1
|
Lange T, Backhaus SJ, Schulz A, Hashemi D, Evertz R, Kowallick JT, Hasenfuß G, Kelle S, Schuster A. CMR-based cardiac phenotyping in different forms of heart failure. Int J Cardiovasc Imaging 2024; 40:1585-1596. [PMID: 38878148 PMCID: PMC11258094 DOI: 10.1007/s10554-024-03145-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 05/15/2024] [Indexed: 07/19/2024]
Abstract
Heart failure (HF) is a heterogenous disease requiring precise diagnostics and knowledge of pathophysiological processes. Since structural and functional imaging data are scarce we hypothesized that cardiac magnetic resonance (CMR)-based analyses would provide accurate characterization and mechanistic insights into different HF groups comprising preserved (HFpEF), mid-range (HFmrEF) and reduced ejection fraction (HFrEF). 22 HFpEF, 17 HFmrEF and 15 HFrEF patients as well as 19 healthy volunteers were included. CMR image assessment contained left atrial (LA) and left ventricular (LV) volumetric evaluation as well as left atrioventricular coupling index (LACI). Furthermore, CMR feature-tracking included LV and LA strain in terms of reservoir (Es), conduit (Ee) and active boosterpump (Ea) function. CMR-based tissue characterization comprised T1 mapping as well as late-gadolinium enhancement (LGE) analyses. HFpEF patients showed predominant atrial impairment (Es 20.8%vs.25.4%, p = 0.02 and Ee 8.3%vs.13.5%, p = 0.001) and increased LACI compared to healthy controls (14.5%vs.23.3%, p = 0.004). Patients with HFmrEF showed LV enlargement but mostly preserved LA function with a compensatory increase in LA boosterpump (LA Ea: 15.0%, p = 0.049). In HFrEF LA and LV functional impairment was documented (Es: 14.2%, Ee: 5.4% p < 0.001 respectively; Ea: 8.8%, p = 0.02). This was paralleled by non-invasively assessed progressive fibrosis (T1 mapping and LGE; HFrEF > HFmrEF > HFpEF). CMR-imaging reveals insights into HF phenotypes with mainly atrial affection in HFpEF, ventricular affection with atrial compensation in HFmrEF and global impairment in HFrEF paralleled by progressive LV fibrosis. These data suggest a necessity for a personalized HF management based on imaging findings for future optimized patient management.
Collapse
Affiliation(s)
- Torben Lange
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
- German Centre of Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Sören J Backhaus
- Department of Cardiology, Campus Kerckhoff of the Justus-Liebig-Universität Gießen, Kerckhoff-Clinic, Bad Nauheim, Germany
| | - Alexander Schulz
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
- German Centre of Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Djawid Hashemi
- Department of Internal Medicine/Cardiology, Charité Campus Virchow Clinic, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Ruben Evertz
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
- German Centre of Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Johannes T Kowallick
- German Centre of Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
- Institute for Diagnostic and Interventional Radiology, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
| | - Gerd Hasenfuß
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Göttingen, Germany
- German Centre of Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany
| | - Sebastian Kelle
- Department of Internal Medicine/Cardiology, Charité Campus Virchow Clinic, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Andreas Schuster
- Department of Cardiology and Pneumology, University Medical Center Göttingen, Georg-August University, Göttingen, Germany.
- German Centre of Cardiovascular Research (DZHK), partner site Lower Saxony, Göttingen, Germany.
| |
Collapse
|
2
|
Barry C, Rouhana S, Braun JL, Geromella MS, Fajardo VA, Pyle WG. Perimenopause Decreases SERCA2a Activity in the Hearts of a Mouse Model of Ovarian Failure. Biomolecules 2024; 14:675. [PMID: 38927078 PMCID: PMC11201532 DOI: 10.3390/biom14060675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Risk of cardiovascular disease mortality rises in women after menopause. While increased cardiovascular risk is largely attributed to postmenopausal declines in estrogens, the molecular changes in the heart that contribute to risk are poorly understood. Disruptions in intracellular calcium handling develop in ovariectomized mice and have been implicated in cardiac dysfunction. Using a mouse model of menopause in which ovarian failure occurs over 120 days, we sought to determine if perimenopause impacted calcium removal mechanisms in the heart and identify the molecular mechanisms. Mice were injected with 4-vinylcyclohexene diepoxide (VCD) to induce ovarian failure over 120 days, mimicking perimenopause. Hearts were removed at 60 and 120 days after VCD injections, representing the middle and end of perimenopause. SERCA2a function was significantly diminished at the end of perimenopause. Neither SERCA2a nor phospholamban expression changed at either time point, but phospholamban phosphorylation at S16 and T17 was dynamically altered. Intrinsic SERCA inhibitors sarcolipin and myoregulin increased >4-fold at day 60, as did the native activator DWORF. At the end of perimenopause, sarcolipin and myoregulin returned to baseline levels while DWORF was significantly reduced below controls. Sodium-calcium exchanger expression was significantly increased at the end of perimenopause. These results show that the foundation for increased cardiovascular disease mortality develops in the heart during perimenopause and that regulators of calcium handling exhibit significant fluctuations over time. Understanding the temporal development of cardiovascular risk associated with menopause and the underlying mechanisms is critical to developing interventions that mitigate the rise in cardiovascular mortality that arises after menopause.
Collapse
Affiliation(s)
- Ciara Barry
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
| | - Sarah Rouhana
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
| | - Jessica L. Braun
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Mia S. Geromella
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Val A. Fajardo
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada (V.A.F.)
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - W. Glen Pyle
- IMPART Team Canada Investigator Network, Dalhousie Medicine, Saint John, NB E2K 5E2, Canada
- Women’s Health Research Institute at BC Women’s Hospital + Health Centre, Vancouver, BC V6H 2N9, Canada
| |
Collapse
|
3
|
Guo YJ, Yao JJ, Guo ZZ, Ding M, Zhang KL, Shen QH, Li Y, Yu SF, Wan T, Xu FP, Wang Y, Qi XX, Wu JJ, Chen JX, Liu ZQ, Lu LL. HBB contributes to individualized aconitine-induced cardiotoxicity in mice via interfering with ABHD5/AMPK/HDAC4 axis. Acta Pharmacol Sin 2024; 45:1224-1236. [PMID: 38467717 PMCID: PMC11130212 DOI: 10.1038/s41401-023-01206-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 11/19/2023] [Indexed: 03/13/2024] Open
Abstract
The root of Aconitum carmichaelii Debx. (Fuzi) is an herbal medicine used in China that exerts significant efficacy in rescuing patients from severe diseases. A key toxic compound in Fuzi, aconitine (AC), could trigger unpredictable cardiotoxicities with high-individualization, thus hinders safe application of Fuzi. In this study we investigated the individual differences of AC-induced cardiotoxicities, the biomarkers and underlying mechanisms. Diversity Outbred (DO) mice were used as a genetically heterogeneous model for mimicking individualization clinically. The mice were orally administered AC (0.3, 0.6, 0.9 mg· kg-1 ·d-1) for 7 d. We found that AC-triggered cardiotoxicities in DO mice shared similar characteristics to those observed in clinic patients. Most importantly, significant individual differences were found in DO mice (variation coefficients: 34.08%-53.17%). RNA-sequencing in AC-tolerant and AC-sensitive mice revealed that hemoglobin subunit beta (HBB), a toxic-responsive protein in blood with 89% homology to human, was specifically enriched in AC-sensitive mice. Moreover, we found that HBB overexpression could significantly exacerbate AC-induced cardiotoxicity while HBB knockdown markedly attenuated cell death of cardiomyocytes. We revealed that AC could trigger hemolysis, and specifically bind to HBB in cell-free hemoglobin (cf-Hb), which could excessively promote NO scavenge and decrease cardioprotective S-nitrosylation. Meanwhile, AC bound to HBB enhanced the binding of HBB to ABHD5 and AMPK, which correspondingly decreased HDAC-NT generation and led to cardiomyocytes death. This study not only demonstrates HBB achievement a novel target of AC in blood, but provides the first clue for HBB as a novel biomarker in determining the individual differences of Fuzi-triggered cardiotoxicity.
Collapse
Affiliation(s)
- Ya-Juan Guo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jing-Jing Yao
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhen-Zhen Guo
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ming Ding
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Kun-Lin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Qing-Hong Shen
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yu Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Shao-Fang Yu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ting Wan
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Fu-Ping Xu
- Guandong Provincial hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Ying Wang
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiao-Xiao Qi
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jin-Jun Wu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jian-Xin Chen
- Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Zhong-Qiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
| | - Lin-Lin Lu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
4
|
Saad NS, Mashali MA, Repas SJ, Janssen PML. Altering Calcium Sensitivity in Heart Failure: A Crossroads of Disease Etiology and Therapeutic Innovation. Int J Mol Sci 2023; 24:17577. [PMID: 38139404 PMCID: PMC10744146 DOI: 10.3390/ijms242417577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Heart failure (HF) presents a significant clinical challenge, with current treatments mainly easing symptoms without stopping disease progression. The targeting of calcium (Ca2+) regulation is emerging as a key area for innovative HF treatments that could significantly alter disease outcomes and enhance cardiac function. In this review, we aim to explore the implications of altered Ca2+ sensitivity, a key determinant of cardiac muscle force, in HF, including its roles during systole and diastole and its association with different HF types-HF with preserved and reduced ejection fraction (HFpEF and HFrEF, respectively). We further highlight the role of the two rate constants kon (Ca2+ binding to Troponin C) and koff (its dissociation) to fully comprehend how changes in Ca2+ sensitivity impact heart function. Additionally, we examine how increased Ca2+ sensitivity, while boosting systolic function, also presents diastolic risks, potentially leading to arrhythmias and sudden cardiac death. This suggests that strategies aimed at moderating myofilament Ca2+ sensitivity could revolutionize anti-arrhythmic approaches, reshaping the HF treatment landscape. In conclusion, we emphasize the need for precision in therapeutic approaches targeting Ca2+ sensitivity and call for comprehensive research into the complex interactions between Ca2+ regulation, myofilament sensitivity, and their clinical manifestations in HF.
Collapse
Affiliation(s)
- Nancy S. Saad
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
| | - Mohammed A. Mashali
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
- Department of Surgery, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22514, Egypt
| | - Steven J. Repas
- Department of Emergency Medicine, Wright State University Boonshoft School of Medicine, Dayton, OH 45324, USA;
| | - Paul M. L. Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
5
|
Abstract
Chronic kidney disease is associated with an increased risk for the development and progression of cardiovascular disorders including hypertension, dyslipidemia, and coronary artery disease. Chronic kidney disease may also affect the myocardium through complex systemic changes, resulting in structural remodeling such as hypertrophy and fibrosis, as well as impairments in both diastolic and systolic function. These cardiac changes in the setting of chronic kidney disease define a specific cardiomyopathic phenotype known as uremic cardiomyopathy. Cardiac function is tightly linked to its metabolism, and research over the past 3 decades has revealed significant metabolic remodeling in the myocardium during the development of heart failure. Because the concept of uremic cardiomyopathy has only been recognized in recent years, there are limited data on metabolism in the uremic heart. Nonetheless, recent findings suggest overlapping mechanisms with heart failure. This work reviews key features of metabolic remodeling in the failing heart in the general population and extends this to patients with chronic kidney disease. The knowledge of similarities and differences in cardiac metabolism between heart failure and uremic cardiomyopathy may help identify new targets for mechanistic and therapeutic research on uremic cardiomyopathy.
Collapse
Affiliation(s)
- T Dung Nguyen
- Department of Internal Medicine I, University Hospital Jena, Jena, Germany
| | | |
Collapse
|
6
|
Dulce RA, Kanashiro-Takeuchi RM, Takeuchi LM, Salerno AG, Wanschel ACBA, Kulandavelu S, Balkan W, Zuttion MSSR, Cai R, Schally AV, Hare JM. Synthetic growth hormone-releasing hormone agonist ameliorates the myocardial pathophysiology characteristic of heart failure with preserved ejection fraction. Cardiovasc Res 2023; 118:3586-3601. [PMID: 35704032 DOI: 10.1093/cvr/cvac098] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 05/06/2022] [Accepted: 05/25/2022] [Indexed: 02/07/2023] Open
Abstract
AIMS To test the hypothesis that the activation of the growth hormone-releasing hormone (GHRH) receptor signalling pathway within the myocardium both prevents and reverses diastolic dysfunction and pathophysiologic features consistent with heart failure with preserved ejection fraction (HFpEF). Impaired myocardial relaxation, fibrosis, and ventricular stiffness, among other multi-organ morbidities, characterize the phenotype underlying the HFpEF syndrome. Despite the rapidly increasing prevalence of HFpEF, few effective therapies have emerged. Synthetic agonists of the GHRH receptors reduce myocardial fibrosis, cardiomyocyte hypertrophy, and improve performance in animal models of ischaemic cardiomyopathy, independently of the growth hormone axis. METHODS AND RESULTS CD1 mice received 4- or 8-week continuous infusion of angiotensin-II (Ang-II) to generate a phenotype with several features consistent with HFpEF. Mice were administered either vehicle or a potent synthetic agonist of GHRH, MR-356 for 4-weeks beginning concurrently or 4-weeks following the initiation of Ang-II infusion. Ang-II-treated animals exhibited diastolic dysfunction, ventricular hypertrophy, interstitial fibrosis, and normal ejection fraction. Cardiomyocytes isolated from these animals exhibited incomplete relaxation, depressed contractile responses, altered myofibrillar protein phosphorylation, and disturbed calcium handling mechanisms (ex vivo). MR-356 both prevented and reversed the development of the pathological phenotype in vivo and ex vivo. Activation of the GHRH receptors increased cAMP and cGMP in cardiomyocytes isolated from control animals but only cAMP in cardiac fibroblasts, suggesting that GHRH-A exert differential effects on cardiomyocytes and fibroblasts. CONCLUSION These findings indicate that the GHRH receptor signalling pathway(s) represents a new molecular target to counteract dysfunctional cardiomyocyte relaxation by targeting myofilament phosphorylation and fibrosis. Accordingly, activation of GHRH receptors with potent, synthetic GHRH agonists may provide a novel therapeutic approach to management of the myocardial alterations associated with the HFpEF syndrome.
