1
|
Nguyen DC, Wells CK, Taylor MS, Martinez-Ondaro Y, Singhal R, Brittian KR, Brainard RE, Moore JB, Hill BG. Dietary Branched-Chain Amino Acids Modify Postinfarct Cardiac Remodeling and Function in the Murine Heart. J Am Heart Assoc 2025; 14:e037637. [PMID: 39950451 DOI: 10.1161/jaha.124.037637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 01/06/2025] [Indexed: 02/19/2025]
Abstract
BACKGROUND Branched-chain amino acids (BCAAs), which are derived from the diet, are markedly elevated in cardiac tissue following myocardial infarction (MI). Nevertheless, it remains unclear whether dietary BCAA levels influence post-MI remodeling. METHODS To investigate the impact of dietary BCAAs on cardiac remodeling and function after MI, we fed mice a low or a high BCAA diet for 2 weeks before MI and for 4 weeks after MI. Cardiac structural and functional changes were evaluated by echocardiography, gravimetry, and histopathological analyses. Immunoblotting was used to evaluate the effects of BCAAs on isolated cardiac myofibroblast differentiation. RESULTS The low BCAA diet decreased circulating BCAA concentrations by >2-fold when compared with the high BCAA diet. Although neither body weights nor heart masses were different in female mice fed the custom diets, male mice fed the high BCAA diet had significantly higher body and heart masses than those on the low BCAA diet. The low BCAA diet preserved stroke volume and cardiac output after MI, whereas the high BCAA diet promoted progressive decreases in cardiac function. Although BCAAs were required for myofibroblast differentiation in vitro, cardiac fibrosis, scar collagen topography, and cardiomyocyte cross-sectional area were not different between the dietary groups; however, male mice fed the high BCAA diet had longer cardiomyocytes and higher capillary density compared with the low BCAA group. CONCLUSIONS A low BCAA diet mitigates eccentric cardiomyocyte remodeling and loss of cardiac function after MI in mice, with dietary effects more prominent in males.
Collapse
Affiliation(s)
- Daniel C Nguyen
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
- Department of Physiology University of Louisville Louisville KY USA
| | - Collin K Wells
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
| | - Madison S Taylor
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
| | - Yania Martinez-Ondaro
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
| | - Richa Singhal
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
| | - Kenneth R Brittian
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
| | | | - Joseph B Moore
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
| | - Bradford G Hill
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine University of Louisville Louisville KY USA
| |
Collapse
|
2
|
Nguyen DC, Wells CK, Taylor MS, Martinez-Ondaro Y, Brittian KR, Brainard RE, Moore IV JB, Hill BG. Dietary Branched Chain Amino Acids Modify Post-Infarct Cardiac Remodeling and Function in the Murine Heart. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603348. [PMID: 39071416 PMCID: PMC11275808 DOI: 10.1101/2024.07.12.603348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Introduction Branch-chain amino acids (BCAA) are markedly elevated in the heart following myocardial infarction (MI) in both humans and animal models. Nevertheless, it remains unclear whether dietary BCAA levels influence post-MI remodeling. We hypothesize that lowering dietary BCAA levels prevents adverse cardiac remodeling after MI. Methods and Results To assess whether altering dietary BCAA levels would impact circulating BCAA concentrations, mice were fed a low (1/3×), normal (1×), or high (2×) BCAA diet over a 7-day period. We found that mice fed the low BCAA diet had >2-fold lower circulating BCAA concentrations when compared with normal and high BCAA diet feeding strategies; notably, the high BCAA diet did not further increase BCAA levels over the normal chow diet. To investigate the impact of dietary BCAAs on cardiac remodeling and function after MI, male and female mice were fed either the low or high BCAA diet for 2 wk prior to MI and for 4 wk after MI. Although body weights or heart masses were not different in female mice fed the custom diets, male mice fed the high BCAA diet had significantly higher body and heart masses than those on the low BCAA diet. Echocardiographic assessments revealed that the low BCAA diet preserved stroke volume and cardiac output for the duration of the study, while the high BCAA diet led to progressive decreases in cardiac function. Although no discernible differences in cardiac fibrosis, scar collagen topography, or cardiomyocyte cross-sectional area were found between the dietary groups, male mice fed the high BCAA diet showed longer cardiomyocytes and higher capillary density compared with the low BCAA group. Conclusions Provision of a diet low in BCAAs to mice mitigates eccentric cardiomyocyte remodeling and loss of cardiac function after MI, with dietary effects more prominent in males.
