mechanical scenarios of pressure- and volume overload, from the initial insult, to compensated, and ultimately decompensated stage. We discuss how different hemodynamic conditions occur and are underlined by distinct phenotypic and molecular changes. We complete the review by exploring how current basic cardiac research should leverage available cardiac models to study mechanical load in its different presentations.
Collapse
|
80
|
Chung CS, Hiske MA, Chadha A, Mueller PJ. Compliant Titin Isoform Content Is Reduced in Left Ventricles of Sedentary Versus Active Rats. Front Physiol 2020; 11:15. [PMID: 32116740 PMCID: PMC7025574 DOI: 10.3389/fphys.2020.00015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/13/2020] [Indexed: 11/30/2022] Open
Abstract
A sedentary lifestyle is associated with increased cardiovascular risk factors and reduced cardiac compliance when compared to a lifestyle that includes exercise training. Exercise training increases cardiac compliance in humans, but the mechanisms underlying this improvement are unknown. A major determinant of cardiac compliance is the compliance of the giant elastic protein titin. Experimentally reducing titin compliance in animal models reduces exercise tolerance, but it is not known whether sedentary versus chronic exercise conditions cause differences in titin isoform content. We hypothesized that sedentary conditions would be associated with a reduction in the content of the longer, more compliant N2BA isoform relative to the stiffer N2B isoform (yielding a reduced N2BA:N2B ratio) compared to age-matched exercising controls. We obtained left ventricles from 16-week old rats housed for 12 weeks in standard (sedentary) or voluntary running wheel (exercised) housing. The N2BA:N2B ratio was decreased in the hearts of sedentary versus active rats (p = 0.041). Gene expression of a titin mRNA splicing factor, RNA Binding Motif 20 protein (RBM20), correlated negatively with N2BA:N2B ratios (p = 0.006, r = -0.449), but was not different between groups, suggesting that RBM20 may be regulated post-transcriptionally. Total phosphorylation of cardiac titin was not different between the active and sedentary groups. This study is the first to demonstrate that sedentary rats exhibit reduced cardiac titin N2BA:N2B isoform ratios, which implies reduced cardiac compliance. These data suggest that a lack of exercise (running wheel) reduces cardiac compliance and that exercise itself increases cardiac compliance.
Collapse
|
81
|
Henning RJ. Diagnosis and treatment of heart failure with preserved left ventricular ejection fraction. World J Cardiol 2020; 12:7-25. [PMID: 31984124 PMCID: PMC6952725 DOI: 10.4330/wjc.v12.i1.7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/17/2019] [Accepted: 11/26/2019] [Indexed: 02/07/2023] Open
Abstract
Nearly six million people in United States have heart failure. Fifty percent of these people have normal left ventricular (LV) systolic heart function but abnormal diastolic function due to increased LV myocardial stiffness. Most commonly, these patients are elderly women with hypertension, ischemic heart disease, atrial fibrillation, obesity, diabetes mellitus, renal disease, or obstructive lung disease. The annual mortality rate of these patients is 8%-12% per year. The diagnosis is based on the history, physical examination, laboratory data, echocardiography, and, when necessary, by cardiac catheterization. Patients with obesity, hypertension, atrial fibrillation, and volume overload require weight reduction, an exercise program, aggressive control of blood pressure and heart rate, and diuretics. Miniature devices inserted into patients for pulmonary artery pressure monitoring provide early warning of increased pulmonary pressure and congestion. If significant coronary heart disease is present, coronary revascularization should be considered.
Collapse
Affiliation(s)
- Robert J Henning
- College of Public Health, University of South Florida, Tampa, FL33612, United States
| |
Collapse
|
82
|
Krueger W, Bender N, Haeusler M, Henneberg M. The role of mechanotransduction in heart failure pathobiology-a concise review. Heart Fail Rev 2020; 26:981-995. [PMID: 31965473 DOI: 10.1007/s10741-020-09915-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
This review evaluates the role of mechanotransduction (MT) in heart failure (HF) pathobiology. Cardiac functional and structural modifications are regulated by biomechanical forces. Exposing cardiomyocytes and the myocardial tissue to altered biomechanical stress precipitates changes in the end-diastolic wall stress (EDWS). Thereby various interconnected biomolecular pathways, essentially mediated and orchestrated by MT, are launched and jointly contribute to adapt and remodel the myocardium. This cardiac MT-mediated feedback decisively determines the primary cardiac cellular and tissue response, the sort (concentric or eccentric) of hypertrophy/remodeling, to mechanical and/or hemodynamic alterations. Moreover, the altered EDWS affects the diastolic myocardial properties independent of the systolic function, and elevated EDWS causes diastolic dysfunction. The close interconnection between MT pathways and the cell nucleus, the genetic endowment, principally allows for the wide variety of phenotypic appearances. However, demographic, environmental features, comorbidities, and also the genetic make-up may modulate the phenotypic result. Cardiac MT takes a fundamental and superordinate position in the myocardial adaptation and remodeling processes in all HF categories and phenotypes. Therefore, the effects of MT should be integrated in all our scientific, clinical, and therapeutic considerations.