Collapse
Affiliation(s)
- Raul A Dulce
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Rosemeire M Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Alessandro G Salerno
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Amarylis C B A Wanschel
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Shathiyah Kulandavelu
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wayne Balkan
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Marilia S S R Zuttion
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA
| | - Renzhi Cai
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, FL 33125, USA
| | - Andrew V Schally
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.,Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, FL 33125, USA.,Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, 1501 N.W. 10th Avenue, Room 908, Miami, FL 33136, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
7
|
Karsenty C, Guilbeau-Frugier C, Genet G, Seguelas MH, Alzieu P, Cazorla O, Montagner A, Blum Y, Dubroca C, Maupoint J, Tramunt B, Cauquil M, Sulpice T, Richard S, Arcucci S, Flores-Flores R, Pataluch N, Montoriol R, Sicard P, Deney A, Couffinhal T, Senard JM, Galés C. Ephrin-B1 regulates the adult diastolic function through a late postnatal maturation of cardiomyocyte surface crests. eLife 2023; 12:e80904. [PMID: 36649053 PMCID: PMC9844986 DOI: 10.7554/elife.80904] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
The rod-shaped adult cardiomyocyte (CM) harbors a unique architecture of its lateral surface with periodic crests, relying on the presence of subsarcolemmal mitochondria (SSM) with unknown role. Here, we investigated the development and functional role of CM crests during the postnatal period. We found in rodents that CM crest maturation occurs late between postnatal day 20 (P20) and P60 through both SSM biogenesis, swelling and crest-crest lateral interactions between adjacent CM, promoting tissue compaction. At the functional level, we showed that the P20-P60 period is dedicated to the improvement of relaxation. Interestingly, crest maturation specifically contributes to an atypical CM hypertrophy of its short axis, without myofibril addition, but relying on CM lateral stretching. Mechanistically, using constitutive and conditional CM-specific knock-out mice, we identified ephrin-B1, a lateral membrane stabilizer, as a molecular determinant of P20-P60 crest maturation, governing both the CM lateral stretch and the diastolic function, thus highly suggesting a link between crest maturity and diastole. Remarkably, while young adult CM-specific Efnb1 KO mice essentially exhibit an impairment of the ventricular diastole with preserved ejection fraction and exercise intolerance, they progressively switch toward systolic heart failure with 100% KO mice dying after 13 months, indicative of a critical role of CM-ephrin-B1 in the adult heart function. This study highlights the molecular determinants and the biological implication of a new late P20-P60 postnatal developmental stage of the heart in rodents during which, in part, ephrin-B1 specifically regulates the maturation of the CM surface crests and of the diastolic function.
Collapse
Affiliation(s)
- Clement Karsenty
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Pediatric Cardiology, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Celine Guilbeau-Frugier
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Forensic Medicine, Centre Hospitalier Universitaire de Toulouse, Université de ToulouseToulouseFrance
| | - Gaël Genet
- Department of Cell Biology, University of Virginia School of MedicineCharlottesvilleUnited States
| | - Marie-Helene Seguelas
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Philippe Alzieu
- Université de Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
| | - Olivier Cazorla
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Alexandra Montagner
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Yuna Blum
- IGDR UMR 6290, CNRS, Université de Rennes 1RennesFrance
| | | | | | - Blandine Tramunt
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Diabetology, Metabolic Diseases & Nutrition, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Marie Cauquil
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | | | - Sylvain Richard
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Silvia Arcucci
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Remy Flores-Flores
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Nicolas Pataluch
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Romain Montoriol
- Department of Forensic Medicine, Centre Hospitalier Universitaire de Toulouse, Université de ToulouseToulouseFrance
| | - Pierre Sicard
- Université de Montpellier, INSERM, CNRS, PhyMedExpMontpellierFrance
| | - Antoine Deney
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| | - Thierry Couffinhal
- Université de Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
- Service des Maladies Cardiaques et Vasculaires, CHU de BordeauxBordeauxFrance
| | - Jean-Michel Senard
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
- Department of Clinical Pharmacology, Centre Hospitalier Universitaire de ToulouseToulouseFrance
| | - Celine Galés
- INSERM, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de ToulouseToulouseFrance
| |
Collapse
|
8
|
Hegner P, Drzymalski M, Biedermann A, Memmel B, Durczok M, Wester M, Floerchinger B, Provaznik Z, Schmid C, Zausig Y, Maier LS, Wagner S. SAR296968, a Novel Selective Na+/Ca2+ Exchanger Inhibitor, Improves Ca2+ Handling and Contractile Function in Human Atrial Cardiomyocytes. Biomedicines 2022; 10:biomedicines10081932. [PMID: 36009478 PMCID: PMC9406204 DOI: 10.3390/biomedicines10081932] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 07/30/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background: In reverse-mode, cardiac sodium-calcium exchanger (NCX) can increase the cytoplasmic Ca2+ concentration in response to high intracellular Na+ levels, which may contribute to diastolic contractile dysfunction. Furthermore, increased spontaneous Ca2+ release from intracellular stores can activate forward mode NCX. The resulting transient inward current causes delayed afterdepolarization (DAD)-dependent arrhythmias. Moreover, recently, NCX has been associated with impaired relaxation and reduced cardiac function in heart failure with preserved ejection fraction (HFpEF). Since NCX is upregulated in human chronic atrial fibrillation (AF) as well as heart failure (HF), specific inhibition may have therapeutic potential. Objective: We tested the antiarrhythmic, lusitropic and inotropic effects of a novel selective NCX-inhibitor (SAR296968) in human atrial myocardium. Methods and Results: Right atrial appendage biopsies of 46 patients undergoing elective cardiac surgery in a predominant HFpEF cohort (n = 24/46) were investigated. In isolated human atrial cardiomyocytes, SAR296968 reduced the frequency of spontaneous SR Ca2+ release events and increased caffeine transient amplitude. In accordance, in isolated atrial trabeculae, SAR296968 enhanced the developed tension after a 30 s pause of electrical stimulation consistent with reduced diastolic sarcoplasmic reticulum (SR) Ca2+ leak. Moreover, compared to vehicle, SAR296968 decreased steady-state diastolic tension (at 1 Hz) without impairing developed systolic tension. Importantly, SAR296968 did not affect the safety parameters, such as resting membrane potential or action potential duration as measured by patch clamp. Conclusion: The novel selective NCX-inhibitor SAR296968 inhibits atrial pro-arrhythmic activity and improves diastolic and contractile function in human atrial myocardium, which may have therapeutic implications, especially for treatment of HFpEF.
Collapse
Affiliation(s)
- Philipp Hegner
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Marzena Drzymalski
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Alexander Biedermann
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Bernadette Memmel
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Melanie Durczok
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Michael Wester
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Bernhard Floerchinger
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Zdenek Provaznik
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - York Zausig
- Department of Anesthesiology, University Medical Center Regensburg, 93053 Regensburg, Germany
- Department of Anesthesiology and Operative Intensive Care Medicine, Aschaffenburg-Alzenau Hospital, 63739 Aschaffenburg, Germany
| | - Lars S. Maier
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Medical Center Regensburg, 93053 Regensburg, Germany
- Correspondence: ; Tel.: +49-941-944-7206
| |
Collapse
|
9
|
Zhang M, Shu H, Chen C, He Z, Zhou Z, Wang DW. Epoxyeicosatrienoic acid: A potential therapeutic target of heart failure with preserved ejection fraction. Biomed Pharmacother 2022; 153:113326. [PMID: 35759865 DOI: 10.1016/j.biopha.2022.113326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 11/02/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) reduces the quality of life, costs substantial medical resources, and has a high mortality. However, we lack an effective therapy for HFpEF due to our limited knowledge of its mechanism. Therefore, it is crucial to explore novel therapeutics, such as those with endogenous protective roles, and seek new targeted therapies. Epoxyeicosatrienoic acids (EETs) are endogenous bioactive metabolites of arachidonic acids produced by cytochrome P450 (CYP) epoxygenases. EETs can function as endogenous cardioprotective factors with potent inhibitory roles in inflammation, endothelial dysfunction, cardiac remodeling, and fibrosis, which are the fundamental mechanisms of HFpEF. This suggests that EETs have the potential function to protect against HFpEF. Therefore, we present an overview of the ever-expanding world of EETs and how they might help alleviate the pathophysiology underlying HFpEF to provide new insights for research in this field.
Collapse
Affiliation(s)
- Min Zhang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Zuowen He
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Zhou Zhou
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan 430030, China.
| |
Collapse
|
10
|
Chen X, Lin H, Xiong W, Pan J, Huang S, Xu S, He S, Lei M, Chang ACY, Zhang H. p53-Dependent Mitochondrial Compensation in Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2022; 11:e024582. [PMID: 35656994 PMCID: PMC9238719 DOI: 10.1161/jaha.121.024582] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Heart failure with preserved ejection fraction (HFpEF) accounts for 50% of patients with heart failure. Clinically, HFpEF prevalence shows age and gender biases. Although the majority of patients with HFpEF are elderly, there is an emergence of young patients with HFpEF. A better understanding of the underlying pathogenic mechanism is urgently needed. Here, we aimed to determine the role of aging in the pathogenesis of HFpEF. Methods and Results HFpEF dietary regimen (high‐fat diet + Nω‐Nitro‐L‐arginine methyl ester hydrochloride) was used to induce HFpEF in wild type and telomerase RNA knockout mice (second‐generation and third‐generation telomerase RNA component knockout), an aging murine model. First, both male and female animals develop HFpEF equally. Second, cardiac wall thickening preceded diastolic dysfunction in all HFpEF animals. Third, accelerated HFpEF onset was observed in second‐generation telomerase RNA component knockout (at 6 weeks) and third‐generation telomerase RNA component knockout (at 4 weeks) compared with wild type (8 weeks). Fourth, we demonstrate that mitochondrial respiration transitioned from compensatory state (normal basal yet loss of maximal respiratory capacity) to dysfunction (loss of both basal and maximal respiratory capacity) in a p53 dosage dependent manner. Last, using myocardial‐specific p53 knockout animals, we demonstrate that loss of p53 activation delays the development of HFpEF. Conclusions Here we demonstrate that p53 activation plays a role in the pathogenesis of HFpEF. We show that short telomere animals exhibit a basal level of p53 activation, mitochondria upregulate mtDNA encoded genes as a mean to compensate for blocked mitochondrial biogenesis, and loss of myocardial p53 delays HFpEF onset in high fat diet + Nω‐Nitro‐L‐arginine methyl ester hydrochloride challenged murine model.
Collapse
Affiliation(s)
- Xiaonan Chen
- Department of Cardiology Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Hao Lin
- Department of Cardiology Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Weiyao Xiong
- Shanghai Institute of Precision MedicineNinth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Jianan Pan
- Department of Cardiology Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Shuying Huang
- Department of Cardiology Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Shan Xu
- Shanghai Institute of Precision MedicineNinth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Shufang He
- Shanghai Institute of Precision MedicineNinth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Ming Lei
- Shanghai Institute of Precision MedicineNinth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Alex Chia Yu Chang
- Department of Cardiology Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China.,Shanghai Institute of Precision MedicineNinth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| | - Huili Zhang
- Department of Cardiology Ninth People's HospitalShanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
11
|
Grabowski K, Herlan L, Witten A, Qadri F, Eisenreich A, Lindner D, Schädlich M, Schulz A, Subrova J, Mhatre KN, Primessnig U, Plehm R, van Linthout S, Escher F, Bader M, Stoll M, Westermann D, Heinzel FR, Kreutz R. Cpxm2 as a novel candidate for cardiac hypertrophy and failure in hypertension. Hypertens Res 2022; 45:292-307. [PMID: 34916661 PMCID: PMC8766285 DOI: 10.1038/s41440-021-00826-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/08/2021] [Accepted: 10/29/2021] [Indexed: 12/18/2022]
Abstract
Treatment of hypertension-mediated cardiac damage with left ventricular (LV) hypertrophy (LVH) and heart failure remains challenging. To identify novel targets, we performed comparative transcriptome analysis between genetic models derived from stroke-prone spontaneously hypertensive rats (SHRSP). Here, we identified carboxypeptidase X 2 (Cpxm2) as a genetic locus affecting LV mass. Analysis of isolated rat cardiomyocytes and cardiofibroblasts indicated Cpxm2 expression and intrinsic upregulation in genetic hypertension. Immunostaining indicated that CPXM2 associates with the t-tubule network of cardiomyocytes. The functional role of Cpxm2 was further investigated in Cpxm2-deficient (KO) and wild-type (WT) mice exposed to deoxycorticosterone acetate (DOCA). WT and KO animals developed severe and similar systolic hypertension in response to DOCA. WT mice developed severe LV damage, including increases in LV masses and diameters, impairment of LV systolic and diastolic function and reduced ejection fraction. These changes were significantly ameliorated or even normalized (i.e., ejection fraction) in KO-DOCA animals. LV transcriptome analysis showed a molecular cardiac hypertrophy/remodeling signature in WT but not KO mice with significant upregulation of 1234 transcripts, including Cpxm2, in response to DOCA. Analysis of endomyocardial biopsies from patients with cardiac hypertrophy indicated significant upregulation of CPXM2 expression. These data support further translational investigation of CPXM2.