Collapse
Affiliation(s)
- Daniel C. Nguyen
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
- Department of Physiology, University of Louisville, Louisville, KY
| | - Collin K. Wells
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Madison S. Taylor
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Yania Martinez-Ondaro
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Kenneth R. Brittian
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | | | - Joseph B. Moore IV
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| | - Bradford G. Hill
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Division of Environmental Medicine, Department of Medicine, University of Louisville, Louisville, KY
| |
Collapse
|
3
|
Li Y, Johnson JP, Yang Y, Yu D, Kubo H, Berretta RM, Wang T, Zhang X, Foster M, Yu J, Tilley DG, Houser SR, Chen X. Effects of maternal hypothyroidism on postnatal cardiomyocyte proliferation and cardiac disease responses of the progeny. Am J Physiol Heart Circ Physiol 2023; 325:H702-H719. [PMID: 37539452 PMCID: PMC10659327 DOI: 10.1152/ajpheart.00320.2023] [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: 05/31/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/05/2023]
Abstract
Maternal hypothyroidism (MH) could adversely affect the cardiac disease responses of the progeny. This study tested the hypothesis that MH reduces early postnatal cardiomyocyte (CM) proliferation so that the adult heart of MH progeny has a smaller number of larger cardiac myocytes, which imparts adverse cardiac disease responses following injury. Thyroidectomy (TX) was used to establish MH. The progeny from mice that underwent sham or TX surgery were termed Ctrl (control) or MH (maternal hypothyroidism) progeny, respectively. MH progeny had similar heart weight (HW) to body weight (BW) ratios and larger CM size consistent with fewer CMs at postnatal day 60 (P60) compared with Ctrl (control) progeny. MH progeny had lower numbers of EdU+, Ki67+, and phosphorylated histone H3 (PH3)+ CMs, which suggests they had a decreased CM proliferation in the postnatal timeframe. RNA-seq data showed that genes related to DNA replication were downregulated in P5 MH hearts, including bone morphogenetic protein 10 (Bmp10). Both in vivo and in vitro studies showed Bmp10 treatment increased CM proliferation. After transverse aortic constriction (TAC), the MH progeny had more severe cardiac pathological remodeling compared with the Ctrl progeny. Thyroid hormone (T4) treatment for MH mothers preserved their progeny's postnatal CM proliferation capacity and prevented excessive pathological remodeling after TAC. Our results suggest that CM proliferation during early postnatal development was significantly reduced in MH progeny, resulting in fewer CMs with hypertrophy in adulthood. These changes were associated with more severe cardiac disease responses after pressure overload.NEW & NOTEWORTHY Our study shows that compared with Ctrl (control) progeny, the adult progeny of mothers who have MH (MH progeny) had fewer CMs. This reduction of CM numbers was associated with decreased postnatal CM proliferation. Gene expression studies showed a reduced expression of Bmp10 in MH progeny. Bmp10 has been linked to myocyte proliferation. In vivo and in vitro studies showed that Bmp10 treatment of MH progeny and their myocytes could increase CM proliferation. Differences in CM number and size in adult hearts of MH progeny were linked to more severe cardiac structural and functional remodeling after pressure overload. T4 (synthetic thyroxine) treatment of MH mothers during their pregnancy, prevented the reduction in CM number in their progeny and the adverse response to disease stress.
Collapse
Affiliation(s)
- Yijia Li
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Jaslyn P Johnson
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Yijun Yang
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Daohai Yu
- Department of Biomedical Education and Data Science, Center for Biostatistics and Epidemiology, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Hajime Kubo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Remus M Berretta
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Tao Wang
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Xiaoying Zhang
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Michael Foster
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Jun Yu
- Department of Cardiovascular Sciences, Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Douglas G Tilley
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Cardiovascular Research Center, Philadelphia, Pennsylvania, United States
| | - Steven R Houser
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Xiongwen Chen
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
4
|
Frisk M, Norseng PA, Stenersen Espe EK, Louch WE. Tubulator: an automated approach to analysis of t-tubule and dyadic organization in cardiomyocytes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210468. [PMID: 36189810 PMCID: PMC9527907 DOI: 10.1098/rstb.2021.0468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 04/13/2022] [Indexed: 11/12/2022] Open
Abstract