Collapse
Affiliation(s)
- Wolfgang Krueger
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland. .,Medical University Department, Kantonsspital Aarau, Aarau, Switzerland.
| | - Nicole Bender
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Martin Haeusler
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland
| | - Maciej Henneberg
- Institute of Evolutionary Medicine, University of Zurich, Zurich, Switzerland.,Adelaide Medical School, The University of Adelaide, Adelaide, Australia
| |
Collapse
|
83
|
Cellular and Molecular Differences between HFpEF and HFrEF: A Step Ahead in an Improved Pathological Understanding. Cells 2020; 9:cells9010242. [PMID: 31963679 PMCID: PMC7016826 DOI: 10.3390/cells9010242] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/15/2020] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
Heart failure (HF) is the most rapidly growing cardiovascular health burden worldwide. HF can be classified into three groups based on the percentage of the ejection fraction (EF): heart failure with reduced EF (HFrEF), heart failure with mid-range-also called mildly reduced EF- (HFmrEF), and heart failure with preserved ejection fraction (HFpEF). HFmrEF can progress into either HFrEF or HFpEF, but its phenotype is dominated by coronary artery disease, as in HFrEF. HFrEF and HFpEF present with differences in both the development and progression of the disease secondary to changes at the cellular and molecular level. While recent medical advances have resulted in efficient and specific treatments for HFrEF, these treatments lack efficacy for HFpEF management. These differential response rates, coupled to increasing rates of HF, highlight the significant need to understand the unique pathogenesis of HFrEF and HFpEF. In this review, we summarize the differences in pathological development of HFrEF and HFpEF, focussing on disease-specific aspects of inflammation and endothelial function, cardiomyocyte hypertrophy and death, alterations in the giant spring titin, and fibrosis. We highlight the areas of difference between the two diseases with the aim of guiding research efforts for novel therapeutics in HFrEF and HFpEF.
Collapse
|
84
|
Cauwenberghs N, Ravassa S, Thijs L, Haddad F, Yang WY, Wei FF, López B, González A, Díez J, Staessen JA, Kuznetsova T. Circulating Biomarkers Predicting Longitudinal Changes in Left Ventricular Structure and Function in a General Population. J Am Heart Assoc 2020; 8:e010430. [PMID: 30638123 PMCID: PMC6497333 DOI: 10.1161/jaha.118.010430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Serial imaging studies in the general population remain important to evaluate the usefulness of pathophysiologically relevant biomarkers in predicting progression of left ventricular (LV) remodeling and dysfunction. Here, we assessed in a general population whether these circulating biomarkers at baseline predict longitudinal changes in LV structure and function. Methods and Results In 592 participants (mean age, 50.8 years; 51.4% women; 40.5% hypertensive), we derived echocardiographic indexes reflecting LV structure and function at baseline and after 4.7 years. At baseline, we measured alkaline phosphatase, markers of collagen turnover (procollagen type I, C-terminal telopeptide, matrix metalloproteinase-1) and high-sensitivity cardiac troponin T. We regressed longitudinal changes in LV indexes on baseline biomarker levels and reported standardized effect sizes as a fraction of the standard deviation of LV change. After full adjustment, a decline in LV longitudinal strain (-14.2%) and increase in E/e' ratio over time (+18.9%; P≤0.019) was associated with higher alkaline phosphatase activity at baseline. Furthermore, longitudinal strain decreased with higher levels of collagen I production and degradation at baseline (procollagen type I, -14.2%; C-terminal telopeptide, -16.4%; P≤0.029). An increase in E/e' ratio over time was borderline associated with lower matrix metalloproteinase-1 (+9.8%) and lower matrix metalloproteinase-1/tissue inhibitor of metalloproteinase-1 ratio (+11.9%; P≤0.041). Higher high-sensitivity cardiac troponin T levels at baseline correlated significantly with an increase in relative wall thickness (+23.1%) and LV mass index (+18.3%) during follow-up ( P≤0.035). Conclusions We identified a set of biomarkers predicting adverse changes in LV structure and function over time. Circulating biomarkers reflecting LV stiffness, injury, and collagen composition might improve the identification of subjects at risk for subclinical cardiac maladaptation.