Collapse
Affiliation(s)
- Katja Grabowski
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institut für Klinische Pharmakologie und Toxikologie, 10178 Berlin, Germany
| | - Laura Herlan
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institut für Klinische Pharmakologie und Toxikologie, 10178 Berlin, Germany
| | - Anika Witten
- grid.16149.3b0000 0004 0551 4246Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany
| | - Fatimunnisa Qadri
- grid.419491.00000 0001 1014 0849Max-Delbrück Center for Molecular Medicine (MDC), Berlin-Buch, Berlin, Germany
| | - Andreas Eisenreich
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institut für Klinische Pharmakologie und Toxikologie, 10178 Berlin, Germany
| | - Diana Lindner
- grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Clinic for Cardiology, University Heart and Vascular Center Hamburg, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Schädlich
- grid.16149.3b0000 0004 0551 4246Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany
| | - Angela Schulz
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institut für Klinische Pharmakologie und Toxikologie, 10178 Berlin, Germany
| | - Jana Subrova
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institut für Klinische Pharmakologie und Toxikologie, 10178 Berlin, Germany
| | - Ketaki Nitin Mhatre
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Cardiology, Campus Virchow Klinikum, 10178 Berlin, Germany
| | - Uwe Primessnig
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Cardiology, Campus Virchow Klinikum, 10178 Berlin, Germany ,grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Ralph Plehm
- grid.419491.00000 0001 1014 0849Max-Delbrück Center for Molecular Medicine (MDC), Berlin-Buch, Berlin, Germany
| | - Sophie van Linthout
- grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany ,grid.6363.00000 0001 2218 4662Charité—Universitätsmedizin Berlin, BCRT—Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Felicitas Escher
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Cardiology, Campus Virchow Klinikum, 10178 Berlin, Germany ,grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany ,grid.486773.9Institute of Cardiac Diagnostics and Therapy, IKDT GmbH, Berlin, Germany
| | - Michael Bader
- grid.419491.00000 0001 1014 0849Max-Delbrück Center for Molecular Medicine (MDC), Berlin-Buch, Berlin, Germany ,grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany ,grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 10178 Berlin, Germany ,grid.4562.50000 0001 0057 2672University of Lübeck, Institute for Biology, Ratzeburger Allee 160, 23562 Lübeck, Germany
| | - Monika Stoll
- grid.16149.3b0000 0004 0551 4246Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany ,grid.5012.60000 0001 0481 6099Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Dirk Westermann
- grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner site Hamburg/Kiel/Lübeck, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Clinic for Cardiology, University Heart and Vascular Center Hamburg, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Frank R. Heinzel
- grid.7468.d0000 0001 2248 7639Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Cardiology, Campus Virchow Klinikum, 10178 Berlin, Germany ,grid.452396.f0000 0004 5937 5237German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Reinhold Kreutz
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institut für Klinische Pharmakologie und Toxikologie, 10178, Berlin, Germany.
| |
Collapse
|
12
|
Benitah JP, Perrier R, Mercadier JJ, Pereira L, Gómez AM. RyR2 and Calcium Release in Heart Failure. Front Physiol 2021; 12:734210. [PMID: 34690808 PMCID: PMC8533677 DOI: 10.3389/fphys.2021.734210] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/30/2021] [Indexed: 12/24/2022] Open
Abstract
Heart Failure (HF) is defined as the inability of the heart to efficiently pump out enough blood to maintain the body's needs, first at exercise and then also at rest. Alterations in Ca2+ handling contributes to the diminished contraction and relaxation of the failing heart. While most Ca2+ handling protein expression and/or function has been shown to be altered in many models of experimental HF, in this review, we focus in the sarcoplasmic reticulum (SR) Ca2+ release channel, the type 2 ryanodine receptor (RyR2). Various modifications of this channel inducing alterations in its function have been reported. The first was the fact that RyR2 is less responsive to activation by Ca2+ entry through the L-Type calcium channel, which is the functional result of an ultrastructural remodeling of the ventricular cardiomyocyte, with fewer and disorganized transverse (T) tubules. HF is associated with an elevated sympathetic tone and in an oxidant environment. In this line, enhanced RyR2 phosphorylation and oxidation have been shown in human and experimental HF. After several controversies, it is now generally accepted that phosphorylation of RyR2 at the Calmodulin Kinase II site (S2814) is involved in both the depressed contractile function and the enhanced arrhythmic susceptibility of the failing heart. Diminished expression of the FK506 binding protein, FKBP12.6, may also contribute. While these alterations have been mostly studied in the left ventricle of HF with reduced ejection fraction, recent studies are looking at HF with preserved ejection fraction. Moreover, alterations in the RyR2 in HF may also contribute to supraventricular defects associated with HF such as sinus node dysfunction and atrial fibrillation.
Collapse
Affiliation(s)
| | | | | | | | - Ana M. Gómez
- Signaling and Cardiovascular Pathophysiology—UMR-S 1180, INSERM, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
13
|
Kovács ZZA, Szűcs G, Freiwan M, Kovács MG, Márványkövi FM, Dinh H, Siska A, Farkas K, Kovács F, Kriston A, Horváth P, Kővári B, Cserni BG, Cserni G, Földesi I, Csont T, Sárközy M. Comparison of the antiremodeling effects of losartan and mirabegron in a rat model of uremic cardiomyopathy. Sci Rep 2021; 11:17495. [PMID: 34471171 PMCID: PMC8410807 DOI: 10.1038/s41598-021-96815-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023] Open
Abstract
Uremic cardiomyopathy is characterized by diastolic dysfunction (DD), left ventricular hypertrophy (LVH), and fibrosis. Angiotensin-II plays a major role in the development of uremic cardiomyopathy via nitro-oxidative and inflammatory mechanisms. In heart failure, the beta-3 adrenergic receptor (β3-AR) is up-regulated and coupled to endothelial nitric oxide synthase (eNOS)-mediated pathways, exerting antiremodeling effects. We aimed to compare the antiremodeling effects of the angiotensin-II receptor blocker losartan and the β3-AR agonist mirabegron in uremic cardiomyopathy. Chronic kidney disease (CKD) was induced by 5/6th nephrectomy in male Wistar rats. Five weeks later, rats were randomized into four groups: (1) sham-operated, (2) CKD, (3) losartan-treated (10 mg/kg/day) CKD, and (4) mirabegron-treated (10 mg/kg/day) CKD groups. At week 13, echocardiographic, histologic, laboratory, qRT-PCR, and Western blot measurements proved the development of uremic cardiomyopathy with DD, LVH, fibrosis, inflammation, and reduced eNOS levels, which were significantly ameliorated by losartan. However, mirabegron showed a tendency to decrease DD and fibrosis; but eNOS expression remained reduced. In uremic cardiomyopathy, β3-AR, sarcoplasmic reticulum ATPase (SERCA), and phospholamban levels did not change irrespective of treatments. Mirabegron reduced the angiotensin-II receptor 1 expression in uremic cardiomyopathy that might explain its mild antiremodeling effects despite the unchanged expression of the β3-AR.
Collapse
Affiliation(s)
- Zsuzsanna Z A Kovács
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Gergő Szűcs
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Marah Freiwan
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Mónika G Kovács
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Fanni M Márványkövi
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Hoa Dinh
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Andrea Siska
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6720, Hungary
| | - Katalin Farkas
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6720, Hungary
| | - Ferenc Kovács
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726, Hungary
- Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726, Hungary
| | - András Kriston
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726, Hungary
- Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Péter Horváth
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, Szeged, 6726, Hungary
- Single-Cell Technologies Ltd, Temesvári krt. 62, Szeged, 6726, Hungary
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, 00014, Helsinki, Finland
| | - Bence Kővári
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720, Hungary
| | - Bálint Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720, Hungary
| | - Gábor Cserni
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged, 6720, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis utca 6, Szeged, 6720, Hungary
| | - Tamás Csont
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary.
| | - Márta Sárközy
- MEDICS Research Group, Department of Biochemistry, Interdisciplinary Center of Excellence, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary.
| |
Collapse
|
14
|
Chang D, Xu TT, Zhang SJ, Cai Y, Min SD, Zhao Z, Lu CQ, Wang YC, Ju S. Telmisartan ameliorates cardiac fibrosis and diastolic function in cardiorenal heart failure with preserved ejection fraction. Exp Biol Med (Maywood) 2021; 246:2511-2521. [PMID: 34342551 DOI: 10.1177/15353702211035058] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Chronic kidney disease (CKD) is a major contributor to the development of heart failure with preserved ejection fraction (HFpEF), whereas the underlying mechanism of cardiorenal HFpEF is still elusive. The aim of this study was to investigate the role of cardiac fibrosis in a rat model of cardiorenal HFpEF and explore whether treatment with Telmisartan, an inhibitor of renin-angiotensin-aldosterone system (RAAS), can ameliorate cardiac fibrosis and preserve diastolic function in cardiorenal HFpEF. Male rats were subjected to 5/6 subtotal nephrectomy (SNX) or sham operation (Sham), and rats were allowed four weeks to recover and form a stable condition of CKD. Telmisartan or vehicle was then administered p.o. (8 mg/kg/d) for 12 weeks. Blood pressure, brain natriuretic peptide (BNP), echocardiography, and cardiac magnetic resonance imaging were acquired to evaluate cardiac structural and functional alterations. Histopathological staining, real-time polymerase chain reaction (PCR) and western blot were performed to evaluate cardiac remodeling. SNX rats showed an HFpEF phenotype with increased BNP, decreased early to late diastolic transmitral flow velocity (E/A) ratio, increased left ventricular (LV) hypertrophy and preserved ejection fraction (EF). Pathology revealed increased cardiac fibrosis in cardiorenal HFpEF rats compared with the Sham group, while chronic treatment with Telmisartan significantly decreased cardiac fibrosis, accompanied by reduced markers of fibrosis (collagen I and collagen III) and profibrotic cytokines (α-smooth muscle actin, transforming growth factor-β1, and connective tissue growth factor). In addition, myocardial inflammation was decreased after Telmisartan treatment, which was in a linear correlation with cardiac fibrosis. Telmisartan also reversed LV hypertrophy and E/A ratio, indicating that Telmisartan can improve LV remodeling and diastolic function in cardiorenal HFpEF. In conclusion, cardiac fibrosis is central to the pathology of cardiorenal HFpEF, and RAAS modulation with Telmisartan is capable of alleviating cardiac fibrosis and preserving diastolic dysfunction in this rat model.
Collapse
Affiliation(s)
- Di Chang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Ting-Ting Xu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Shi-Jun Zhang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Yu Cai
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Shu-Dan Min
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Zhen Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Chun-Qiang Lu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Yuan-Cheng Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| |
Collapse
|
15
|
Odening KE, Gomez AM, Dobrev D, Fabritz L, Heinzel FR, Mangoni ME, Molina CE, Sacconi L, Smith G, Stengl M, Thomas D, Zaza A, Remme CA, Heijman J. ESC working group on cardiac cellular electrophysiology position paper: relevance, opportunities, and limitations of experimental models for cardiac electrophysiology research. Europace 2021; 23:1795-1814. [PMID: 34313298 DOI: 10.1093/europace/euab142] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/19/2021] [Indexed: 12/19/2022] Open
Abstract
Cardiac arrhythmias are a major cause of death and disability. A large number of experimental cell and animal models have been developed to study arrhythmogenic diseases. These models have provided important insights into the underlying arrhythmia mechanisms and translational options for their therapeutic management. This position paper from the ESC Working Group on Cardiac Cellular Electrophysiology provides an overview of (i) currently available in vitro, ex vivo, and in vivo electrophysiological research methodologies, (ii) the most commonly used experimental (cellular and animal) models for cardiac arrhythmias including relevant species differences, (iii) the use of human cardiac tissue, induced pluripotent stem cell (hiPSC)-derived and in silico models to study cardiac arrhythmias, and (iv) the availability, relevance, limitations, and opportunities of these cellular and animal models to recapitulate specific acquired and inherited arrhythmogenic diseases, including atrial fibrillation, heart failure, cardiomyopathy, myocarditis, sinus node, and conduction disorders and channelopathies. By promoting a better understanding of these models and their limitations, this position paper aims to improve the quality of basic research in cardiac electrophysiology, with the ultimate goal to facilitate the clinical translation and application of basic electrophysiological research findings on arrhythmia mechanisms and therapies.
Collapse
Affiliation(s)
- Katja E Odening
- Translational Cardiology, Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland.,Institute of Physiology, University of Bern, Bern, Switzerland
| | - Ana-Maria Gomez
- Signaling and cardiovascular pathophysiology-UMR-S 1180, Inserm, Université Paris-Saclay, 92296 Châtenay-Malabry, France
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.,Department of Cardiology, University Hospital Birmingham NHS Trust, Birmingham, UK
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| | - Matteo E Mangoni
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Cristina E Molina
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site, Hamburg/Kiel/Lübeck, Germany
| | - Leonardo Sacconi
- National Institute of Optics and European Laboratory for Non Linear Spectroscopy, Italy.,Institute for Experimental Cardiovascular Medicine, University Freiburg, Germany
| | - Godfrey Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Milan Stengl
- Department of Physiology, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Dierk Thomas
- Department of Cardiology, University Hospital Heidelberg, Heidelberg, Germany; Heidelberg Center for Heart Rhythm Disorders (HCR), University Hospital Heidelberg, Heidelberg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site, Heidelberg/Mannheim, Germany
| | - Antonio Zaza
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Jordi Heijman
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
16
|
Trum M, Riechel J, Wagner S. Cardioprotection by SGLT2 Inhibitors-Does It All Come Down to Na +? Int J Mol Sci 2021; 22:ijms22157976. [PMID: 34360742 PMCID: PMC8347698 DOI: 10.3390/ijms22157976] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/22/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
Sodium-glucose co-transporter 2 inhibitors (SGLT2i) are emerging as a new treatment strategy for heart failure with reduced ejection fraction (HFrEF) and—depending on the wistfully awaited results of two clinical trials (DELIVER and EMPEROR-Preserved)—may be the first drug class to improve cardiovascular outcomes in patients suffering from heart failure with preserved ejection fraction (HFpEF). Proposed mechanisms of action of this class of drugs are diverse and include metabolic and hemodynamic effects as well as effects on inflammation, neurohumoral activation, and intracellular ion homeostasis. In this review we focus on the growing body of evidence for SGLT2i-mediated effects on cardiac intracellular Na+ as an upstream mechanism. Therefore, we will first give a short overview of physiological cardiomyocyte Na+ handling and its deterioration in heart failure. On this basis we discuss the salutary effects of SGLT2i on Na+ homeostasis by influencing NHE1 activity, late INa as well as CaMKII activity. Finally, we highlight the potential relevance of these effects for systolic and diastolic dysfunction as well as arrhythmogenesis.