During cardiac disease, t-tubules and dyads are remodelled and disrupted within cardiomyocytes, thereby reducing cardiac performance. Given the pathological implications of such dyadic remodelling, robust and versatile tools for characterizing these sub-cellular structures are needed. While analysis programs for continuous and regular structures such as rodent ventricular t-tubules are available, at least in two dimensions, these approaches are less appropriate for assessment of more irregular structures, such as dyadic proteins and non-rodent t-tubules. Here, we demonstrate versatile, easy-to-use software that performs such analyses. This software, called Tubulator, enables automated analysis of t-tubules and dyadic proteins alike, in both tissue sections and isolated myocytes. The program measures densities of subcellular structures and proteins in individual cells, quantifies their distribution into transversely and longitudinally oriented elements, and supports detailed co-localization analyses. Importantly, Tubulator provides tools for three-dimensional assessment and rendering of image stacks, extending examinations from the single plane to the whole-myocyte level. To provide insight into the consequences of dyadic organization for synchrony of Ca2+ handling, Tubulator also creates 'distance maps', by calculating the distance from all cytosolic positions to the nearest t-tubule and/or dyad. In conclusion, this freely accessible program provides detailed automated analysis of the three-dimensional nature of dyadic and t-tubular structures. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
- K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
- K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Emil Knut Stenersen Espe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
- K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, 0424 Oslo, Norway
- K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
5
|
Role of ranolazine in heart failure: From cellular to clinic perspective. Eur J Pharmacol 2022; 919:174787. [PMID: 35114190 DOI: 10.1016/j.ejphar.2022.174787] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/25/2021] [Accepted: 01/25/2022] [Indexed: 12/17/2022]
Abstract
Ranolazine was approved by the US Food and Drug Administration as an antianginal drug in 2006, and has been used since in certain groups of patients with stable angina. The therapeutic action of ranolazine was initially attributed to inhibitory effects on fatty acids metabolism. As investigations went on, however, it developed that the main beneficial effects of ranolazine arise from its action on the late sodium current in the heart. Since late sodium currents were discovered to be involved in various heart pathologies such as ischemia, arrhythmias, systolic and diastolic dysfunctions, and all these conditions are associated with heart failure, ranolazine has in some way been tested either directly or indirectly on heart failure in numerous experimental and clinical studies. As the heart continuously remodels following any sort of severe injury, the inhibition by ranolazine of the underlying mechanisms of cardiac remodeling including ion disturbances, oxidative stress, inflammation, apoptosis, fibrosis, metabolic dysregulation, and neurohormonal impairment are discussed, along with unresolved issues. A projection of pathologies targeted by ranolazine from cellular level to clinical is provided in this review.
Collapse
|
6
|
Zhou J, Singh N, Monnier C, Marszalec W, Gao L, Jin J, Frisk M, Louch WE, Verma S, Krishnamurthy P, Nico E, Mulla M, Aistrup GL, Kishore R, Wasserstrom JA. Phosphatidylinositol-4,5-Bisphosphate Binding to Amphiphysin-II Modulates T-Tubule Remodeling: Implications for Heart Failure. Front Physiol 2022; 12:782767. [PMID: 35002765 PMCID: PMC8733645 DOI: 10.3389/fphys.2021.782767] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
BIN1 (amphyphysin-II) is a structural protein involved in T-tubule (TT) formation and phosphatidylinositol-4,5-bisphosphate (PIP2) is responsible for localization of BIN1 to sarcolemma. The goal of this study was to determine if PIP2-mediated targeting of BIN1 to sarcolemma is compromised during the development of heart failure (HF) and is responsible for TT remodeling. Immunohistochemistry showed co-localization of BIN1, Cav1.2, PIP2, and phospholipase-Cβ1 (PLCβ1) in TTs in normal rat and human ventricular myocytes. PIP2 levels were reduced in spontaneously hypertensive rats during HF progression compared to age-matched controls. A PIP Strip assay of two native mouse cardiac-specific isoforms of BIN1 including the longest (cardiac BIN1 #4) and shortest (cardiac BIN1 #1) isoforms as well human skeletal BIN1 showed that all bound PIP2. In addition, overexpression of all three BIN1 isoforms caused tubule formation in HL-1 cells. A triple-lysine motif in a short loop segment between two helices was mutated and replaced by negative charges which abolished tubule formation, suggesting a possible location for PIP2 interaction aside from known consensus binding sites. Pharmacological PIP2 depletion in rat ventricular myocytes caused TT loss and was associated with changes in Ca2+ release typically found in myocytes during HF, including a higher variability in release along the cell length and a slowing in rise time, time to peak, and decay time in treated myocytes. These results demonstrate that depletion of PIP2 can lead to TT disruption and suggest that PIP2 interaction with cardiac BIN1 is required for TT maintenance and function.