Collapse
Affiliation(s)
- Nicholas Cauwenberghs
- 1 Research Unit Hypertension and Cardiovascular Epidemiology KU Leuven Department of Cardiovascular Sciences University of Leuven Belgium
| | - Susana Ravassa
- 2 Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra Pamplona Spain.,3 IdiSNA Navarra Institute for Health Research Pamplona Spain.,4 CIBERCV Carlos III Institute of Health Madrid Spain
| | - Lutgarde Thijs
- 1 Research Unit Hypertension and Cardiovascular Epidemiology KU Leuven Department of Cardiovascular Sciences University of Leuven Belgium
| | - Francois Haddad
- 5 Division of Cardiovascular Medicine Stanford University School of Medicine and Stanford Cardiovascular Institute Stanford CA
| | - Wen-Yi Yang
- 1 Research Unit Hypertension and Cardiovascular Epidemiology KU Leuven Department of Cardiovascular Sciences University of Leuven Belgium
| | - Fang-Fei Wei
- 1 Research Unit Hypertension and Cardiovascular Epidemiology KU Leuven Department of Cardiovascular Sciences University of Leuven Belgium
| | - Begoña López
- 2 Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra Pamplona Spain.,3 IdiSNA Navarra Institute for Health Research Pamplona Spain.,4 CIBERCV Carlos III Institute of Health Madrid Spain
| | - Arantxa González
- 2 Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra Pamplona Spain.,3 IdiSNA Navarra Institute for Health Research Pamplona Spain.,4 CIBERCV Carlos III Institute of Health Madrid Spain
| | - Javier Díez
- 2 Program of Cardiovascular Diseases Centre for Applied Medical Research University of Navarra Pamplona Spain.,3 IdiSNA Navarra Institute for Health Research Pamplona Spain.,4 CIBERCV Carlos III Institute of Health Madrid Spain.,6 Department of Cardiology and Cardiac Surgery University of Navarra Pamplona Spain.,7 Department of Nephrology University of Navarra Pamplona Spain
| | - Jan A Staessen
- 1 Research Unit Hypertension and Cardiovascular Epidemiology KU Leuven Department of Cardiovascular Sciences University of Leuven Belgium
| | - Tatiana Kuznetsova
- 1 Research Unit Hypertension and Cardiovascular Epidemiology KU Leuven Department of Cardiovascular Sciences University of Leuven Belgium
| |
Collapse
|
85
|
Lazar S, Rayner B, Lopez Campos G, McGrath K, McClements L. Mechanisms of heart failure with preserved ejection fraction in the presence of diabetes mellitus. TRANSLATIONAL METABOLIC SYNDROME RESEARCH 2020. [DOI: 10.1016/j.tmsr.2020.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
86
|
Tharp CA, Haywood ME, Sbaizero O, Taylor MRG, Mestroni L. The Giant Protein Titin's Role in Cardiomyopathy: Genetic, Transcriptional, and Post-translational Modifications of TTN and Their Contribution to Cardiac Disease. Front Physiol 2019; 10:1436. [PMID: 31849696 PMCID: PMC6892752 DOI: 10.3389/fphys.2019.01436] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a leading cause of heart failure, sudden cardiac death and heart transplant. DCM is inherited in approximately 50% of cases, in which the most frequent genetic defects are truncation variants of the titin gene (TTNtv). TTN encodes titin, which is the largest protein in the body and is an essential component of the sarcomere. Titin serves as a biological spring, spanning half of the sarcomere and connecting the Z-disk to the M-line, with scaffold and signaling functions. Truncations of titin are believed to lead to either haploinsufficiency and loss-of-function, or to a “poison peptide” effect. However, other titin mechanisms are postulated to influence cardiac function including post-translational modifications, in particular changes in titin phosphorylation that alters the stiffness of the protein, and diversity of alternative splicing that generates different titin isoforms. In this article, we review the role of TTN mutations in development of DCM, how differential expression of titin isoforms relate to DCM pathophysiology, and discuss how post-translational modifications of titin can affect cardiomyocyte function. Current research efforts aim to elucidate the contribution of titin to myofibril assembly, stability, and signal transduction, and how mutant titin leads to cardiac dysfunction and human disease. Future research will need to translate this knowledge toward novel therapeutic approaches that can modulate titin transcriptional and post-translational defects to treat DCM and heart failure.
Collapse
Affiliation(s)
- Charles A Tharp
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Mary E Haywood
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Trieste, Italy
| | - Matthew R G Taylor
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Luisa Mestroni
- Adult Medical Genetics Program and Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
87
|
Huttner IG, Wang LW, Santiago CF, Horvat C, Johnson R, Cheng D, von Frieling-Salewsky M, Hillcoat K, Bemand TJ, Trivedi G, Braet F, Hesselson D, Alford K, Hayward CS, Seidman JG, Seidman CE, Feneley MP, Linke WA, Fatkin D. A-Band Titin Truncation in Zebrafish Causes Dilated Cardiomyopathy and Hemodynamic Stress Intolerance. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e002135. [PMID: 30354343 DOI: 10.1161/circgen.118.002135] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Truncating variants in the TTN gene ( TTNtv) are common in patients with dilated cardiomyopathy (DCM) but also occur in the general population. Whether TTNtv are sufficient to cause DCM or require a second hit for DCM manifestation is an important clinical issue. Methods We generated a zebrafish model of an A-band TTNtv identified in 2 human DCM families in which early-onset disease appeared to be precipitated by ventricular volume overload. Cardiac phenotypes were serially assessed from 0 to 12 months using video microscopy, high-frequency echocardiography, and histopathologic analysis. The effects of sustained hemodynamic stress resulting from an anemia-induced hyperdynamic state were also evaluated. Results Homozygous ttna mutants had severe cardiac dysmorphogenesis and premature death, whereas heterozygous mutants ( ttnatv/+) survived into adulthood and spontaneously developed DCM. Six-month-old ttnatv/+ fish had reduced baseline ventricular systolic function and failed to mount a hypercontractile response when challenged by hemodynamic stress. Pulsed wave and tissue Doppler analysis also revealed unsuspected ventricular diastolic dysfunction in ttnatv/+ fish with prolonged isovolumic relaxation and increased diastolic passive stiffness in the absence of myocardial fibrosis. These defects reduced diastolic reserve under stress conditions and resulted in disproportionately greater atrial dilation than observed in wild-type fish. Conclusions Heterozygosity for A-band titin truncation is sufficient to cause DCM in adult zebrafish. Abnormalities of systolic and diastolic reserve in titin-truncated fish reduce stress tolerance and may contribute to a substrate for atrial arrhythmogenesis. These data suggest that hemodynamic stress may be an important modifiable risk factor in human TTNtv-related DCM.