Collapse
|
17
|
Wang L, Tian X, Cao Y, Ma X, Shang L, Li H, Zhang X, Deng F, Li S, Guo T, Yang P. Cardiac Shock Wave Therapy Improves Ventricular Function by Relieving Fibrosis Through PI3K/Akt Signaling Pathway: Evidence From a Rat Model of Post-infarction Heart Failure. Front Cardiovasc Med 2021; 8:693875. [PMID: 34222384 PMCID: PMC8241915 DOI: 10.3389/fcvm.2021.693875] [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: 04/12/2021] [Accepted: 05/14/2021] [Indexed: 01/06/2023] Open
Abstract
Objection: Cumulative studies have identified the effectiveness of cardiac shock wave therapy (CSWT) in treating heart failure after acute myocardial infarction (AMI), but little have been discussed with regard to the beneficial effects of CSWT on anti-fibrosis along with the underlying mechanism. In this study, we investigated whether CSWT could reduce post-AMI fibrosis and further explored the molecular mechanism. Methods: Rat heart failure (HF) models induced by ligating the left anterior descending coronary artery were established and validated by echocardiography. Eligible animals were randomly categorized into five groups: the sham group, the HF group, the HF + CSWT group, the HF + LY294002 group, and the HF + CSWT + LY294002 group. The cardiac weight, serum level of BNP, NT-pro BNP and echocardiography parameters were measured to assess cardiac function in different groups. Masson's trichrome staining was used to assess the proportions of the fibrotic area. The expression level of CD34, αSMA was measured by RT-PCR, Immunohistochemistry and Immunofluorescent analyses and the level of PI3K/Akt was quantified by Immunohistochemistry and Western blotting. Results: The application of CSWT significantly improved cardiac function and reduced myocardial fibrosis and level of CD34 and αSMA, compared to the HF group. CSWT led to significant elevations of p-PI3K and p-Akt expression levels compared to that of the HF group and the inhibition of the PI3K/Akt pathway abolished the observed beneficial effects of CSWT. Conclusion: CSWT can facilitate the alleviation of cardiac fibrosis induced by AMI through the activation of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Luqiao Wang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xin Tian
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuting Cao
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xuejuan Ma
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Leilei Shang
- Department of Cardiology, Suizhou Central Hospital, Suizhou, China
| | - Hao Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xueting Zhang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Furong Deng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shumin Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tao Guo
- Department of Cardiology, Yunnan Fuwai Cardiovascular Hospital, Kunming, China
| | - Ping Yang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
18
|
Mishra S, Kass DA. Cellular and molecular pathobiology of heart failure with preserved ejection fraction. Nat Rev Cardiol 2021; 18:400-423. [PMID: 33432192 PMCID: PMC8574228 DOI: 10.1038/s41569-020-00480-6] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2020] [Indexed: 01/30/2023]
Abstract
Heart failure with preserved ejection fraction (HFpEF) affects half of all patients with heart failure worldwide, is increasing in prevalence, confers substantial morbidity and mortality, and has very few effective treatments. HFpEF is arguably the greatest unmet medical need in cardiovascular disease. Although HFpEF was initially considered to be a haemodynamic disorder characterized by hypertension, cardiac hypertrophy and diastolic dysfunction, the pandemics of obesity and diabetes mellitus have modified the HFpEF syndrome, which is now recognized to be a multisystem disorder involving the heart, lungs, kidneys, skeletal muscle, adipose tissue, vascular system, and immune and inflammatory signalling. This multiorgan involvement makes HFpEF difficult to model in experimental animals because the condition is not simply cardiac hypertrophy and hypertension with abnormal myocardial relaxation. However, new animal models involving both haemodynamic and metabolic disease, and increasing efforts to examine human pathophysiology, are revealing new signalling pathways and potential therapeutic targets. In this Review, we discuss the cellular and molecular pathobiology of HFpEF, with the major focus being on mechanisms relevant to the heart, because most research has focused on this organ. We also highlight the involvement of other important organ systems, including the lungs, kidneys and skeletal muscle, efforts to characterize patients with the use of systemic biomarkers, and ongoing therapeutic efforts. Our objective is to provide a roadmap of the signalling pathways and mechanisms of HFpEF that are being characterized and which might lead to more patient-specific therapies and improved clinical outcomes.
Collapse
Affiliation(s)
- Sumita Mishra
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David A. Kass
- Department of Medicine, Division of Cardiology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.,
| |
Collapse
|
19
|
Hegemann N, Primessnig U, Bode D, Wakula P, Beindorff N, Klopfleisch R, Michalick L, Grune J, Hohendanner F, Messroghli D, Pieske B, Kuebler WM, Heinzel FR. Right-ventricular dysfunction in HFpEF is linked to altered cardiomyocyte Ca 2+ homeostasis and myofilament sensitivity. ESC Heart Fail 2021; 8:3130-3144. [PMID: 34002482 PMCID: PMC8318431 DOI: 10.1002/ehf2.13419] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/17/2022] Open
Abstract
Aims Heart failure with preserved ejection fraction (HFpEF) is frequently (30%) associated with right ventricular (RV) dysfunction, which increases morbidity and mortality in these patients. Yet cellular mechanisms of RV remodelling and RV dysfunction in HFpEF are not well understood. Here, we evaluated RV cardiomyocyte function in a rat model of metabolically induced HFpEF. Methods and results Heart failure with preserved ejection fraction‐prone animals (ZSF‐1 obese) and control rats (Wistar Kyoto) were fed a high‐caloric diet for 13 weeks. Haemodynamic characterization by echocardiography and invasive catheterization was performed at 22 and 23 weeks of age, respectively. After sacrifice, organ morphometry, RV histology, isolated RV cardiomyocyte function, and calcium (Ca2+) transients were assessed. ZSF‐1 obese rats showed a HFpEF phenotype with left ventricular (LV) hypertrophy, LV diastolic dysfunction (including increased LV end‐diastolic pressures and E/e′ ratio), and preserved LV ejection fraction. ZSF‐1 obese animals developed RV dilatation (50% increased end‐diastolic area) and mildly impaired RV ejection fraction (42%) with evidence of RV hypertrophy. In isolated RV cardiomyocytes from ZSF‐1 obese rats, cell shortening amplitude was preserved, but cytosolic Ca2+ transient amplitude was reduced. In addition, augmentation of cytosolic Ca2+ release with increased stimulation frequency was lost in ZSF‐1 obese rats. Myofilament sensitivity was increased, while contractile kinetics were largely unaffected in intact isolated RV cardiomyocytes from ZSF‐1 obese rats. Western blot analysis revealed significantly increased phosphorylation of cardiac myosin‐binding protein C (Ser282 cMyBP‐C) but no change in phosphorylation of troponin I (Ser23, 24 TnI) in RV myocardium from ZSF‐1 obese rats. Conclusions Right ventricular dysfunction in obese ZSF‐1 rats with HFpEF is associated with intrinsic RV cardiomyocyte remodelling including reduced cytosolic Ca2+ amplitudes, loss of frequency‐dependent augmentation of Ca2+ release, and increased myofilament Ca2+ sensitivity.
Collapse
Affiliation(s)
- Niklas Hegemann
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Uwe Primessnig
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - David Bode
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Paulina Wakula
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Nicola Beindorff
- Berlin Experimental Radionuclide Imaging Center (BERIC), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, Free University of Berlin, Berlin, Germany
| | - Laura Michalick
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jana Grune
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Hohendanner
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Daniel Messroghli
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany
| | - Wolfgang M Kuebler
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
20
|
Bode D, Rolim NPL, Guthof T, Hegemann N, Wakula P, Primessnig U, Berre AMO, Adams V, Wisløff U, Pieske BM, Heinzel FR, Hohendanner F. Effects of different exercise modalities on cardiac dysfunction in heart failure with preserved ejection fraction. ESC Heart Fail 2021; 8:1806-1818. [PMID: 33768692 PMCID: PMC8120378 DOI: 10.1002/ehf2.13308] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 01/13/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is an increasingly prevalent disease. Physical exercise has been shown to alter disease progression in HFpEF. We examined cardiomyocyte Ca2+ homeostasis and left ventricular function in a metabolic HFpEF model in sedentary and trained rats following 8 weeks of moderate-intensity continuous training (MICT) or high-intensity interval training (HIIT). METHODS AND RESULTS Left ventricular in vivo function (echocardiography) and cardiomyocyte Ca2+ transients (CaTs) (Fluo-4, confocal) were compared in ZSF-1 obese (metabolic syndrome, HFpEF) and ZSF-1 lean (control) 21- and 28-week-old rats. At 21 weeks, cardiomyocytes from HFpEF rats showed prolonged Ca2+ reuptake in cytosolic and nuclear CaTs and impaired Ca2+ release kinetics in nuclear CaTs. At 28 weeks, HFpEF cardiomyocytes had depressed CaT amplitudes, decreased sarcoplasmic reticulum (SR) Ca2+ content, increased SR Ca2+ leak, and elevated diastolic [Ca2+ ] following increased pacing rate (5 Hz). In trained HFpEF rats (HIIT or MICT), cardiomyocyte SR Ca2+ leak was significantly reduced. While HIIT had no effects on the CaTs (1-5 Hz), MICT accelerated early Ca2+ release, reduced the amplitude, and prolonged the CaT without increasing diastolic [Ca2+ ] or cytosolic Ca2+ load at basal or increased pacing rate (1-5 Hz). MICT lowered pro-arrhythmogenic Ca2+ sparks and attenuated Ca2+ -wave propagation in cardiomyocytes. MICT was associated with increased stroke volume in HFpEF. CONCLUSIONS In this metabolic rat model of HFpEF at an advanced stage, Ca2+ release was impaired under baseline conditions. HIIT and MICT differentially affected Ca2+ homeostasis with positive effects of MICT on stroke volume, end-diastolic volume, and cellular arrhythmogenicity.
Collapse
Affiliation(s)
- David Bode
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Natale P L Rolim
- The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Tim Guthof
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany
| | - Niklas Hegemann
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Paulina Wakula
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany
| | - Uwe Primessnig
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Anne Marie Ormbostad Berre
- The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Volker Adams
- Laboratory of Molecular and Experimental Cardiology, TU Dresden, Heart Center Dresden, Dresden, Germany
| | - Ulrik Wisløff
- The Cardiac Exercise Research Group at Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,School of Human Movement and Nutrition Science, University of Queensland, Brisbane, Australia
| | - Burkert M Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Department of Internal Medicine and Cardiology, German Heart Center Berlin, Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Felix Hohendanner
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | | |
Collapse
|
21
|
Miranda-Silva D, Lima T, Rodrigues P, Leite-Moreira A, Falcão-Pires I. Mechanisms underlying the pathophysiology of heart failure with preserved ejection fraction: the tip of the iceberg. Heart Fail Rev 2021; 26:453-478. [PMID: 33411091 DOI: 10.1007/s10741-020-10042-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifaceted syndrome with a complex aetiology often associated with several comorbidities, such as left ventricle pressure overload, diabetes mellitus, obesity, and kidney disease. Its pathophysiology remains obscure mainly due to the complex phenotype induced by all these associated comorbidities and to the scarcity of animal models that adequately mimic HFpEF. Increased oxidative stress, inflammation, and endothelial dysfunction are currently accepted as key players in HFpEF pathophysiology. However, we have just started to unveil HFpEF complexity and the role of calcium handling, energetic metabolism, and mitochondrial function remain to clarify. Indeed, the enlightenment of such cellular and molecular mechanisms represents an opportunity to develop novel therapeutic approaches and thus to improve HFpEF treatment options. In the last decades, the number of research groups dedicated to studying HFpEF has increased, denoting the importance and the magnitude achieved by this syndrome. In the current technological and web world, the amount of information is overwhelming, driving us not only to compile the most relevant information about the theme but also to explore beyond the tip of the iceberg. Thus, this review aims to encompass the most recent knowledge related to HFpEF or HFpEF-associated comorbidities, focusing mainly on myocardial metabolism, oxidative stress, and energetic pathways.
Collapse
Affiliation(s)
- Daniela Miranda-Silva
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Tânia Lima
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Patrícia Rodrigues
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
22
|
Jin G, Manninger M, Adelsmayr G, Schwarzl M, Alogna A, Schönleitner P, Zweiker D, Blaschke F, Sherif M, Radulovic S, Wakula P, Schauer S, Höfler G, Reiter U, Reiter G, Post H, Scherr D, Acsai K, Antoons G, Pieske B, Heinzel FR. Cellular contribution to left and right atrial dysfunction in chronic arterial hypertension in pigs. ESC Heart Fail 2020; 8:151-161. [PMID: 33251761 PMCID: PMC7835565 DOI: 10.1002/ehf2.13087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Aims Atrial contractile dysfunction contributes to worse prognosis in hypertensive heart disease (HHD), but the role of cardiomyocyte dysfunction in atrial remodelling in HHD is not well understood. We investigated and compared cellular mechanisms of left (LA) and right atrial (RA) contractile dysfunction in pigs with HHD. Methods and results In vivo electrophysiological and magnetic resonance imaging studies were performed in control and pigs treated with 11‐deoxycorticosterone acetate (DOCA)/high‐salt/glucose diet (12 weeks) to induce HHD. HHD leads to significant atrial remodelling and loss of contractile function in LA and a similar trend in RA (magnetic resonance imaging). Atrial remodelling was associated with a higher inducibility of atrial fibrillation but unrelated to changes in atrial refractory period or fibrosis (histology). Reduced atrial function in DOCA pigs was related to reduced contraction amplitude of isolated LA (already at baseline) and RA myocytes (at higher frequencies) due to reduced intracellular Ca release (Fura 2‐AM, field stimulation). However, Ca regulation differed in LA and RA cardiomyocytes: LA cardiomyocytes showed reduced sarcoplasmic reticulum (SR) [Ca], whereas in RA, SR [Ca] was unchanged and SR Ca2+‐ATPase activity was increased. Sodium–calcium exchanger (NCX) activity was not significantly altered. We used ORM‐10103 (3 μM), a specific NCX inhibitor to improve Ca availability in LA and RA cardiomyocytes from DOCA pigs. Partial inhibition of NCX increased Ca2+ transient amplitude and SR Ca in LA, but not RA cells. Conclusions In this large animal model of HHD, atrial remodelling in sinus rhythm in vivo was related to differential LA and RA cardiomyocyte dysfunction and Ca signalling. Selective acute inhibition of NCX improved Ca release in diseased LA cardiomyocytes, suggesting a potential therapeutic approach to improve atrial inotropy in HHD.
Collapse
Affiliation(s)
- Ge Jin
- Division of Cardiology, Medical University of Graz, Graz, Austria.,The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Martin Manninger
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | | | - Michael Schwarzl
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Alessio Alogna
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | | | - David Zweiker
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Florian Blaschke
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Mohammad Sherif
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany
| | | | - Paulina Wakula
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Sylvia Schauer
- Department of Pathology, Medical University of Graz, Graz, Austria
| | - Gerald Höfler
- Department of Pathology, Medical University of Graz, Graz, Austria
| | - Ursula Reiter
- Department of Radiology, Medical University of Graz, Graz, Austria
| | - Gert Reiter
- Research & Development, Siemens AG Healthcare, Vienna, Austria
| | - Heiner Post
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Daniel Scherr
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Karoly Acsai
- Division of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Gudrun Antoons
- Faculty of Sciences, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Department of Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
23
|
Heinzel FR, Hegemann N, Hohendanner F, Primessnig U, Grune J, Blaschke F, de Boer RA, Pieske B, Schiattarella GG, Kuebler WM. Left ventricular dysfunction in heart failure with preserved ejection fraction-molecular mechanisms and impact on right ventricular function. Cardiovasc Diagn Ther 2020; 10:1541-1560. [PMID: 33224773 PMCID: PMC7666919 DOI: 10.21037/cdt-20-477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
Abstract
The current classification of heart failure (HF) based on left ventricular (LV) ejection fraction (EF) identifies a large group of patients with preserved ejection fraction (HFpEF) with significant morbidity and mortality but without prognostic benefit from current HF therapy. Co-morbidities and conditions such as arterial hypertension, diabetes mellitus, chronic kidney disease, adiposity and aging shape the clinical phenotype and contribute to mortality. LV diastolic dysfunction and LV structural remodeling are hallmarks of HFpEF, and are linked to remodeling of the cardiomyocyte and extracellular matrix. Pulmonary hypertension (PH) and right ventricular dysfunction (RVD) are particularly common in HFpEF, and mortality is up to 10-fold higher in HFpEF patients with vs. without RV dysfunction. Here, we review alterations in cardiomyocyte function (i.e., ion homeostasis, sarcomere function and cellular metabolism) associated with diastolic dysfunction and summarize the main underlying cellular pathways. The contribution and interaction of systemic and regional upstream signaling such as chronic inflammation, neurohumoral activation, and NO-cGMP-related pathways are outlined in detail, and their diagnostic and therapeutic potential is discussed in the context of preclinical and clinical studies. In addition, we summarize prevalence and pathomechanisms of RV dysfunction in the context of HFpEF and discuss mechanisms connecting LV and RV dysfunction in HFpEF. Dissecting the molecular mechanisms of LV and RV dysfunction in HFpEF may provide a basis for an improved classification of HFpEF and for therapeutic approaches tailored to the molecular phenotype.