Collapse
Affiliation(s)
- Junlan Zhou
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Neha Singh
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Chloe Monnier
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - William Marszalec
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Li Gao
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jing Jin
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Michael Frisk
- Institute for Experimental Medical Research (IEMR), Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research (IEMR), Oslo University Hospital, Oslo, Norway.,K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Suresh Verma
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Prasanna Krishnamurthy
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Elsa Nico
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Maaz Mulla
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Gary L Aistrup
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Raj Kishore
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - J Andrew Wasserstrom
- Department of Medicine (Cardiology), Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
7
|
Yu J, Zhao H, Qi X, Wei L, Li Z, Li C, Zhang X, Wu H. Dapagliflozin Mediates Plin5/PPARα Signaling Axis to Attenuate Cardiac Hypertrophy. Front Pharmacol 2021; 12:730623. [PMID: 34630108 PMCID: PMC8495133 DOI: 10.3389/fphar.2021.730623] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: The purpose of this study was to investigate the effect of dapagliflozin (DAPA), a sodium-glucose cotransporter 2 inhibitor, on relieving cardiac hypertrophy and its potential molecular mechanism. Methods: Cardiac hypertrophy induced by abdominal aortic constriction (AAC) in mice, dapagliflozin were administered in the drinking water at a dose of 25 mg/kg/d for 12 weeks was observed. Echocardiography was used to detect the changes of cardiac function, including LVEF, LVFS, LVEDd, LVEDs, HR and LV mass. Histological morphological changes were evaluated by Masson trichrome staining and wheat germ agglutinin (WGA) staining. The enrichment of differential genes and signal pathways after treatment was analyzed by gene microarray cardiomyocyte hypertrophy was induced by AngII (2 μM) and the protective effect of dapagliflozin (1 μM) was observed in vitro. The morphological changes of myocardial cells were detected by cTnI immunofluorescence staining. ELISA and qRT-PCR assays were performed to detect the expressions levels of cardiac hypertrophy related molecules. Results: After 12 weeks of treatment, DAPA significantly ameliorated cardiac function and inhibited cardiac hypertrophy in AAC-induced mice. In vitro, DAPA significantly inhibited abnormal hypertrophy in AngII-induced cardiacmyocytes. Both in vivo and in vitro experiments have confirmed that DAPA could mediate the Plin5/PPARα signaling axis to play a protective role in inhibiting cardiac hypertrophy. Conclusion: Dapagliflozin activated the Plin5/PPARα signaling axis and exerts a protective effect against cardiac hypertrophy.
Collapse
Affiliation(s)
- Jing Yu
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Huanhuan Zhao
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xin Qi
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China.,Nankai University School of Medicine, Tianjin, China
| | - Liping Wei
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China.,Nankai University School of Medicine, Tianjin, China
| | - Zihao Li
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Chunpeng Li
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xiaoying Zhang
- Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Hao Wu
- Department of Cardiology, Tianjin Union Medical Center, Nankai University Affiliated Hospital, Tianjin, China.,Nankai University School of Medicine, Tianjin, China
| |
Collapse
|
8
|
Yamakawa S, Wu D, Dasgupta M, Pedamallu H, Gupta B, Modi R, Mufti M, O'Callaghan C, Frisk M, Louch WE, Arora R, Shiferaw Y, Burrell A, Ryan J, Nelson L, Chow M, Shah SJ, Aistrup G, Zhou J, Marszalec W, Wasserstrom JA. Role of t-tubule remodeling on mechanisms of abnormal calcium release during heart failure development in canine ventricle. Am J Physiol Heart Circ Physiol 2021; 320:H1658-H1669. [PMID: 33635163 PMCID: PMC8260383 DOI: 10.1152/ajpheart.00946.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/16/2021] [Accepted: 02/22/2021] [Indexed: 11/22/2022]
Abstract
The goal of this work was to investigate the role of t-tubule (TT) remodeling in abnormal Ca2+ cycling in ventricular myocytes of failing dog hearts. Heart failure (HF) was induced using rapid right ventricular pacing. Extensive changes in echocardiographic parameters, including left and right ventricular dilation and systolic dysfunction, diastolic dysfunction, elevated left ventricular filling pressures, and abnormal cardiac mechanics, indicated that severe HF developed. TT loss was extensive when measured as the density of total cell volume, derived from three-dimensional confocal image analysis, and significantly increased the distances in the cell interior to closest cell membrane. Changes in Ca2+ transients indicated increases in heterogeneity of Ca2+ release along the cell length. When critical properties of Ca2+ release variability were plotted as a function of TT organization, there was a complex, nonlinear relationship between impaired calcium release and decreasing TT organization below a certain threshold of TT organization leading to increased sensitivity in Ca2+ release below a TT density threshold of 1.5%. The loss of TTs was also associated with a greater incidence of triggered Ca2+ waves during rapid pacing. Finally, virtually all of these observations were replicated by acute detubulation by formamide treatment, indicating an important role of TT remodeling in impaired Ca2+ cycling. We conclude that TT remodeling itself is a major contributor to abnormal Ca2+ cycling in HF, reducing myocardial performance. The loss of TTs is also responsible for a greater incidence of triggered Ca2+ waves that may play a role in ventricular arrhythmias arising in HF.NEW & NOTEWORTHY Three-dimensional analysis of t-tubule density showed t-tubule disruption throughout the whole myocyte in failing dog ventricle. A double-linear relationship between Ca2+ release and t-tubule density displays a steeper slope at t-tubule densities below a threshold value (∼1.5%) above which there is little effect on Ca2+ release (T-tubule reserve). T-tubule loss increases incidence of triggered Ca2+ waves. Chemically induced t-tubule disruption suggests that t-tubule loss alone is a critical component of abnormal Ca2+ cycling in heart failure.