Collapse
Affiliation(s)
- Inken G Huttner
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.).,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington (I.G.H., L.W.W., C.F.S., D.H., C.S.H., M.P.F., D.F.)
| | - Louis W Wang
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.).,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington (I.G.H., L.W.W., C.F.S., D.H., C.S.H., M.P.F., D.F.).,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia (L.W.W., C.S.H., M.P.F., D.F.)
| | - Celine F Santiago
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.).,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington (I.G.H., L.W.W., C.F.S., D.H., C.S.H., M.P.F., D.F.)
| | - Claire Horvat
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.)
| | - Renee Johnson
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.)
| | - Delfine Cheng
- School of Medical Sciences, Bosch Institute, University of Sydney, Camperdown, NSW, Australia (D.C., F.B.)
| | | | - Karen Hillcoat
- Kevin Alford Cardiology, Port Macquarie, NSW Australia (K.H., K.A.)
| | - Timothy J Bemand
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.)
| | - Gunjan Trivedi
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.)
| | - Filip Braet
- School of Medical Sciences, Bosch Institute, University of Sydney, Camperdown, NSW, Australia (D.C., F.B.).,Cellular Imaging Facility, Charles Perkins Centre (F.B.).,Australian Centre for Microscopy and Microanalysis (F.B.)
| | - Dan Hesselson
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington (I.G.H., L.W.W., C.F.S., D.H., C.S.H., M.P.F., D.F.).,University of Sydney, Camperdown, NSW, Australia. Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia (D.H.)
| | - Kevin Alford
- Kevin Alford Cardiology, Port Macquarie, NSW Australia (K.H., K.A.)
| | - Christopher S Hayward
- Cardiac Physiology and Transplantation Division (C.S.H., M.P.F.).,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington (I.G.H., L.W.W., C.F.S., D.H., C.S.H., M.P.F., D.F.).,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia (L.W.W., C.S.H., M.P.F., D.F.)
| | - J G Seidman
- Howard Hughes Medical Institute, MD (J.G.S.).,Department of Genetics, Harvard Medical School (J.G.S., C.E.S.)
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School (J.G.S., C.E.S.).,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA (C.E.S.)
| | - Michael P Feneley
- Cardiac Physiology and Transplantation Division (C.S.H., M.P.F.).,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington (I.G.H., L.W.W., C.F.S., D.H., C.S.H., M.P.F., D.F.).,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia (L.W.W., C.S.H., M.P.F., D.F.)
| | - Wolfgang A Linke
- Institute of Physiology II, University of Muenster, Germany (M.v.F.-S., W.A.L.)
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division (I.G.H., L.W.W., C.F.S., C.H., R.J., T.J.B., G.T., D.F.).,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia. St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington (I.G.H., L.W.W., C.F.S., D.H., C.S.H., M.P.F., D.F.).,Cardiology Department, St Vincent's Hospital, Darlinghurst, NSW, Australia (L.W.W., C.S.H., M.P.F., D.F.)
| |
Collapse
|
88
|
Jeong MY, Lin YH, Wennersten SA, Demos-Davies KM, Cavasin MA, Mahaffey JH, Monzani V, Saripalli C, Mascagni P, Reece TB, Ambardekar AV, Granzier HL, Dinarello CA, McKinsey TA. Histone deacetylase activity governs diastolic dysfunction through a nongenomic mechanism. Sci Transl Med 2019; 10:10/427/eaao0144. [PMID: 29437146 DOI: 10.1126/scitranslmed.aao0144] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/07/2017] [Accepted: 01/05/2018] [Indexed: 12/21/2022]
Abstract
There are no approved drugs for the treatment of heart failure with preserved ejection fraction (HFpEF), which is characterized by left ventricular (LV) diastolic dysfunction. We demonstrate that ITF2357 (givinostat), a clinical-stage inhibitor of histone deacetylase (HDAC) catalytic activity, is efficacious in two distinct murine models of diastolic dysfunction with preserved EF. ITF2357 blocked LV diastolic dysfunction due to hypertension in Dahl salt-sensitive (DSS) rats and suppressed aging-induced diastolic dysfunction in normotensive mice. HDAC inhibitor-mediated efficacy was not due to lowering blood pressure or inhibiting cellular and molecular events commonly associated with diastolic dysfunction, including cardiac fibrosis, cardiac hypertrophy, or changes in cardiac titin and myosin isoform expression. Instead, ex vivo studies revealed impairment of cardiac myofibril relaxation as a previously unrecognized, myocyte-autonomous mechanism for diastolic dysfunction, which can be ameliorated by HDAC inhibition. Translating these findings to humans, cardiac myofibrils from patients with diastolic dysfunction and preserved EF also exhibited compromised relaxation. These data suggest that agents such as HDAC inhibitors, which potentiate cardiac myofibril relaxation, hold promise for the treatment of HFpEF in humans.