Collapse
Affiliation(s)
- Frank R. Heinzel
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Niklas Hegemann
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Hohendanner
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Uwe Primessnig
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Jana Grune
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Blaschke
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Rudolf A. de Boer
- Department of Cardiology, Groningen, University Medical Center Groningen, University of Groningen, The Netherlands
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Department of Internal Medicine and Cardiology, German Heart Center, Berlin, Germany
| | | | - Wolfgang M. Kuebler
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
24
|
Manolis AS, Manolis AA, Manolis TA, Apostolaki NE, Apostolopoulos EJ, Melita H, Katsiki N. Mitochondrial dysfunction in cardiovascular disease: Current status of translational research/clinical and therapeutic implications. Med Res Rev 2020; 41:275-313. [PMID: 32959403 DOI: 10.1002/med.21732] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
Mitochondria provide energy to the cell during aerobic respiration by supplying ~95% of the adenosine triphosphate (ATP) molecules via oxidative phosphorylation. These organelles have various other functions, all carried out by numerous proteins, with the majority of them being encoded by nuclear DNA (nDNA). Mitochondria occupy ~1/3 of the volume of myocardial cells in adults, and function at levels of high-efficiency to promptly meet the energy requirements of the myocardial contractile units. Mitochondria have their own DNA (mtDNA), which contains 37 genes and is maternally inherited. Over the last several years, a variety of functions of these organelles have been discovered and this has led to a growing interest in their involvement in various diseases, including cardiovascular (CV) diseases. Mitochondrial dysfunction relates to the status where mitochondria cannot meet the demands of a cell for ATP and there is an enhanced formation of reactive-oxygen species. This dysfunction may occur as a result of mtDNA and/or nDNA mutations, but also as a response to aging and various disease and environmental stresses, leading to the development of cardiomyopathies and other CV diseases. Designing mitochondria-targeted therapeutic strategies aiming to maintain or restore mitochondrial function has been a great challenge as a result of variable responses according to the etiology of the disorder. There have been several preclinical data on such therapies, but clinical studies are scarce. A major challenge relates to the techniques needed to eclectically deliver the therapeutic agents to cardiac tissues and to damaged mitochondria for successful clinical outcomes. All these issues and progress made over the last several years are herein reviewed.
Collapse
Affiliation(s)
- Antonis S Manolis
- First Department of Cardiology, Athens University School of Medicine, Athens, Greece
| | | | | | | | | | | | - Niki Katsiki
- First Department of Internal Medicine, Division of Endocrinology and Metabolism, Diabetes Center, Medical School, AHEPA University Hospital, Thessaloniki, Greece
| |
Collapse
|
25
|
Djalinac N, Ljubojevic-Holzer S, Matzer I, Kolesnik E, Jandl K, Lohberger B, Rainer P, Heinemann A, Sedej S, von Lewinski D, Bisping E. The role of stretch, tachycardia and sodium-calcium exchanger in induction of early cardiac remodelling. J Cell Mol Med 2020; 24:8732-8743. [PMID: 32573098 PMCID: PMC7412684 DOI: 10.1111/jcmm.15504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/20/2020] [Accepted: 05/24/2020] [Indexed: 12/14/2022] Open
Abstract
Stretch and tachycardia are common triggers for cardiac remodelling in various conditions, but a comparative characterization of their role in the excitation‐transcription coupling (ETC) and early regulation of gene expression and structural changes is lacking. Here, we show that stretch and tachycardia directly induced hypertrophy of neonatal rat cardiac myocytes and also of non‐myocytes. Both triggers induced similar patterns of hypertrophy but had largely distinct gene expression profiles. ACTA1 served as good hypertrophy marker upon stretch, while RCAN1 was found increased in response to tachycardia in a rate‐dependent fashion. Mechanistically, several calcium‐handling proteins, including the sodium‐calcium exchanger (NCX), contributed to ETC. Phosphorylation of the calcium/calmodulin‐dependent protein kinase II (CaMKII) was elevated and occurred downstream of NCX activation upon tachycardia, but not stretch. Microarray profiling revealed that stretch and tachycardia regulated around 33% and 20% genes in a NCX‐dependent manner, respectively. In conclusion, our data show that hypertrophy induction by stretch and tachycardia is associated with different gene expression profiles with a significant contribution of the NCX.
Collapse
Affiliation(s)
- Natasa Djalinac
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | | | - Ingrid Matzer
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Ewald Kolesnik
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Katharina Jandl
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria.,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Birgit Lohberger
- Department of Orthopedics and Trauma, Medical University of Graz, Graz, Austria
| | - Peter Rainer
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | | | - Egbert Bisping
- Department of Cardiology, Medical University of Graz, Graz, Austria
| |
Collapse
|
26
|
Eisner DA, Caldwell JL, Trafford AW, Hutchings DC. The Control of Diastolic Calcium in the Heart: Basic Mechanisms and Functional Implications. Circ Res 2020; 126:395-412. [PMID: 31999537 PMCID: PMC7004450 DOI: 10.1161/circresaha.119.315891] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Normal cardiac function requires that intracellular Ca2+ concentration be reduced to low levels in diastole so that the ventricle can relax and refill with blood. Heart failure is often associated with impaired cardiac relaxation. Little, however, is known about how diastolic intracellular Ca2+ concentration is regulated. This article first discusses the reasons for this ignorance before reviewing the basic mechanisms that control diastolic intracellular Ca2+ concentration. It then considers how the control of systolic and diastolic intracellular Ca2+ concentration is intimately connected. Finally, it discusses the changes that occur in heart failure and how these may result in heart failure with preserved versus reduced ejection fraction.
Collapse
Affiliation(s)
- David A Eisner
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Jessica L Caldwell
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - Andrew W Trafford
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| | - David C Hutchings
- From the Unit of Cardiac Physiology, Division of Cardiovascular Sciences, University of Manchester, United Kingdom
| |
Collapse
|
27
|
Primessnig U, Bracic T, Levijoki J, Otsomaa L, Pollesello P, Falcke M, Pieske B, Heinzel FR. Long-term effects of Na + /Ca 2+ exchanger inhibition with ORM-11035 improves cardiac function and remodelling without lowering blood pressure in a model of heart failure with preserved ejection fraction. Eur J Heart Fail 2019; 21:1543-1552. [PMID: 31762174 DOI: 10.1002/ejhf.1619] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 08/26/2019] [Accepted: 09/02/2019] [Indexed: 12/28/2022] Open
Abstract
AIMS Heart failure with preserved ejection fraction (HFpEF) is increasingly common but there is currently no established pharmacological therapy. We hypothesized that ORM-11035, a novel specific Na+ /Ca2+ exchanger (NCX) inhibitor, improves cardiac function and remodelling independent of effects on arterial blood pressure in a model of cardiorenal HFpEF. METHODS AND RESULTS Rats were subjected to subtotal nephrectomy (NXT) or sham operation. Eight weeks after intervention, treatment for 16 weeks with ORM-11035 (1 mg/kg body weight) or vehicle was initiated. At 24 weeks, blood pressure measurements, echocardiography and pressure-volume loops were performed. Contractile function, Ca2+ transients and NCX-mediated Ca2+ extrusion were measured in isolated ventricular cardiomyocytes. NXT rats (untreated) showed a HFpEF phenotype with left ventricular (LV) hypertrophy, LV end-diastolic pressure (LVEDP) elevation, increased brain natriuretic peptide (BNP) levels, preserved ejection fraction and pulmonary congestion. In cardiomyocytes from untreated NXT rats, early relaxation was prolonged and NCX-mediated Ca2+ extrusion was decreased. Chronic treatment with ORM-11035 significantly reduced LV hypertrophy and cardiac remodelling without lowering systolic blood pressure. LVEDP [14 ± 3 vs. 9 ± 2 mmHg; NXT (n = 12) vs. NXT + ORM (n = 12); P = 0.0002] and BNP levels [71 ± 12 vs. 49 ± 11 pg/mL; NXT (n = 12) vs. NXT + ORM (n = 12); P < 0.0001] were reduced after ORM treatment. LV cardiomyocytes from ORM-treated rats showed improved active relaxation and diastolic cytosolic Ca2+ decay as well as restored NCX-mediated Ca2+ removal, indicating NCX modulation with ORM-11035 as a promising target in the treatment of HFpEF. CONCLUSION Chronic inhibition of NCX with ORM-11035 significantly attenuated cardiac remodelling and diastolic dysfunction without lowering systemic blood pressure in this model of HFpEF. Therefore, long-term treatment with selective NCX inhibitors such as ORM-11035 should be evaluated further in the treatment of heart failure.
Collapse
Affiliation(s)
- Uwe Primessnig
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,Department of Cardiology, Medical University of Graz, Graz, Austria.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Taja Bracic
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | | | | | | | - Martin Falcke
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.,Department of Physics, Humboldt Universität, Berlin, Germany
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site, Berlin, Germany
| |
Collapse
|
28
|
Gollmann-Tepeköylü C, Lobenwein D, Theurl M, Primessnig U, Lener D, Kirchmair E, Mathes W, Graber M, Pölzl L, An A, Koziel K, Pechriggl E, Voelkl J, Paulus P, Schaden W, Grimm M, Kirchmair R, Holfeld J. Shock Wave Therapy Improves Cardiac Function in a Model of Chronic Ischemic Heart Failure: Evidence for a Mechanism Involving VEGF Signaling and the Extracellular Matrix. J Am Heart Assoc 2019; 7:e010025. [PMID: 30371289 PMCID: PMC6474945 DOI: 10.1161/jaha.118.010025] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Mechanical stimulation of acute ischemic myocardium by shock wave therapy (SWT) is known to improve cardiac function by induction of angiogenesis. However, SWT in chronic heart failure is poorly understood. We aimed to study whether mechanical stimulation upon SWT improves heart function in chronic ischemic heart failure by induction of angiogenesis and postnatal vasculogenesis and to dissect underlying mechanisms. Methods and Results SWT was applied in a mouse model of chronic myocardial ischemia. To study effects of SWT on postnatal vasculogenesis, wild‐type mice received bone marrow transplantation from green fluorescence protein donor mice. Underlying mechanisms were elucidated in vitro in endothelial cells and murine aortic rings. Echocardiography and pressure/volume measurements revealed improved left ventricular ejection fraction, myocardial contractility, and diastolic function and decreased myocardial fibrosis after treatment. Concomitantly, numbers of capillaries and arterioles were increased. SWT resulted in enhanced expression of the chemoattractant stromal cell–derived factor 1 in ischemic myocardium and serum. Treatment induced recruitment of bone marrow–derived endothelial cells to the site of injury. In vitro, SWT resulted in endothelial cell proliferation, enhanced survival, and capillary sprouting. The effects were vascular endothelial growth factor receptor 2 and heparan sulfate proteoglycan dependent. Conclusions SWT positively affects heart function in chronic ischemic heart failure by induction of angiogenesis and postnatal vasculogenesis. SWT upregulated pivotal angiogenic and vasculogenic factors in the myocardium in vivo and induced proliferative and anti‐apoptotic effects on endothelial cells in vitro. Mechanistically, these effects depend on vascular endothelial growth factor signaling and heparan sulfate proteoglycans. SWT is a promising treatment option for regeneration of ischemic myocardium.
Collapse
Affiliation(s)
| | | | - Markus Theurl
- 3 Internal Medicine III Medical University of Innsbruck Austria
| | - Uwe Primessnig
- 4 Department of Internal Medicine and Cardiology Charité - Universitätsmedizin Berlin Germany
| | - Daniela Lener
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | - Elke Kirchmair
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | | | - Michael Graber
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | - Leo Pölzl
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | - Angela An
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | | | - Elisabeth Pechriggl
- 1 Division of Clinical and Functional Anatomy Department of Anatomy, Histology and Embryology Medical University of Innsbruck Austria
| | - Jakob Voelkl
- 4 Department of Internal Medicine and Cardiology Charité - Universitätsmedizin Berlin Germany
| | - Patrick Paulus
- 5 Department of Anaesthesiology and Operative Intensive Care Medicine Kepler University Hospital Linz Austria
| | - Wolfgang Schaden
- 6 Ludwig Boltzmann Institute for Experimental and Clinical Traumatology AUVA Research Centre Vienna Austria.,7 Austrian Cluster for Tissue Regeneration Vienna Austria
| | - Michael Grimm
- 2 Cardiac Surgery Medical University of Innsbruck Austria
| | | | - Johannes Holfeld
- 2 Cardiac Surgery Medical University of Innsbruck Austria.,7 Austrian Cluster for Tissue Regeneration Vienna Austria
| |
Collapse
|
29
|
Mavrakanas T, Khattak A, Wang W, Singh K, Charytan D. Association of Chronic Kidney Disease with Preserved Ejection Fraction Heart Failure Is Independent of Baseline Cardiac Function. Kidney Blood Press Res 2019; 44:1247-1258. [DOI: 10.1159/000502874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background/Aims: Chronic kidney disease (CKD) is common among patients with heart failure with preserved ejection fraction (HFpEF) and is associated with worse clinical outcomes. This study aims to identify whether the association of CKD with HFpEF is independent of underlying echocardiographic abnormalities. Materials: We conducted a retrospective cohort study including patients without prevalent heart failure referred for echocardiography. Patients with serial echocardiograms, baseline left ventricular ejection fraction (LVEF) ≥50% and estimated glomerular filtration rate (eGFR) ≥90 mL/min/1.73 m2 were matched 1:1 with patients with eGFR <60 mL/min/1.73 m2 for age (±5 years), sex, history of hypertension or diabetes, use of renin-angiotensin inhibitors, and LVEF (±5%). A secondary analysis included patients with preserved LVEF and normal left ventricular mass index matched for the same parameters except use of renin-angiotensin inhibitors. Results: Patients with CKD were at increased risk for HFpEF admission: crude hazard ratio (HR) 1.79 (95% confidence interval [CI] 1.38–2.32, p < 0.001) and adjusted HR (for coronary disease, loop diuretics, left atrial diameter) 1.64 (95% CI 1.22–2.21, p = 0.001). LVEF and left ventricular diameter decreased over time in both groups but no difference was observed in rate of dropping. Results were similar in the secondary analysis (crude HR 1.99, 95% CI 1.07–3.71, p = 0.03 and HR adjusted for left atrial diameter 1.98, 95% CI 1.05–3.75, p = 0.04). Rate of change was similar for LVEF, pulmonary artery pressure, and left ventricular mass index in both groups. Conclusion: CKD is independently associated with incident HFpEF despite a similar change in relevant echocardiographic parameters in patients with or without CKD.