Collapse
Affiliation(s)
- Sean Yamakawa
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Daniel Wu
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Mona Dasgupta
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Havisha Pedamallu
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Binita Gupta
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Rishi Modi
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Maryam Mufti
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Caitlin O'Callaghan
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - William E Louch
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Rishi Arora
- California State University Northridge, Los Angeles, California
| | - Yohannes Shiferaw
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Amy Burrell
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Juliet Ryan
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Lauren Nelson
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Madeleine Chow
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sanjiv J Shah
- The Masonic Medical Research Institute, Utica, New York
| | - Gary Aistrup
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Junlan Zhou
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William Marszalec
- Feinberg Cardiovascular and Renal Research Institute and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | |
Collapse
|
9
|
Tawfik MK, Ameen AM. Cardioprotective effect of ranolazine in nondiabetic and diabetic male rats subjected to isoprenaline-induced acute myocardial infarction involves modulation of AMPK and inhibition of apoptosis. Can J Physiol Pharmacol 2019; 97:661-674. [DOI: 10.1139/cjpp-2018-0571] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diabetes increases the sensitivity of myocardium to ischemic damage and impairs response of the myocardium to cardioprotective interventions. The present study aimed to elucidate the potential cardioprotective effect provided by ranolazine during myocardial infarction in nondiabetic and diabetic male rats. As AMP-activated protein kinase (AMPK) has been shown to be involved in the cellular response to ischemic injury, in this context, the present animal study evaluated the modulating role of ranolazine in the AMPK expression in isoprenaline-induced myocardial ischemic rat model. Male rats were divided into 2 experiments: experiment I and II (nondiabetic and diabetic rats) and assigned to normal control, saline control for isoprenaline, isoprenaline control, and ranolazine-treated groups. Ranolazine administration revealed effectiveness in attenuating the severity of isoprenaline-induced myocardial injury in both nondiabetic and diabetic rats as revealed by ECG signs, histopathological score, and apoptotic markers via abrogating the increments in the inflammatory and oxidative stress markers and modulating AMPK expression. Therefore, the current cardioprotective effect of ranolazine was, at least in part, mediated through inhibition of apoptosis and modulation of AMPK expression, encouraging considering the utility of ranolazine in protection from acute myocardial infarction.
Collapse
Affiliation(s)
- Mona K. Tawfik
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Angie M. Ameen
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
10
|
Ghosh GC, Ghosh RK, Bandyopadhyay D, Chatterjee K, Aneja A. Ranolazine: Multifaceted Role beyond Coronary Artery Disease, a Recent Perspective. Heart Views 2019; 19:88-98. [PMID: 31007857 PMCID: PMC6448470 DOI: 10.4103/heartviews.heartviews_18_18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ranolazine is a piperazine derivative approved as an antianginal. Primarily used as a second-line antianginal in stable coronary artery disease. Ranolazine blocks the late Na + current and prevents the rise of cytosolic calcium. It decreases myocardial wall tension and improves coronary blood flow. Ranolazine is effective in atrial fibrillation (AF) as an adjunct to electrical or pharmacological cardioversion. It can be used in combination with amiodarone or dronedarone. It has also been used in AF arising after coronary artery bypass grafting surgery. Role of ranolazine is also being evaluated in pulmonary arterial hypertension, diastolic dysfunction, and chemotherapy-induced cardiotoxicity. Ranolazine has some anti-glycemic effect and has shown a reduction of hemoglobin A1c in multiple trials. The antianginal effect of ranolazine has also been seen to be more in patients with diabetes compared to those without diabetes. Ranolazine is being evaluated in patients with the peripheral arterial disease with intermittent claudication and hypertrophic cardiomyopathy. Pilot studies have shown that ranolazine may be beneficial in neurological conditions with myotonia. The evidence-base on the use of ranolazine in various conditions is rapidly increasing with results of further trials eagerly awaited. Accumulating evidence may see ranolazine in routine clinical use for many conditions beyond its traditional role as an antianginal.
Collapse
Affiliation(s)
- Gopal Chandra Ghosh
- Department of Cardiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Raktim Kumar Ghosh
- MetroHealth Medical Center, Case Western Reserve University, Heart and Vascular Institute, Cleveland, OH, USA
| | | | - Krishnarpan Chatterjee
- Department of Cardiology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Ashish Aneja
- MetroHealth Medical Center, Case Western Reserve University, Heart and Vascular Institute, Cleveland, OH, USA
| |
Collapse
|
11
|
Ke HY, Yang HY, Francis AJ, Collins TP, Surendran H, Alvarez-Laviada A, Firth JM, MacLeod KT. Changes in cellular Ca 2+ and Na + regulation during the progression towards heart failure in the guinea pig. J Physiol 2019; 598:1339-1359. [PMID: 30811606 PMCID: PMC7187457 DOI: 10.1113/jp277038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 02/26/2019] [Indexed: 12/19/2022] Open
Abstract
Key points During compensated hypertrophy in vivo fractional shortening (FS) remains constant until heart failure (HF) develops, when FS decreases from 70% to 39%. Compensated hypertrophy is accompanied by an increase in INa,late and a decrease in Na+,K+‐ATPase current. These changes persist as HF develops. SR Ca2+ content increases during compensated hypertrophy then decreases in HF. In healthy cells, increases in SR Ca2+ content and Ca2+ transients can be achieved by the same amount of inhibition of the Na+,K+‐ATPase as measured in the diseased cells. SERCA function remains constant during compensated hypertrophy then decreases in HF, when there is also an increase in spark frequency and spark‐mediated Ca2+ leak. We suggest an increase in INa,late and a decrease in Na+,K+‐ATPase current and function alters the balance of Ca2+ flux mediated by the Na+/Ca2+ exchange that limits early contractile impairment.