Collapse
Affiliation(s)
- Mark Y Jeong
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ying H Lin
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sara A Wennersten
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kimberly M Demos-Davies
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Maria A Cavasin
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jennifer H Mahaffey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | - Chandrasekhar Saripalli
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85724, USA
| | | | - T Brett Reece
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Amrut V Ambardekar
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Henk L Granzier
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85724, USA
| | - Charles A Dinarello
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy A McKinsey
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA. .,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
89
|
Ulanova A, Gritsyna Y, Salmov N, Lomonosova Y, Belova S, Nemirovskaya T, Shenkman B, Vikhlyantsev I. Effect of L-Arginine on Titin Expression in Rat Soleus Muscle After Hindlimb Unloading. Front Physiol 2019; 10:1221. [PMID: 31616317 PMCID: PMC6764413 DOI: 10.3389/fphys.2019.01221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 09/06/2019] [Indexed: 01/18/2023] Open
Abstract
Nitric oxide (NO), produced by NO-synthases via L-arginine oxidation, is an essential trigger for signaling processes involved in structural and metabolic changes in muscle fibers. Recently, it was shown that L-arginine administration prevented the decrease in levels of the muscle cytoskeletal proteins, desmin and dystrophin, in rat soleus muscle after 14 days of hindlimb unloading. Therefore, in this study, we investigated the effect of L-arginine administration on the degree of atrophy changes in the rat soleus muscles under unloading conditions, and on the content, gene expression, and phosphorylation level of titin, the giant protein of striated muscles, able to form a third type of myofilaments—elastic filaments. A 7-day gravitational unloading [hindlimb suspension (HS) group] resulted in a decrease in the soleus weight:body weight ratio (by 31.8%, p < 0.05), indicating muscle atrophy development. The content of intact titin (T1) decreased (by 22.4%, p < 0.05) and the content of proteolytic fragments of titin (T2) increased (by 66.7%, p < 0.05) in the soleus muscle of HS rats, compared to control rats. The titin gene expression and phosphorylation level of titin between these two groups were not significantly different. L-Arginine administration under 7-day gravitational unloading decreased the degree of atrophy changes and also prevented the decrease in levels of T1 in the soleus muscle as compared to HS group. Furthermore, L-arginine administration under unloading resulted in increased titin mRNA level (by 76%, p < 0.05) and decreased phosphorylation level of T2 (by 28%, p < 0.05), compared to those in the HS group. These results suggest that administration of L-arginine, the NO precursor, under unloading decreased the degree of atrophy changes, increased gene expression of titin and prevented the decrease in levels of T1 in the rat soleus muscle. The results can be used to search for approaches to reduce the development of negative changes caused by gravitational unloading in the muscle.
Collapse
Affiliation(s)
- Anna Ulanova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia.,Pushchino State Institute of Natural Sciences, Pushchino, Russia
| | - Yuliya Gritsyna
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Nikolai Salmov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Yuliya Lomonosova
- State Scientific Center RF, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Svetlana Belova
- State Scientific Center RF, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Tatyana Nemirovskaya
- State Scientific Center RF, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Boris Shenkman
- State Scientific Center RF, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Ivan Vikhlyantsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| |
Collapse
|
90
|
Abstract
Hypertensive heart disease represents a spectrum of illnesses from uncontrolled hypertension to heart failure. The authors discuss the natural history and pathogenesis of heart failure owing to hypertensive heart disease, reviewing the important role of left ventricular hypertrophy as the inciting process leading to diastolic dysfunction and heart failure with preserved ejection fraction. They describe the various mechanisms by which a subset of patients ultimately develops systolic heart failure. They discuss management strategies for hypertensive heart disease at all stages of the disease process. Treatment in the initial stages before onset of heart failure may result in regression of disease.
Collapse
Affiliation(s)
- Jeremy Slivnick
- Ohio State University Wexner Medical Center, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA
| | - Brent C Lampert
- Heart Transplantation and Mechanical Circulatory Support, Ohio State University Wexner Medical Center, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA.
| |
Collapse
|
91
|
Najafi A, van de Locht M, Schuldt M, Schönleitner P, van Willigenburg M, Bollen I, Goebel M, Ottenheijm CAC, van der Velden J, Helmes M, Kuster DWD. End-diastolic force pre-activates cardiomyocytes and determines contractile force: role of titin and calcium. J Physiol 2019; 597:4521-4531. [PMID: 31314138 PMCID: PMC6852589 DOI: 10.1113/jp277985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/10/2019] [Indexed: 12/22/2022] Open
Abstract
Titin functions as a molecular spring, and cardiomyocytes are able, through splicing, to control the length of titin. We hypothesized that together with diastolic [Ca2+], titin‐based stretch pre‐activates cardiomyocytes during diastole and is a major determinant of force production in the subsequent contraction. Through this mechanism titin would play an important role in active force development and length‐dependent activation. Mutations in the splicing factor RNA binding motif protein 20 (RBM20) result in expression of large, highly compliant titin isoforms. We measured single cardiomyocyte work loops that mimic the cardiac cycle in wild‐type (WT) and heterozygous (HET) RBM20‐deficient rats. In addition, we studied the role of diastolic [Ca2+] in membrane‐permeabilized WT and HET cardiomyocytes. Intact cardiomyocytes isolated from HET left ventricles were unable to produce normal levels of work (55% of WT) at low pacing frequencies, but this difference disappeared at high pacing frequencies. Length‐dependent activation (force–sarcomere length relationship) was blunted in HET cardiomyocytes, but the force–end‐diastolic force relationship was not different between HET and WT cardiomyocytes. To delineate the effects of diastolic [Ca2+] and titin pre‐activation on force generation, measurements were performed in detergent‐permeabilized cardiomyocytes. Cardiac twitches were simulated by transiently exposing permeabilized cardiomyocytes to 2 µm Ca2+. Increasing diastolic [Ca2+] from 1 to 80 nm increased force development twofold in WT. Higher diastolic [Ca2+] was needed in HET. These findings are consistent with our hypothesis that pre‐activation increases active force development. Highly compliant titin allows cells to function at higher diastolic [Ca2+].