Collapse
|
30
|
Olver TD, Edwards JC, Jurrissen TJ, Veteto AB, Jones JL, Gao C, Rau C, Warren CM, Klutho PJ, Alex L, Ferreira-Nichols SC, Ivey JR, Thorne PK, McDonald KS, Krenz M, Baines CP, Solaro RJ, Wang Y, Ford DA, Domeier TL, Padilla J, Rector RS, Emter CA. Western Diet-Fed, Aortic-Banded Ossabaw Swine: A Preclinical Model of Cardio-Metabolic Heart Failure. JACC Basic Transl Sci 2019; 4:404-421. [PMID: 31312763 PMCID: PMC6610000 DOI: 10.1016/j.jacbts.2019.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/13/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022]
Abstract
The development of new treatments for heart failure lack animal models that encompass the increasingly heterogeneous disease profile of this patient population. This report provides evidence supporting the hypothesis that Western Diet-fed, aortic-banded Ossabaw swine display an integrated physiological, morphological, and genetic phenotype evocative of cardio-metabolic heart failure. This new preclinical animal model displays a distinctive constellation of findings that are conceivably useful to extending the understanding of how pre-existing cardio-metabolic syndrome can contribute to developing HF.
Collapse
Key Words
- AB, aortic-banded
- CON, control
- EDPVR, end-diastolic pressure−volume relationship
- EF, ejection fraction
- HF, heart failure
- HFpEF, heart failure with preserved ejection fraction
- HFrEF, heart failure with reduced ejection fraction
- IL1RL1, interleukin 1 receptor-like 1
- LV, left ventricle
- NF, nuclear factor
- PTX3, pentraxin-3
- WD, Western Diet
- cardio-metabolic disease
- heart failure
- integrative pathophysiology
- preclinical model of cardiovascular disease
Collapse
Affiliation(s)
- T. Dylan Olver
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Jenna C. Edwards
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Thomas J. Jurrissen
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Adam B. Veteto
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - John L. Jones
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Chen Gao
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Christoph Rau
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Chad M. Warren
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Paula J. Klutho
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Linda Alex
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | | | - Jan R. Ivey
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Pamela K. Thorne
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| | - Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Christopher P. Baines
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - R. John Solaro
- Department of Physiology and Biophysics, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, Illinois
| | - Yibin Wang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - David A. Ford
- Department of Biochemistry and Molecular Biology and Center for Cardiovascular Research, Saint Louis University- School of Medicine, St. Louis, Missouri
| | - Timothy L. Domeier
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
- Department of Child Health, University of Missouri-Columbia, Columbia, Missouri
| | - R. Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri-Columbia, Columbia, Missouri
- Department of Medicine – University of Missouri-Columbia, Columbia, Missouri
- Research Service, Harry S Truman Memorial VA Hospital, University of Missouri-Columbia, Columbia, Missouri
| | - Craig A. Emter
- Department of Biomedical Science, University of Missouri-Columbia, Columbia, Missouri
| |
Collapse
|
31
|
Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, Luo X, Jiang N, May HI, Wang ZV, Hill TM, Mammen PPA, Huang J, Lee DI, Hahn VS, Sharma K, Kass DA, Lavandero S, Gillette TG, Hill JA. Nitrosative stress drives heart failure with preserved ejection fraction. Nature 2019; 568:351-356. [PMID: 30971818 PMCID: PMC6635957 DOI: 10.1038/s41586-019-1100-z] [Citation(s) in RCA: 493] [Impact Index Per Article: 98.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a common syndrome with high morbidity and mortality for which there are no evidence-based therapies. Here we report that concomitant metabolic and hypertensive stress in mice-elicited by a combination of high-fat diet and inhibition of constitutive nitric oxide synthase using Nω-nitro-L-arginine methyl ester (L-NAME)-recapitulates the numerous systemic and cardiovascular features of HFpEF in humans. Expression of one of the unfolded protein response effectors, the spliced form of X-box-binding protein 1 (XBP1s), was reduced in the myocardium of our rodent model and in humans with HFpEF. Mechanistically, the decrease in XBP1s resulted from increased activity of inducible nitric oxide synthase (iNOS) and S-nitrosylation of the endonuclease inositol-requiring protein 1α (IRE1α), culminating in defective XBP1 splicing. Pharmacological or genetic suppression of iNOS, or cardiomyocyte-restricted overexpression of XBP1s, each ameliorated the HFpEF phenotype. We report that iNOS-driven dysregulation of the IRE1α-XBP1 pathway is a crucial mechanism of cardiomyocyte dysfunction in HFpEF.
Collapse
Affiliation(s)
- Gabriele G Schiattarella
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Francisco Altamirano
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dan Tong
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kristin M French
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elisa Villalobos
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Soo Young Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nan Jiang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Herman I May
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Theodore M Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pradeep P A Mammen
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jian Huang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dong I Lee
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sergio Lavandero
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Molecular Studies of the Cell (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
32
|
Rouhana S, Farah C, Roy J, Finan A, Rodrigues de Araujo G, Bideaux P, Scheuermann V, Saliba Y, Reboul C, Cazorla O, Aimond F, Richard S, Thireau J, Fares N. Early calcium handling imbalance in pressure overload-induced heart failure with nearly normal left ventricular ejection fraction. Biochim Biophys Acta Mol Basis Dis 2019; 1865:230-242. [DOI: 10.1016/j.bbadis.2018.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 07/13/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023]
|
33
|
Metra M. December 2018 at a glance: new imaging methods, translational cardiology and non-medical treatment. Eur J Heart Fail 2018; 20:1613-1614. [PMID: 30561162 DOI: 10.1002/ejhf.1016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Italy
| |
Collapse
|
34
|
Barandiarán Aizpurua A, Schroen B, van Bilsen M, van Empel V. Targeted HFpEF therapy based on matchmaking of human and animal models. Am J Physiol Heart Circ Physiol 2018; 315:H1670-H1683. [PMID: 30239232 DOI: 10.1152/ajpheart.00024.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The diversity in clinical phenotypes and poor understanding of the underlying pathophysiology of heart failure with preserved ejection fraction (HFpEF) is the main reason why no effective treatments have been found yet. Targeted, instead of one size fits all, treatment seems the only promising approach for treating HFpEF. To be able to design a targeted, phenotype-specific HFpEF treatment, the matrix relating clinical phenotypes and underlying pathophysiological mechanisms has to be clarified. This review discusses the opportunities for additional evaluation of the underlying pathophysiological processes, e.g., to evaluate biological phenotypes on top of clinical routine, to guide us toward a phenotype-specific HFpEF treatment. Moreover, a translational approach with matchmaking of animal models to biological HFpEF phenotypes will be a valuable step to test the effectiveness of novel, targeted interventions in HFpEF. Listen to this article's corresponding podcast at https://ajpheart.podbean.com/e/personalized-medicine-in-hfpef/ .
Collapse
Affiliation(s)
- Arantxa Barandiarán Aizpurua
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Blanche Schroen
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre , Maastricht , The Netherlands
| | - Marc van Bilsen
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Department of Physiology, Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases, Maastricht University , Maastricht , The Netherlands
| | - Vanessa van Empel
- Department of Cardiology, Maastricht University Medical Centre , Maastricht , The Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre , Maastricht , The Netherlands
| |
Collapse
|
35
|
Michalska-Kasiczak M, Bielecka-Dabrowa A, von Haehling S, Anker SD, Rysz J, Banach M. Biomarkers, myocardial fibrosis and co-morbidities in heart failure with preserved ejection fraction: an overview. Arch Med Sci 2018; 14:890-909. [PMID: 30002709 PMCID: PMC6040115 DOI: 10.5114/aoms.2018.76279] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is steadily increasing. Its diagnosis remains difficult and controversial and relies mostly on non-invasive echocardiographic detection of left ventricular diastolic dysfunction and elevated filling pressures. The large phenotypic heterogeneity of HFpEF from pathophysiologic al underpinnings to clinical manifestations presents a major obstacle to the development of new therapies targeted towards specific HF phenotypes. Recent studies suggest that natriuretic peptides have the potential to improve the diagnosis of early HFpEF, but they still have significant limitations, and the cut-off points for diagnosis and prognosis in HFpEF remain open to debate. The purpose of this review is to present potential targets of intervention in patients with HFpEF, starting with myocardial fibrosis and methods of its detection. In addition, co-morbidities are discussed as a means to treat HFpEF according to cut-points of biomarkers that are different from usual. Biomarkers and approaches to co-morbidities may be able to tailor therapies according to patients' pathophysiological needs. Recently, soluble source of tumorigenicity 2 (sST2), growth differentiation factor 15 (GDF-15), galectin-3, and other cardiac markers have emerged, but evidence from large cohorts is still lacking. Furthermore, the field of miRNA is a very promising area of research, and further exploration of miRNA may offer diagnostic and prognostic applications and insight into the pathology, pointing to new phenotype-specific therapeutic targets.
Collapse
Affiliation(s)
- Marta Michalska-Kasiczak
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
- Department of Endocrine Disorders and Bone Metabolism, 1 Chair of Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Agata Bielecka-Dabrowa
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Göttingen, Germany
| | - Stefan D. Anker
- Division of Cardiology and Metabolism – Heart Failure, Cachexia and Sarcopenia, Department of Cardiology, Campus Virchow-Klinikum, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
36
|
Curl CL, Danes VR, Bell JR, Raaijmakers AJA, Ip WTK, Chandramouli C, Harding TW, Porrello ER, Erickson JR, Charchar FJ, Kompa AR, Edgley AJ, Crossman DJ, Soeller C, Mellor KM, Kalman JM, Harrap SB, Delbridge LMD. Cardiomyocyte Functional Etiology in Heart Failure With Preserved Ejection Fraction Is Distinctive-A New Preclinical Model. J Am Heart Assoc 2018; 7:JAHA.117.007451. [PMID: 29858360 PMCID: PMC6015350 DOI: 10.1161/jaha.117.007451] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Among the growing numbers of patients with heart failure, up to one half have heart failure with preserved ejection fraction (HFpEF). The lack of effective treatments for HFpEF is a substantial and escalating unmet clinical need—and the lack of HFpEF‐specific animal models represents a major preclinical barrier in advancing understanding of HFpEF. As established treatments for heart failure with reduced ejection fraction (HFrEF) have proven ineffective for HFpEF, the contention that the intrinsic cardiomyocyte phenotype is distinct in these 2 conditions requires consideration. Our goal was to validate and characterize a new rodent model of HFpEF, undertaking longitudinal investigations to delineate the associated cardiac and cardiomyocyte pathophysiology. Methods and Results The selectively inbred Hypertrophic Heart Rat (HHR) strain exhibits adult cardiac enlargement (without hypertension) and premature death (40% mortality at 50 weeks) compared to its control strain, the normal heart rat. Hypertrophy was characterized in vivo by maintained systolic parameters (ejection fraction at 85%–90% control) with marked diastolic dysfunction (increased E/E′). Surprisingly, HHR cardiomyocytes were hypercontractile, exhibiting high Ca2+ operational levels and markedly increased L‐type Ca2+ channel current. In HHR, prominent regions of reparative fibrosis in the left ventricle free wall adjacent to the interventricular septum were observed. Conclusions Thus, the cardiomyocyte remodeling process in the etiology of this HFpEF model contrasts dramatically with the suppressed Ca2+ cycling state that typifies heart failure with reduced ejection fraction. These findings may explain clinical observations, that treatments considered appropriate for heart failure with reduced ejection fraction are of little benefit for HFpEF—and suggest a basis for new therapeutic strategies.
Collapse
Affiliation(s)
- Claire L Curl
- Department of Physiology, University of Melbourne, Victoria, Australia
| | - Vennetia R Danes
- Department of Physiology, University of Melbourne, Victoria, Australia
| | - James R Bell
- Department of Physiology, University of Melbourne, Victoria, Australia
| | | | - Wendy T K Ip
- Department of Physiology, University of Melbourne, Victoria, Australia
| | | | - Tristan W Harding
- Department of Physiology, University of Melbourne, Victoria, Australia
| | - Enzo R Porrello
- Department of Physiology, University of Melbourne, Victoria, Australia.,Murdoch Children's Research Institute, Melbourne, Australia
| | | | - Fadi J Charchar
- School of Applied & Biomedical Sciences, Federation University, Ballarat, Australia
| | - Andrew R Kompa
- Department of Medicine, St. Vincent's Hospital The University of Melbourne, Australia
| | - Amanda J Edgley
- Department of Medicine, St. Vincent's Hospital The University of Melbourne, Australia
| | | | | | | | - Jonathan M Kalman
- Department of Medicine, University of Melbourne, Victoria, Australia
| | - Stephen B Harrap
- Department of Physiology, University of Melbourne, Victoria, Australia
| | - Lea M D Delbridge
- Department of Physiology, University of Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Røe ÅT, Aronsen JM, Skårdal K, Hamdani N, Linke WA, Danielsen HE, Sejersted OM, Sjaastad I, Louch WE. Increased passive stiffness promotes diastolic dysfunction despite improved Ca2+ handling during left ventricular concentric hypertrophy. Cardiovasc Res 2018; 113:1161-1172. [PMID: 28472418 PMCID: PMC5852536 DOI: 10.1093/cvr/cvx087] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/03/2017] [Indexed: 02/06/2023] Open
Abstract
Aims Concentric hypertrophy following pressure-overload is linked to preserved systolic function but impaired diastolic function, and is an important substrate for heart failure with preserved ejection fraction. While increased passive stiffness of the myocardium is a suggested mechanism underlying diastolic dysfunction in these hearts, the contribution of active diastolic Ca2+ cycling in cardiomyocytes remains unclear. In this study, we sought to dissect contributions of passive and active mechanisms to diastolic dysfunction in the concentrically hypertrophied heart following pressure-overload. Methods and results Rats were subjected to aortic banding (AB), and experiments were performed 6 weeks after surgery using sham-operated rats as controls. In vivo ejection fraction and fractional shortening were normal, confirming preservation of systolic function. Left ventricular concentric hypertrophy and diastolic dysfunction following AB were indicated by thickening of the ventricular wall, reduced peak early diastolic tissue velocity, and higher E/e' values. Slowed relaxation was also observed in left ventricular muscle strips isolated from AB hearts, during both isometric and isotonic stimulation, and accompanied by increases in passive tension, viscosity, and extracellular collagen. An altered titin phosphorylation profile was observed with hypophosphorylation of the phosphosites S4080 and S3991 sites within the N2Bus, and S12884 within the PEVK region. Increased titin-based stiffness was confirmed by salt-extraction experiments. In contrast, isolated, unloaded cardiomyocytes exhibited accelerated relaxation in AB compared to sham, and less contracture at high pacing frequencies. Parallel enhancement of diastolic Ca2+ handling was observed, with augmented NCX and SERCA2 activity and lowered resting cytosolic [Ca2+]. Conclusion In the hypertrophied heart with preserved systolic function, in vivo diastolic dysfunction develops as cardiac fibrosis and alterations in titin phosphorylation compromise left ventricular compliance, and despite compensatory changes in cardiomyocyte Ca2+ homeostasis.