Abstract We followed changes in cardiac myocyte Ca2+ and Na+ regulation from the formation of compensated hypertrophy (CH) until signs of heart failure (HF) are apparent using a trans‐aortic pressure overload (TAC) model. In this model, in vivo fractional shortening (FS) remained constant despite HW:BW ratio increasing by 39% (CH) until HF developed 150 days post‐TAC when FS decreased from 70% to 39%. Using live and fixed fluorescence imaging and electrophysiological techniques, we found an increase in INa,late from –0.34 to –0.59 A F−1 and a decrease in Na+,K+‐ATPase current from 1.09 A F−1 to 0.54 A F−1 during CH. These changes persisted as HF developed (INa,late increased to –0.82 A F−1 and Na+,K+‐ATPase current decreased to 0.51 A F−1). Sarcoplasmic reticulum (SR) Ca2+ content increased during CH then decreased in HF (from 32 to 15 μm l−1) potentially supporting the maintenance of FS in the whole heart and Ca2+ transients in single myocytes during the former stage. We showed using glycoside blockade in healthy myocytes that increases in SR Ca2+ content and Ca2+ transients can be driven by the same amount of inhibition of the Na+,K+‐ATPase as measured in the diseased cells. SERCA function remains constant in CH but decreases (τ for SERCA‐mediated Ca2+ removal changed from 6.3 to 3.0 s−1) in HF. In HF there was an increase in spark frequency and spark‐mediated Ca2+ leak. We suggest an increase in INa,late and a decrease in Na+,K+‐ATPase current and function alters the balance of Ca2+ flux mediated by the Na+/Ca2+ exchange that limits early contractile impairment. During compensated hypertrophy in vivo fractional shortening (FS) remains constant until heart failure (HF) develops, when FS decreases from 70% to 39%. Compensated hypertrophy is accompanied by an increase in INa,late and a decrease in Na+,K+‐ATPase current. These changes persist as HF develops. SR Ca2+ content increases during compensated hypertrophy then decreases in HF. In healthy cells, increases in SR Ca2+ content and Ca2+ transients can be achieved by the same amount of inhibition of the Na+,K+‐ATPase as measured in the diseased cells. SERCA function remains constant during compensated hypertrophy then decreases in HF, when there is also an increase in spark frequency and spark‐mediated Ca2+ leak. We suggest an increase in INa,late and a decrease in Na+,K+‐ATPase current and function alters the balance of Ca2+ flux mediated by the Na+/Ca2+ exchange that limits early contractile impairment.
Collapse
Affiliation(s)
- H-Y Ke
- Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (ROC)
| | - H-Y Yang
- Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan (ROC)
| | - A J Francis
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - T P Collins
- The Wellcome Trust, Gibbs Building, 215 Euston Road, London, NW1 2BE, UK
| | - H Surendran
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - A Alvarez-Laviada
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - J M Firth
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - K T MacLeod
- National Heart and Lung Institute, Imperial College, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
12
|
Calloe K. Doctoral Dissertation: The transient outward potassium current in healthy and diseased hearts. Acta Physiol (Oxf) 2019; 225 Suppl 717:e13225. [PMID: 30628199 DOI: 10.1111/apha.13225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 11/20/2018] [Accepted: 11/21/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Kirstine Calloe
- Section for Anatomy; Biochemistry and Physiology; Department for Veterinary and Animal Sciences; Faculty of Health and Medical Sciences; University of Copenhagen; Frederiksberg C Denmark
| |
Collapse
|
13
|
Cannata' A, Merlo M, Artico J, Gentile P, Camparini L, Cristallini J, Porcari A, Loffredo F, Sinagra G. Cardiovascular aging: the unveiled enigma from bench to bedside. J Cardiovasc Med (Hagerstown) 2018; 19:517-526. [PMID: 30024423 DOI: 10.2459/jcm.0000000000000694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
: The rapid increase in the median age of the world's population requires particular attention towards older and more fragile people. Cardiovascular risk factors, time and comorbidities play a vicious role in the development of heart failure, both with reduced and preserved ejection fraction, in the elderly. Understanding the mechanisms underlying the pathophysiological processes observed with aging is pivotal to target those patients and their therapeutic needs properly. This review aims to investigate and to dissect the main pathways leading to the aging cardiomyopathy, helping to understand the relationship from bench to bedside of the clinical phenotype.