Collapse
Affiliation(s)
- Aref Najafi
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands.,Netherlands Heart Institute, PO box 19258, 3501 DG, Utrecht, the Netherlands
| | - Martijn van de Locht
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Maike Schuldt
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | | | | | - Ilse Bollen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Max Goebel
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Coen A C Ottenheijm
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| | - Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands.,Netherlands Heart Institute, PO box 19258, 3501 DG, Utrecht, the Netherlands
| | - Michiel Helmes
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands.,Ionoptix, de Boelelaan 1108, 1081 HV, Amsterdam, the Netherlands.,CytoCypher, de Boelelaan 1108, 1081 HV, Amsterdam, the Netherlands
| | - Diederik W D Kuster
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, de Boelelaan 1117, 1081 HZ, Amsterdam, the Netherlands
| |
Collapse
|
92
|
Ovchinnikov AG, Potekhina AV, Ibragimova NM, Barabanova EA, Yushchyuk EN, Ageev FT. [Mechanisms of exercise intolerance in patients with heart failure and preserved ejection fraction. Part I: The role of impairments in the left heart chambers]. ACTA ACUST UNITED AC 2019; 59:4-16. [PMID: 31340744 DOI: 10.18087/cardio.n394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 07/24/2019] [Indexed: 11/18/2022]
Abstract
During exercise an increase in oxygen delivery to working muscles is achieved through well‑coordinated interaction of many organs and systems: the heart, lungs, blood vessels, skeletal muscles, and the autonomic nervous system. In heart failure with preserved left ventricular ejection fraction, all mechanisms involved in the normal exercise tolerance are impaired. In the first part of this review, the impairments of the left heart chambers are considered ‑ left ventricular diastolic dysfunction, the weakening of the contractile and chronotropic reserves, left atrium dysfunction; the possible ways of their medical correction are also presented.
Collapse
Affiliation(s)
- A G Ovchinnikov
- FSBO National Medical research center of cardiology of the Ministry of healthcare of the Russian Federation
| | - A V Potekhina
- FSBO National Medical research center of cardiology of the Ministry of healthcare of the Russian Federation
| | - N M Ibragimova
- FSBO National Medical research center of cardiology of the Ministry of healthcare of the Russian Federation
| | - E A Barabanova
- I. M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E N Yushchyuk
- A. I. Evdokimov Moscow State University for Medicine and Dentistry
| | - F T Ageev
- FSBO National Medical research center of cardiology of the Ministry of healthcare of the Russian Federation
| |
Collapse
|
93
|
Michels da Silva D, Langer H, Graf T. Inflammatory and Molecular Pathways in Heart Failure-Ischemia, HFpEF and Transthyretin Cardiac Amyloidosis. Int J Mol Sci 2019; 20:ijms20092322. [PMID: 31083399 PMCID: PMC6540104 DOI: 10.3390/ijms20092322] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
Elevated pro-inflammatory biomarkers and cytokines are associated with morbidity and mortality in heart failure (HF). Preclinical and clinical studies have shown multiple inflammatory mechanisms causing cardiac remodeling, dysfunction and chronic failure. Therapeutics in trials targeting the immune response in heart failure and its effects did not result in evident benefits regarding clinical endpoints and mortality. This review elaborates pathways of immune cytokines in pathogenesis and worsening of heart failure in clinical and cellular settings. Besides the well-known mechanisms of immune activation and inflammation in atherosclerosis causing ischemic cardiomyopathy or myocarditis, attention is focused on other mechanisms leading to heart failure such as transthyretin (TTR) amyloidosis or heart failure with preserved ejection fraction. The knowledge of the pathogenesis in heart failure and amyloidosis on a molecular and cellular level might help to highlight new disease defining biomarkers and to lead the way to new therapeutic targets.
Collapse
Affiliation(s)
- Diana Michels da Silva
- Department of Cardiology, Angiology and Intensive Care, Medicine Medical Clinic II, University Heart Center Lübeck, 23562 Lübeck, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany.
| | - Harald Langer
- Department of Cardiology, Angiology and Intensive Care, Medicine Medical Clinic II, University Heart Center Lübeck, 23562 Lübeck, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany.
| | - Tobias Graf
- Department of Cardiology, Angiology and Intensive Care, Medicine Medical Clinic II, University Heart Center Lübeck, 23562 Lübeck, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany.
| |
Collapse
|
94
|
Koser F, Loescher C, Linke WA. Posttranslational modifications of titin from cardiac muscle: how, where, and what for? FEBS J 2019; 286:2240-2260. [PMID: 30989819 PMCID: PMC6850032 DOI: 10.1111/febs.14854] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/27/2019] [Accepted: 04/13/2019] [Indexed: 12/11/2022]
Abstract
Titin is a giant elastic protein expressed in the contractile units of striated muscle cells, including the sarcomeres of cardiomyocytes. The last decade has seen enormous progress in our understanding of how titin molecular elasticity is modulated in a dynamic manner to help cardiac sarcomeres adjust to the varying hemodynamic demands on the heart. Crucial events mediating the rapid modulation of cardiac titin stiffness are post‐translational modifications (PTMs) of titin. In this review, we first recollect what is known from earlier and recent work on the molecular mechanisms of titin extensibility and force generation. The main goal then is to provide a comprehensive overview of current insight into the relationship between titin PTMs and cardiomyocyte stiffness, notably the effect of oxidation and phosphorylation of titin spring segments on titin stiffness. A synopsis is given of which type of oxidative titin modification can cause which effect on titin stiffness. A large part of the review then covers the mechanically relevant phosphorylation sites in titin, their location along the elastic segment, and the protein kinases and phosphatases known to target these sites. We also include a detailed coverage of the complex changes in phosphorylation at specific titin residues, which have been reported in both animal models of heart disease and in human heart failure, and their correlation with titin‐based stiffness alterations. Knowledge of the relationship between titin PTMs and titin elasticity can be exploited in the search for therapeutic approaches aimed at softening the pathologically stiffened myocardium in heart failure patients.