Collapse
MESH Headings
- Adaptation, Physiological
- Animals
- Aorta/physiopathology
- Aorta/surgery
- Arterial Pressure
- Calcium/metabolism
- Calcium Signaling
- Collagen/metabolism
- Compliance
- Connectin/metabolism
- Constriction
- Diastole
- Disease Models, Animal
- Fibrosis
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Isolated Heart Preparation
- Male
- Myocardium/metabolism
- Myocardium/pathology
- Phosphorylation
- Rats, Wistar
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Sodium-Calcium Exchanger/metabolism
- Systole
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Åsmund T. Røe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
- Corresponding author. Tel: +47 23 01 68 00; fax: +47 23 01 67 99, E-mail:
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Kristine Skårdal
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| | - Nazha Hamdani
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Wolfgang A. Linke
- Department of Cardiovascular Physiology, Ruhr University Bochum, Bochum, Germany
| | - Håvard E. Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, UK
| | - Ole M. Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, NO-0407 Oslo, Norway
| |
Collapse
|
38
|
Neef S, Steffens A, Pellicena P, Mustroph J, Lebek S, Ort KR, Schulman H, Maier LS. Improvement of cardiomyocyte function by a novel pyrimidine-based CaMKII-inhibitor. J Mol Cell Cardiol 2017; 115:73-81. [PMID: 29294328 DOI: 10.1016/j.yjmcc.2017.12.015] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 12/12/2017] [Accepted: 12/29/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Pathologically increased activity of Ca2+/calmodulin-dependent protein kinase II (CaMKII) and the associated Ca2+-leak from the sarcoplasmic reticulum are recognized to be important novel pharmacotherapeutic targets in heart failure and cardiac arrhythmias. However, CaMKII-inhibitory compounds for therapeutic use are still lacking. We now report on the cellular and molecular effects of a novel pyrimidine-based CaMKII inhibitor developed towards clinical use. METHODS AND RESULTS Our findings demonstrate that AS105 is a high-affinity ATP-competitive CaMKII-inhibitor that by its mode of action is also effective against autophosphorylated CaMKII (in contrast to the commonly used allosteric CaMKII-inhibitor KN-93). In isolated atrial cardiomyocytes from human donors and ventricular myocytes from CaMKIIδC-overexpressing mice with heart failure, AS105 effectively reduced diastolic SR Ca2+ leak by 38% to 65% as measured by Ca2+-sparks or tetracaine-sensitive shift in [Ca2+]i. Consistent with this, we found that AS105 suppressed arrhythmogenic spontaneous cardiomyocyte Ca2+-release (by 53%). Also, the ability of the SR to accumulate Ca2+ was enhanced by AS105, as indicated by improved post-rest potentiation of Ca2+-transient amplitudes and increased SR Ca2+-content in the murine cells. Accordingly, these cells had improved systolic Ca2+-transient amplitudes and contractility during basal stimulation. Importantly, CaMKII inhibition did not compromise systolic fractional Ca2+-release, diastolic SR Ca2+-reuptake via SERCA2a or Ca2+-extrusion via NCX. CONCLUSION AS105 is a novel, highly potent ATP-competitive CaMKII inhibitor. In vitro, it effectively reduced SR Ca2+-leak, thus improving SR Ca2+-accumulation and reducing cellular arrhythmogenic correlates, without negatively influencing excitation-contraction coupling. These findings further validate CaMKII as a key target in cardiovascular disease, implicated by genetic, allosteric inhibitors, and pseudo-substrate inhibitors.
Collapse
Affiliation(s)
- Stefan Neef
- Dept. of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Alexander Steffens
- Dept. of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Julian Mustroph
- Dept. of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Simon Lebek
- Dept. of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Katharina R Ort
- Dept. of Thoracic, Cardiac and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
| | | | - Lars S Maier
- Dept. of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany.
| |
Collapse
|
39
|
Cellular mechanisms of metabolic syndrome-related atrial decompensation in a rat model of HFpEF. J Mol Cell Cardiol 2017; 115:10-19. [PMID: 29289652 DOI: 10.1016/j.yjmcc.2017.12.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/30/2017] [Accepted: 12/27/2017] [Indexed: 11/23/2022]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is present in about 50% of HF patients. Atrial remodeling is common in HFpEF and associated with increased mortality. We postulate that atrial remodeling is associated with atrial dysfunction in vivo related to alterations in cardiomyocyte Calcium (Ca) signaling and remodeling. We examined atrial function in vivo and Ca transients (CaT) (Fluo4-AM, field stim) in atrial cardiomyocytes of ZSF-1 rats without (Ln; lean hypertensive) and with metabolic syndrome (Ob; obese, hypertensive, diabetic) and HFpEF. RESULTS At 21weeks Ln showed an increased left ventricular (LV) mass and left ventricular end-diastolic pressure (LVEDP), but unchanged left atrial (LA) size and preserved atrial ejection fraction vs. wild-type (WT). CaT amplitude in atrial cardiomyocytes was increased in Ln (2.9±0.2 vs. 2.3±0.2F/F0 in WT; n=22 cells/group; p<0.05). Studying subcellular Ca release in more detail, we found that local central cytosolic CaT amplitude was increased, while subsarcolemmal CaT amplitudes remained unchanged. Moreover, Sarcoplasmic reticulum (SR) Ca content (caffeine) was preserved while Ca spark frequency and tetracaine-dependent SR Ca leak were significantly increased in Ln. Ob mice developed a HFpEF phenotype in vivo, LA area was significantly increased and atrial in vivo function was impaired, despite increased atrial CaT amplitudes in vitro (2.8±0.2; p<0.05 vs. WT). Ob cells showed alterations of the tubular network possibly contributing to the observed phenotype. CaT kinetics as well as SR Ca in Ob were not significantly different from WT, but SR Ca leak remained increased. Angiotensin II (Ang II) reduced in vitro cytosolic CaT amplitudes and let to active nuclear Ca release in Ob but not in Ln or WT. SUMMARY In hypertensive ZSF-1 rats, a possibly compensatory increase of cytosolic CaT amplitude and increased SR Ca leak precede atrial remodeling and HFpEF. Atrial remodeling in ZSF-1 HFpEF is associated with an altered tubular network in-vitro and atrial contractile dysfunction in vivo, indicating insufficient compensation. Atrial cardiomyocyte dysfunction in vitro is induced by the addition of angiotensin II.
Collapse
|
40
|
Lewis GA, Schelbert EB, Williams SG, Cunnington C, Ahmed F, McDonagh TA, Miller CA. Biological Phenotypes of Heart Failure With Preserved Ejection Fraction. J Am Coll Cardiol 2017; 70:2186-2200. [PMID: 29050567 DOI: 10.1016/j.jacc.2017.09.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 09/05/2017] [Accepted: 09/05/2017] [Indexed: 12/19/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) involves multiple pathophysiological mechanisms, which result in the heterogeneous phenotypes that are evident clinically, and which have potentially confounded previous HFpEF trials. A greater understanding of the in vivo human processes involved, and in particular, which are the causes and which are the downstream effects, may allow the syndrome of HFpEF to be distilled into distinct diagnoses based on the underlying biology. From this, specific interventions can follow, targeting individuals identified on the basis of their biological phenotype. This review describes the biological phenotypes of HFpEF and therapeutic interventions aimed at targeting these phenotypes.
Collapse
Affiliation(s)
- Gavin A Lewis
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, United Kingdom; University Hospital of South Manchester NHS Foundation Trust, Wythenshawe, Manchester, United Kingdom
| | - Erik B Schelbert
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, Pennsylvania; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Simon G Williams
- University Hospital of South Manchester NHS Foundation Trust, Wythenshawe, Manchester, United Kingdom
| | - Colin Cunnington
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, United Kingdom; Manchester Heart Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Oxford Road, Manchester, United Kingdom
| | - Fozia Ahmed
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, United Kingdom; Manchester Heart Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Sciences Centre, Oxford Road, Manchester, United Kingdom
| | | | - Christopher A Miller
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, United Kingdom; University Hospital of South Manchester NHS Foundation Trust, Wythenshawe, Manchester, United Kingdom; Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, United Kingdom.
| |
Collapse
|
41
|
Tschöpe C, Birner C, Böhm M, Bruder O, Frantz S, Luchner A, Maier L, Störk S, Kherad B, Laufs U. Heart failure with preserved ejection fraction: current management and future strategies : Expert opinion on the behalf of the Nucleus of the "Heart Failure Working Group" of the German Society of Cardiology (DKG). Clin Res Cardiol 2017; 107:1-19. [PMID: 29018938 DOI: 10.1007/s00392-017-1170-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022]
Abstract
About 50% of all patients suffering from heart failure (HF) exhibit a reduced ejection fraction (EF ≤ 40%), termed HFrEF. The others may be classified into HF with midrange EF (HFmrEF 40-50%) or preserved ejection fraction (HFpEF, EF ≥ 50%). Presentation and pathophysiology of HFpEF is heterogeneous and its management remains a challenge since evidence of therapeutic benefits on outcome is scarce. Up to now, there are no therapies improving survival in patients with HFpEF. Thus, the treatment targets symptom relief, quality of life and reduction of cardiac decompensations by controlling fluid retention and managing risk factors and comorbidities. As such, renin-angiotensin-aldosterone inhibitors, diuretics, calcium channel blockers (CBB) and beta-blockers, diet and exercise recommendations are still important in HFpEF, although these interventions are not proven to reduce mortality in large randomized controlled trials. Recently, numerous new treatment targets have been identified, which are further investigated in studies using, e.g. soluble guanylate cyclase stimulators, inorganic nitrates, the angiotensin receptor neprilysin inhibitor LCZ 696, and SGLT2 inhibitors. In addition, several devices such as the CardioMEMS, interatrial septal devices (IASD), cardiac contractility modulation (CCM), renal denervation, and baroreflex activation therapy (BAT) were investigated in different forms of HFpEF populations and some of them have the potency to offer new hopes for patients suffering from HFpEF. On the basic research field side, lot of new disease-modifying strategies are under development including anti-inflammatory drugs, mitochondrial-targeted antioxidants, new anti-fibrotic and microRNA-guided interventions are under investigation and showed already promising results. This review addresses available data of current best clinical practice and management approaches based on expert experiences and summarizes novel approaches towards HFpEF.
Collapse
Affiliation(s)
- Carsten Tschöpe
- Department of Cardiology, Universitätsmedizin Berlin, Charite, Campus Rudolf Virchow Clinic (CVK), Augustenburger Platz 1, 13353, Berlin, Germany. .,Berliner Zentrum für Regenerative Therapien (BCRT), Charite, Campus Virchow Clinic (CVK), Berlin, Germany. .,Deutsches Zentrum für Herz Kreislaufforschung (DZHK), Standort Berlin/Charité, Berlin, Germany.
| | - Christoph Birner
- Germany Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Michael Böhm
- Innere Medizin III-Kardiologie, Angiologie und internistische Intensivmedizin, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg, Germany
| | - Oliver Bruder
- Department of Cardiology and Angiology, Elisabeth Hospital, Essen, Germany
| | - Stefan Frantz
- Department of Internal Medicine III, University Halle, Halle, Germany
| | - Andreas Luchner
- Department of Internal Medicine I, Clinic St. Marien, Amberg, Germany
| | - Lars Maier
- Germany Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Störk
- Deutsches Zentrum für Herzinsuffizienz, Universitätsklinikum und Universität Würzburg, Würzburg, Germany
| | - Behrouz Kherad
- Department of Cardiology, Universitätsmedizin Berlin, Charite, Campus Rudolf Virchow Clinic (CVK), Augustenburger Platz 1, 13353, Berlin, Germany.,Privatpraxis Dr. Kherad, Berlin, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie im Department für Innere Medizin, Neurologie und Dermatologie, Universitätsklinikum Leipzig, Leipzig, Germany
| |
Collapse
|
42
|
Bögeholz N, Schulte JS, Kaese S, Bauer BK, Pauls P, Dechering DG, Frommeyer G, Goldhaber JI, Kirchhefer U, Eckardt L, Pott C, Müller FU. The Effects of SEA0400 on Ca 2+ Transient Amplitude and Proarrhythmia Depend on the Na +/Ca 2+ Exchanger Expression Level in Murine Models. Front Pharmacol 2017; 8:649. [PMID: 28983248 PMCID: PMC5613119 DOI: 10.3389/fphar.2017.00649] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/01/2017] [Indexed: 11/13/2022] Open
Abstract
Background/Objective: The cardiac Na+/Ca2+ exchanger (NCX) has been identified as a promising target to counter arrhythmia in previous studies investigating the benefit of NCX inhibition. However, the consequences of NCX inhibition have not been investigated in the setting of altered NCX expression and function, which is essential, since major cardiac diseases (heart failure/atrial fibrillation) exhibit NCX upregulation. Thus, we here investigated the effects of the NCX inhibitor SEA0400 on the Ca2+ transient amplitude and on proarrhythmia in homozygous NCX overexpressor (OE) and heterozygous NCX knockout (hetKO) mice compared to corresponding wild-types (WTOE/WThetKO). Methods/Results: Ca2+ transients of field-stimulated isolated ventricular cardiomyocytes were recorded with fluo-4-AM or indo-1-AM. SEA0400 (1 μM) significantly reduced NCX forward mode function in all mouse lines. SEA0400 (1 μM) significantly increased the amplitude of field-stimulated Ca2+ transients in WTOE, WThetKO, and hetKO, but not in OE (% of basal; OE = 98.7 ± 5.0; WTOE = 137.8 ± 5.2*; WThetKO = 126.3 ± 6.0*; hetKO = 140.6 ± 12.8*; *p < 0.05 vs. basal). SEA0400 (1 μM) significantly reduced the number of proarrhythmic spontaneous Ca2+ transients (sCR) in OE, but increased it in WTOE, WThetKO and hetKO (sCR per cell; basal/+SEA0400; OE = 12.5/3.7; WTOE = 0.2/2.4; WThetKO = 1.3/8.8; hetKO = 0.2/5.5) and induced Ca2+ overload with subsequent cell death in hetKO. Conclusion: The effects of SEA0400 on Ca2+ transient amplitude and the occurrence of spontaneous Ca2+ transients as a proxy measure for inotropy and cellular proarrhythmia depend on the NCX expression level. The antiarrhythmic effect of SEA0400 in conditions of increased NCX expression promotes the therapeutic concept of NCX inhibition in heart failure/atrial fibrillation. Conversely, in conditions of reduced NCX expression, SEA0400 suppressed the NCX function below a critical level leading to adverse Ca2+ accumulation as reflected by an increase in Ca2+ transient amplitude, proarrhythmia and cell death. Thus, the remaining NCX function under inhibition may be a critical factor determining the inotropic and antiarrhythmic efficacy of SEA0400.