Collapse
Affiliation(s)
- Antonio Cannata'
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Marco Merlo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Jessica Artico
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Piero Gentile
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Luca Camparini
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Jacopo Cristallini
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Aldostefano Porcari
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Francesco Loffredo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| |
Collapse
|
14
|
Singh JK, Barsegyan V, Bassi N, Marszalec W, Tai S, Mothkur S, Mulla M, Nico E, Shiferaw Y, Aistrup GL, Wasserstrom JA. T-tubule remodeling and increased heterogeneity of calcium release during the progression to heart failure in intact rat ventricle. Physiol Rep 2017; 5:5/24/e13540. [PMID: 29279414 PMCID: PMC5742703 DOI: 10.14814/phy2.13540] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 11/07/2017] [Accepted: 11/10/2017] [Indexed: 01/29/2023] Open
Abstract
A highly organized transverse‐tubule (TT) system is essential to normal Ca2+ cycling and cardiac function. We explored the relationship between the progressive disruption of TTs and resulting Ca2+ cycling during the development of heart failure (HF). Confocal imaging was used to measure Ca2+ transients and 2‐D z‐stack images in left ventricular epicardial myocytes of intact hearts from spontaneously hypertensive rats (SHR) and Wistar‐Kyoto control rats. TT organization was measured as the organizational index (OI) derived from a fast Fourier transform of TT organization. We found little decrease in the synchrony of Ca2+ release with TT loss until TT remodeling was severe, suggesting a TT “reserve” characterized by a wide range of TT remodeling with little effect on synchrony of release but beyond which variability in release shows an accelerating sensitivity to TT loss. To explain this observation, we applied a computational model of spatially distributed Ca2+ signaling units to investigate the relationship between OI and excitation‐contraction coupling. Our model showed that release heterogeneity exhibits a nonlinear relationship on both the spatial distribution of release units and the separation between L‐type Ca2+ channels and ryanodine receptors. Our results demonstrate a unique relationship between the synchrony of Ca2+ release and TT organization in myocytes of intact rat ventricle that may contribute to both the compensated and decompensated phases of heart failure.
Collapse
Affiliation(s)
- Jasleen K Singh
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Varderes Barsegyan
- Department of Physics and Astronomy, California State University, Northridge, California
| | - Nikhil Bassi
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William Marszalec
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Shannon Tai
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Shruthi Mothkur
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Maaz Mulla
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Elsa Nico
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Yohannes Shiferaw
- Department of Physics and Astronomy, California State University, Northridge, California
| | - Gary L Aistrup
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - John Andrew Wasserstrom
- Department of Medicine (Cardiology), The Feinberg Cardiovascular Research Institute Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
15
|
Coppini R, Mazzoni L, Ferrantini C, Gentile F, Pioner JM, Laurino A, Santini L, Bargelli V, Rotellini M, Bartolucci G, Crocini C, Sacconi L, Tesi C, Belardinelli L, Tardiff J, Mugelli A, Olivotto I, Cerbai E, Poggesi C. Ranolazine Prevents Phenotype Development in a Mouse Model of Hypertrophic Cardiomyopathy. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003565. [PMID: 28255011 DOI: 10.1161/circheartfailure.116.003565] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 01/30/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND Current therapies are ineffective in preventing the development of cardiac phenotype in young carriers of mutations associated with hypertrophic cardiomyopathy (HCM). Ranolazine, a late Na+ current blocker, reduced the electromechanical dysfunction of human HCM myocardium in vitro. METHODS AND RESULTS To test whether long-term treatment prevents cardiomyopathy in vivo, transgenic mice harboring the R92Q troponin-T mutation and wild-type littermates received an oral lifelong treatment with ranolazine and were compared with age-matched vehicle-treated animals. In 12-months-old male R92Q mice, ranolazine at therapeutic plasma concentrations prevented the development of HCM-related cardiac phenotype, including thickening of the interventricular septum, left ventricular volume reduction, left ventricular hypercontractility, diastolic dysfunction, left-atrial enlargement and left ventricular fibrosis, as evaluated in vivo using echocardiography and magnetic resonance. Left ventricular cardiomyocytes from vehicle-treated R92Q mice showed marked excitation-contraction coupling abnormalities, including increased diastolic [Ca2+] and Ca2+ waves, whereas cells from treated mutants were undistinguishable from those from wild-type mice. Intact trabeculae from vehicle-treated mutants displayed inotropic insufficiency, increased diastolic tension, and premature contractions; ranolazine treatment counteracted the development of myocardial mechanical abnormalities. In mutant myocytes, ranolazine inhibited the enhanced late Na+ current and reduced intracellular [Na+] and diastolic [Ca2+], ultimately preventing the pathological increase of calmodulin kinase activity in treated mice. CONCLUSIONS Owing to the sustained reduction of intracellular Ca2+ and calmodulin kinase activity, ranolazine prevented the development of morphological and functional cardiac phenotype in mice carrying a clinically relevant HCM-related mutation. Pharmacological inhibitors of late Na+ current are promising candidates for an early preventive therapy in young phenotype-negative subjects carrying high-risk HCM-related mutations.