Collapse
|
95
|
Rodrigues PG, Miranda-Silva D, Costa SM, Barros C, Hamdani N, Moura C, Mendes MJ, Sousa-Mendes C, Trindade F, Fontoura D, Vitorino R, Linke WA, Leite-Moreira AF, Falcão-Pires I. Early myocardial changes induced by doxorubicin in the nonfailing dilated ventricle. Am J Physiol Heart Circ Physiol 2019; 316:H459-H475. [DOI: 10.1152/ajpheart.00401.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several studies have demonstrated that administration of doxorubicin (DOXO) results in cardiotoxicity, which eventually progresses to dilated cardiomyopathy. The present work aimed to evaluate the early myocardial changes of DOXO-induced cardiotoxicity. Male New Zealand White rabbits were injected intravenously with DOXO twice weekly for 8 wk [DOXO-induced heart failure (DOXO-HF)] or with an equivolumetric dose of saline (control). Echocardiographic evaluation was performed, and myocardial samples were collected to evaluate myocardial cellular and molecular modifications. The DOXO-HF group presented cardiac hypertrophy and higher left ventricular cavity diameters, showing a dilated phenotype but preserved ejection fraction. Concerning cardiomyocyte function, the DOXO-HF group presented a trend toward increased active tension without significant differences in passive tension. The myocardial GSSG-to-GSH ratio and interstitial fibrosis were increased and Bax-to- Bcl-2 ratio presented a trend toward an increase, suggesting the activation of apoptosis signaling pathways. The macromolecule titin shifted toward the more compliant isoform (N2BA), whereas the stiffer one (N2B) was shown to be hypophosphorylated. Differential protein analysis from the aggregate-enriched fraction through gel liquid chromatography-tandem mass spectrometry revealed an increase in the histidine-rich glycoprotein fragment in DOXO-HF animals. This work describes novel and early myocardial effects of DOXO-induced cardiotoxicity. Thus, tracking these changes appears to be of extreme relevance for the early detection of cardiac damage (as soon as ventricular dilation becomes evident) before irreversible cardiac function deterioration occurs (reduced ejection fraction). Moreover, it allows for the adjustment of the therapeutic approach and thus the prevention of cardiomyopathy progression. NEW & NOTEWORTHY Identification of early myocardial effects of doxorubicin in the heart is essential to hinder the development of cardiac complications and adjust the therapeutic approach. This study describes doxorubicin-induced cellular and molecular modifications before the onset of dilated cardiomyopathy. Myocardial samples from doxorubicin-treated rabbits showed a tendency for higher cardiomyocyte active tension, titin isoform shift from N2B to N2BA, hypophosphorylation of N2B, increased apoptotic genes, left ventricular interstitial fibrosis, and increased aggregation of histidine-rich glycoprotein.
Collapse
Affiliation(s)
- Patricia G. Rodrigues
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Daniela Miranda-Silva
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Sofia M. Costa
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Carla Barros
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Nazha Hamdani
- Department of Systems Physiology, Ruhr University, Bochum, Germany
| | - Cláudia Moura
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Maria J. Mendes
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Cláudia Sousa-Mendes
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Fábio Trindade
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Dulce Fontoura
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Rui Vitorino
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Wolfgang A. Linke
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
- Department of Cardiothoracic Surgery, São João Hospital Centre, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
96
|
van der Velden J, Stienen GJM. Cardiac Disorders and Pathophysiology of Sarcomeric Proteins. Physiol Rev 2019; 99:381-426. [PMID: 30379622 DOI: 10.1152/physrev.00040.2017] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The sarcomeric proteins represent the structural building blocks of heart muscle, which are essential for contraction and relaxation. During recent years, it has become evident that posttranslational modifications of sarcomeric proteins, in particular phosphorylation, tune cardiac pump function at rest and during exercise. This delicate, orchestrated interaction is also influenced by mutations, predominantly in sarcomeric proteins, which cause hypertrophic or dilated cardiomyopathy. In this review, we follow a bottom-up approach starting from a description of the basic components of cardiac muscle at the molecular level up to the various forms of cardiac disorders at the organ level. An overview is given of sarcomere changes in acquired and inherited forms of cardiac disease and the underlying disease mechanisms with particular reference to human tissue. A distinction will be made between the primary defect and maladaptive/adaptive secondary changes. Techniques used to unravel functional consequences of disease-induced protein changes are described, and an overview of current and future treatments targeted at sarcomeric proteins is given. The current evidence presented suggests that sarcomeres not only form the basis of cardiac muscle function but also represent a therapeutic target to combat cardiac disease.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Ger J M Stienen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, Amsterdam , The Netherlands ; and Department of Physiology, Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|
97
|
LeWinter MM. Pericardiectomy to Treat Heart Failure With Preserved Ejection Fraction: Unrestrained Enthusiasm? Circ Heart Fail 2019; 10:e003971. [PMID: 28396502 DOI: 10.1161/circheartfailure.117.003971] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
98
|
Patel RB, Shah SJ. Drug Targets for Heart Failure with Preserved Ejection Fraction: A Mechanistic Approach and Review of Contemporary Clinical Trials. Annu Rev Pharmacol Toxicol 2019; 59:41-63. [PMID: 30296895 PMCID: PMC6327844 DOI: 10.1146/annurev-pharmtox-010818-021136] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for over half of prevalent heart failure (HF) worldwide, and prognosis after hospitalization for HFpEF remains poor. Due, at least in part, to the heterogeneous nature of HFpEF, drug development has proved immensely challenging. Currently, there are no universally accepted therapies that alter the clinical course of HFpEF. Despite these challenges, important mechanistic understandings of the disease have revealed that the pathophysiology of HFpEF is distinct from that of HF with reduced ejection fraction and have also highlighted potential new therapeutic targets for HFpEF. Of note, HFpEF is a systemic syndrome affecting multiple organ systems. Depending on the organ systems involved, certain novel therapies offer promise in reducing the morbidity of the HFpEF syndrome. In this review, we aim to discuss novel pharmacotherapies for HFpEF based on its unique pathophysiology and identify key research strategies to further elucidate mechanistic pathways to develop novel therapeutics in the future.