Collapse
Affiliation(s)
- Nils Bögeholz
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany
| | - Jan S Schulte
- Institute of Pharmacology and Toxicology, University of MünsterMünster, Germany
| | - Sven Kaese
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany
| | - B Klemens Bauer
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany.,Institute of Pharmacology and Toxicology, University of MünsterMünster, Germany
| | - Paul Pauls
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany.,Institute of Pharmacology and Toxicology, University of MünsterMünster, Germany
| | - Dirk G Dechering
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany
| | - Gerrit Frommeyer
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany
| | - Joshua I Goldhaber
- Cedars-Sinai Medical Center, Heart InstituteLos Angeles, CA, United States
| | - Uwe Kirchhefer
- Institute of Pharmacology and Toxicology, University of MünsterMünster, Germany
| | - Lars Eckardt
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany
| | - Christian Pott
- Division of Electrophysiology, Department of Cardiovascular Medicine, University Hospital MünsterMünster, Germany
| | - Frank U Müller
- Institute of Pharmacology and Toxicology, University of MünsterMünster, Germany
| |
Collapse
|
43
|
The profile of selected single nucleotide polymorphisms in patients with hypertension and heart failure with preserved and mid-range ejection fraction. Sci Rep 2017; 7:8974. [PMID: 28827564 PMCID: PMC5566797 DOI: 10.1038/s41598-017-09564-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/25/2017] [Indexed: 12/23/2022] Open
Abstract
The study aimed to assess the clinical significance of selected single nucleotide polymorphisms (SNPs) in patients with diastolic heart failure (HF): inflammation [-174 G/C Interleukin -6 (IL-6) rs1800795, tumor necrosis factor (TNF)-608 G/A rs1800629], fibrosis [Arg25Pro transforming growth factor β (TGF β) rs1800471], endothelial function [-786 T/C nitric oxide synthase (NOS) rs2070744], glucose and lipid metabolism [Pro12Ala peroxisome proliferator activated receptor (PPAR)γ rs1801282], and vitamin D metabolism [cytochrome P450 27B1 (CYP27B1) C-1260A].110 patients with HF with preserved and mid-range ejection fraction (HFpEF and HFmrEF) were recruited. GG homozygotes in 174 G/C of IL6 polymorphism are characterized by higher values of estimated glomerular filtration rate based on the study Modification of Diet in Renal Disease (eGFR MDRD) and C allele in the NOS polymorphism and AA profile in C-1260A of CYP27B1 polymorphism correlated with a lower eGFR (MDRD). In multivariate analysis the CG genotype for 174 G/C of IL-6 and allele A in C-1260A of CYP27B1 are the only SNPs independently associated with worse course of HFpEF and HFmrEF. These data confirm the importance of the selected SNPs in aggravation and complications of hypertension.
Collapse
|
44
|
Peana D, Domeier TL. Cardiomyocyte Ca 2+ homeostasis as a therapeutic target in heart failure with reduced and preserved ejection fraction. Curr Opin Pharmacol 2017; 33:17-26. [PMID: 28437711 DOI: 10.1016/j.coph.2017.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/22/2017] [Indexed: 12/28/2022]
Abstract
Heart failure is a highly prevalent syndrome of multiple etiologies and associated comorbidities, and aberrant intracellular Ca2+ homeostasis is a hallmark finding in heart failure patients. The cyclical changes in Ca2+ concentration within cardiomyocytes control cycles of cardiac contraction and relaxation, and dysregulation of Ca2+ handling processes leads to systolic dysfunction, diastolic dysfunction, and adverse remodeling. For this reason, greater understanding of Ca2+ handling mechanisms in heart failure is critical for selection of appropriate treatment strategies. In this review, we summarize the mechanisms of altered Ca2+ handling in two subsets of heart failure, heart failure with reduced ejection fraction and heart failure with preserved ejection fraction, and outline current and experimental treatments that target cardiomyocyte Ca2+ handling processes.
Collapse
Affiliation(s)
- Deborah Peana
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
45
|
Murphy E, Amanakis G, Fillmore N, Parks RJ, Sun J. Sex Differences in Metabolic Cardiomyopathy. Cardiovasc Res 2017; 113:370-377. [PMID: 28158412 PMCID: PMC5852638 DOI: 10.1093/cvr/cvx008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 12/19/2016] [Accepted: 01/16/2017] [Indexed: 12/23/2022] Open
Abstract
In contrast to ischemic cardiomyopathies which are more common in men, women are over-represented in diabetic cardiomyopathies. Diabetes is a risk factor for cardiovascular disease; however, there is a sexual dimorphism in this risk factor: heart disease is five times more common in diabetic women but only two-times more common in diabetic men. Heart failure with preserved ejection fraction, which is associated with metabolic syndrome, is also more prevalent in women. This review will examine potential mechanisms for the sex differences in metabolic cardiomyopathies. Sex differences in metabolism, calcium handling, nitric oxide, and structural proteins will be evaluated. Nitric oxide synthase and PPARα exhibit sex differences and have also been proposed to mediate the development of hypertrophy and heart failure. We focused on a role for these signalling pathways in regulating sex differences in metabolic cardiomyopathies.
Collapse
Affiliation(s)
- Elizabeth Murphy
- Systems Biology Center, National Heart, Lung and Blood Institute, NIH, MSC 1770, 10 Center Dr, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
46
|
Kriegel AJ, Gartz M, Afzal MZ, de Lange WJ, Ralphe JC, Strande JL. Molecular Approaches in HFpEF: MicroRNAs and iPSC-Derived Cardiomyocytes. J Cardiovasc Transl Res 2016; 10:295-304. [PMID: 28032312 DOI: 10.1007/s12265-016-9723-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/15/2016] [Indexed: 01/04/2023]
Abstract
Heart failure with preserved left ventricular ejection fraction (HFpEF) has emerged as one of the largest unmet needs in cardiovascular medicine. HFpEF is increasing in prevalence and causes significant morbidity, mortality, and health care resource utilization. Patients have multiple co-morbidities which contribute to the disease complexity. To date, no effective treatment for HFpEF has been identified. The paucity of cardiac biopsies from this patient population and the absence of well-accepted animal models limit our understanding of the underlying molecular mechanisms of HFpEF. In this review, we discuss combining state-of-the-art technologies of microRNA profiling and human induced pluripotent cell-derived cardiomyocytes (iPSC-CMs) in order to uncover novel molecular pathways that may contribute to the development of HFpEF. Here, we focus the advantages and limitations of microRNA profiling and iPSC-CMs as a disease model system to discover molecular mechanisms in HFpEF.
Collapse
Affiliation(s)
- Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Melanie Gartz
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Muhammad Z Afzal
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Willem J de Lange
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jennifer L Strande
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
47
|
Brown DA, Perry JB, Allen ME, Sabbah HN, Stauffer BL, Shaikh SR, Cleland JGF, Colucci WS, Butler J, Voors AA, Anker SD, Pitt B, Pieske B, Filippatos G, Greene SJ, Gheorghiade M. Expert consensus document: Mitochondrial function as a therapeutic target in heart failure. Nat Rev Cardiol 2016; 14:238-250. [PMID: 28004807 PMCID: PMC5350035 DOI: 10.1038/nrcardio.2016.203] [Citation(s) in RCA: 493] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Heart failure is a pressing worldwide public-health problem with millions of patients having worsening heart failure. Despite all the available therapies, the condition carries a very poor prognosis. Existing therapies provide symptomatic and clinical benefit, but do not fully address molecular abnormalities that occur in cardiomyocytes. This shortcoming is particularly important given that most patients with heart failure have viable dysfunctional myocardium, in which an improvement or normalization of function might be possible. Although the pathophysiology of heart failure is complex, mitochondrial dysfunction seems to be an important target for therapy to improve cardiac function directly. Mitochondrial abnormalities include impaired mitochondrial electron transport chain activity, increased formation of reactive oxygen species, shifted metabolic substrate utilization, aberrant mitochondrial dynamics, and altered ion homeostasis. In this Consensus Statement, insights into the mechanisms of mitochondrial dysfunction in heart failure are presented, along with an overview of emerging treatments with the potential to improve the function of the failing heart by targeting mitochondria.
Collapse
Affiliation(s)
- David A Brown
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, 1035 Integrated Life Sciences Building, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Justin B Perry
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, 1035 Integrated Life Sciences Building, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Mitchell E Allen
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, 1035 Integrated Life Sciences Building, 1981 Kraft Drive, Blacksburg, Virginia 24060, USA
| | - Hani N Sabbah
- Division of Cardiovascular Medicine, Department of Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, Michigan 48202, USA
| | - Brian L Stauffer
- Division of Cardiology, Department of Medicine, University of Colorado Denver, 12700 East 19th Avenue, B139, Aurora, Colorado 80045, USA
| | - Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA
| | - John G F Cleland
- National Heart &Lung Institute, National Institute of Health Research Cardiovascular Biomedical Research Unit, Royal Brompton &Harefield Hospitals, Imperial College, London, UK
| | - Wilson S Colucci
- Cardiovascular Medicine Section, Boston University School of Medicine and Boston Medical Center, 88 East Newton Street, C-8, Boston, Massachusetts 02118, USA
| | - Javed Butler
- Division of Cardiology, Health Sciences Center, T-16 Room 080, SUNY at Stony Brook, New York 11794, USA
| | - Adriaan A Voors
- University of Groningen, Department of Cardiology, University Medical Center Groningen, Groningen 9713 GZ, Netherlands
| | - Stefan D Anker
- Department of Innovative Clinical Trials, University Medical Centre Göttingen (UMG), Robert-Koch-Straße, D-37075, Göttingen, Germany
| | - Bertram Pitt
- University of Michigan School of Medicine, 1500 East Medical Center Drive, Ann Arbor, Michigan 48109, USA
| | - Burkert Pieske
- Department of Cardiology, Charité University Medicine, Campus Virchow Klinikum, and German Heart Center Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Gerasimos Filippatos
- National and Kopodistrian University of Athens, School of Medicine, Heart Failure Unit, Department of Cardiology, Athens University Hospital Attikon, Rimini 1, Athens 12462, Greece
| | - Stephen J Greene
- Division of Cardiology, Duke University Medical Center, 2301 Erwin Road Suite 7400, Durham, North Carolina 27705, USA
| | - Mihai Gheorghiade
- Center for Cardiovascular Innovation, Northwestern University Feinberg School of Medicine, 201 East Huron, Galter 3-150, Chicago, Illinois 60611, USA
| |
Collapse
|
48
|
Tepeköylü C, Primessnig U, Pölzl L, Graber M, Lobenwein D, Nägele F, Kirchmair E, Pechriggl E, Grimm M, Holfeld J. Shockwaves prevent from heart failure after acute myocardial ischaemia via RNA/protein complexes. J Cell Mol Med 2016; 21:791-801. [PMID: 27995765 PMCID: PMC5345685 DOI: 10.1111/jcmm.13021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022] Open
Abstract
Shock wave treatment (SWT) was shown to induce regeneration of ischaemic myocardium via Toll‐like receptor 3 (TLR3). The antimicrobial peptide LL37 gets released by mechanical stress and is known to form complexes with nucleic acids thus activating Toll‐like receptors. We suggested that SWT in the acute setting prevents from the development of heart failure via RNA/protein release. Myocardial infarction in mice was induced followed by subsequent SWT. Heart function was assessed 4 weeks later via transthoracic echocardiography and pressure–volume measurements. Human umbilical vein endothelial cells (HUVECs) were treated with SWT in the presence of RNase and proteinase and analysed for proliferation, tube formation and LL37 expression. RNA release and uptake after SWT was evaluated. We found significantly improved cardiac function after SWT. SWT resulted in significantly higher numbers of capillaries and arterioles and less left ventricular fibrosis. Supernatants of treated cells activated TLR3 reporter cells. Analysis of the supernatant revealed increased RNA levels. The effect could not be abolished by pre‐treatment of the supernatant with RNase, but only by a sequential digestion with proteinase and RNase hinting strongly towards the involvement of RNA/protein complexes. Indeed, LL37 expression as well as cellular RNA uptake were significantly increased after SWT. We show for the first time that SWT prevents from left ventricular remodelling and cardiac dysfunction via RNA/protein complex release and subsequent induction of angiogenesis. It might therefore develop a potent regenerative treatment alternative for ischaemic heart disease.
Collapse
Affiliation(s)
- Can Tepeköylü
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Uwe Primessnig
- Department of Cardiology, Charité University Berlin, Berlin, Germany
| | - Leo Pölzl
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Graber
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniela Lobenwein
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Nägele
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke Kirchmair
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Elisabeth Pechriggl
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology, Innsbruck Medical University, Innsbruck, Austria
| | - Michael Grimm
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Johannes Holfeld
- Department for Cardiac Surgery, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
49
|
Metra M. August 2016 at a glance: the new ESC guidelines, and pathophysiology, epidemiology and prognosis of heart failure. Eur J Heart Fail 2016; 18:889-90. [DOI: 10.1002/ejhf.625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health; University of Brescia; Italy
| |
Collapse
|
50
|
Thum T. Novel models and mechanisms of heart failure with preserved ejection fraction. Eur J Heart Fail 2016; 18:998-9. [DOI: 10.1002/ejhf.596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 11/08/2022] Open
Affiliation(s)
- Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), IFB-Tx; Hannover Medical School; Hannover Germany
- National Heart and Lung Institute; Imperial College London; U.K
| |
Collapse
|