Collapse
Affiliation(s)
- Ravi B Patel
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Sanjiv J Shah
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
- T1 Center for Cardiovascular Therapeutics, Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA
| |
Collapse
|
99
|
Masci PG, Pavon AG, Berchier G, Schwitter J. Probing the intravascular and interstitial compartments of remodeled myocardium in heart failure patients with preserved and reduced ejection fraction: a CMR study. BMC Med Imaging 2019; 19:1. [PMID: 30611240 PMCID: PMC6320584 DOI: 10.1186/s12880-018-0301-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 12/20/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Recent autopsy studies found microvascular rarefaction in remodeled myocardium of patients who died of heart failure with preserved ejection-fraction (HFpEF). This condition has not been investigated so far by non-invasive methods in patients with HFpEF. The aim was to quantify the intravascular volume (IVV) compartment by CMR in HFpEF patients. METHODS In two separate CMR examinations, HFpEF patients (n = 6; 12 examinations) and post-myocardial infarction patients (post-MI; n = 6; 12 examinations) were studied with T1-mapping (MOLLI-sequence) before and after IV bolus of 0.03 mmol/Kg of the intravascular contrast-medium (CM) Gadofosveset and 0.2 mmol/Kg of the extravascular CM Gadobutrol yielding IVV and extracellular volume (ECV), respectively. Healthy controls (n = 10 with Gadofosveset only, n = 10 with Gadobutrol only) were also studied with the same protocol. IVV and ECV were measured in the basal septum (without ischemic scar in post-MI patients). In post-MI patients, ECV and IVV were also measured in the ischemic scar. Left ventricular (LV) volumes, mass, and ejection-fraction were measured by standard protocol. LV global longitudinal strain (GLS) was calculated by feature tracking on long-axis cine acquisitions. RESULTS LV mass to end-diastolic volume ratio and GLS in HFpEF were higher and lower, respectively, than in healthy controls and post-MI patients, whereas the post-MI patients showed lower LV ejection-fraction. Compared to healthy myocardium of controls, IVV in scar was reduced (0.135 ± 0.018 vs 0.109 ± 0.008, respectively, p = 0.005), while ECV was increased (0.244 ± 0.037 vs 0.698 ± 0.106, respectively, p < 0.001). However, IVV did not differ among HFpEF, post-MI, and healthy controls (0.155 ± 0.033, 0.146 ± 0.038, and 0.135 ± 0.018, respectively, p = 0.413), whereas ECV was higher in HFpEF than in post-MI and healthy controls (0.304 ± 0.159, 0.270 ± 0.017, and 0.244 ± 0.037, respectively, p = 0.003). CONCLUSIONS The T1-mapping technique combined with an intravascular CM shows potential to measure IVV. In infarct scar with substantially increased ECV, IVV was significantly reduced. Unlike in infarct scar, in remodeled myocardium of HFpEF patients, increased ECV was not accompanied by a reduction of IVV.
Collapse
Affiliation(s)
- Pier Giorgio Masci
- Lausanne University Hospital, Cardiovascular Department, and University of Lausanne, Lausanne, Switzerland.
| | - Anna Giulia Pavon
- Lausanne University Hospital, Cardiovascular Department, and University of Lausanne, Lausanne, Switzerland
- Institute of Cardiology, S. Raffaele Hospital, Milan, Italy
| | - Gregoire Berchier
- Radiology Department, University Hospital Lausanne-CHUV, Lausanne, Switzerland
| | - Juerg Schwitter
- Lausanne University Hospital, Cardiovascular Department, and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
100
|
Gritsyna YV, Ulanova AD, Salmov NN, Bobylev AG, Zhalimov VK, Vikhlyantsev IM. Differences in Titin and Nebulin Gene Expression in Skeletal Muscles of Rats Chronically Alcoholized by Different Methods. Mol Biol 2019. [DOI: 10.1134/s0026893319010035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|