Collapse
Affiliation(s)
- Raffaele Coppini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.).
| | - Luca Mazzoni
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Cecilia Ferrantini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Francesca Gentile
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Josè Manuel Pioner
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Annunziatina Laurino
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Lorenzo Santini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Valentina Bargelli
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Matteo Rotellini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Gianluca Bartolucci
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Claudia Crocini
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Leonardo Sacconi
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Chiara Tesi
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Luiz Belardinelli
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Jil Tardiff
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Alessandro Mugelli
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Iacopo Olivotto
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Elisabetta Cerbai
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| | - Corrado Poggesi
- From the Department NeuroFarBa (R.C., L.M., T.L., L. Santini, V.B., G.B., A.M., E.C.) and Department of Experimental and Clinical Medicine (C.F., F.G., J.M.P., C.T., C.P.), University of Florence, Italy; European Laboratory for Non-linear Spectroscopy (LENS), University of Florence & National Institute of Optics (INO-CNR), Sesto Fiorentino, Italy (C.C., L. Sacconi); Gilead Sciences Inc., Foster City, CA (L.B.); Department of Cellular and Molecular Medicine University of Arizona at Tucson, USA (J.T.); and Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy (M.R., I.O.)
| |
Collapse
|
16
|
Arora R, Aistrup GL, Supple S, Frank C, Singh J, Tai S, Zhao A, Chicos L, Marszalec W, Guo A, Song LS, Wasserstrom JA. Regional distribution of T-tubule density in left and right atria in dogs. Heart Rhythm 2016; 14:273-281. [PMID: 27670628 DOI: 10.1016/j.hrthm.2016.09.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND The peculiarities of transverse tubule (T-tubule) morphology and distribution in the atrium-and how they contribute to excitation-contraction coupling-are just beginning to be understood. OBJECTIVES The objectives of this study were to determine T-tubule density in the intact, live right and left atria in a large animal and to determine intraregional differences in T-tubule organization within each atrium. METHODS Using confocal microscopy, T-tubules were imaged in both atria in intact, Langendorf-perfused normal dog hearts loaded with di-4-ANEPPS. T-tubules were imaged in large populations of myocytes from the endocardial surface of each atrium. Computerized data analysis was performed using a new MatLab (Mathworks, Natick, MA) routine, AutoTT. RESULTS There was a large percentage of myocytes that had no T-tubules in both atria with a higher percentage in the right atrium (25.1%) than in the left atrium (12.5%) (P < .02). The density of transverse and longitudinal T-tubule elements was low in cells that did contain T-tubules, but there were no significant differences in density between the left atrial appendage, the pulmonary vein-posterior left atrium, the right atrial appendage, and the right atrial free wall. In contrast, there were significant differences in sarcomere spacing and cell width between different regions of the atria. CONCLUSION There is a sparse T-tubule network in atrial myocytes throughout both dog atria, with significant numbers of myocytes in both atria-the right atrium more so than the left atrium-having no T-tubules at all. These regional differences in T-tubule distribution, along with differences in cell width and sarcomere spacing, may have implications for the emergence of substrate for atrial fibrillation.
Collapse
Affiliation(s)
- Rishi Arora
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois,.
| | - Gary L Aistrup
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Stephen Supple
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Caleb Frank
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jasleen Singh
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Shannon Tai
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Anne Zhao
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Laura Chicos
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - William Marszalec
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ang Guo
- Division of Cardiology, University of Iowa School of Medicine, Iowa City, Iowa
| | - Long-Sheng Song
- Division of Cardiology, University of Iowa School of Medicine, Iowa City, Iowa
| | - J Andrew Wasserstrom
- Division of Cardiology, Department of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
17
|
Cannatà A, Camparini L, Sinagra G, Giacca M, Loffredo FS. Pathways for salvage and protection of the heart under stress: novel routes for cardiac rejuvenation. Cardiovasc Res 2016; 111:142-53. [DOI: 10.1093/cvr/cvw106] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/10/2016] [Indexed: 01/07/2023] Open
|
18
|
Frisk M, Ruud M, Espe EKS, Aronsen JM, Røe ÅT, Zhang L, Norseng PA, Sejersted OM, Christensen GA, Sjaastad I, Louch WE. Elevated ventricular wall stress disrupts cardiomyocyte t-tubule structure and calcium homeostasis. Cardiovasc Res 2016; 112:443-51. [PMID: 27226008 PMCID: PMC5031949 DOI: 10.1093/cvr/cvw111] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/16/2016] [Indexed: 11/13/2022] Open
Abstract
AIMS Invaginations of the cellular membrane called t-tubules are essential for maintaining efficient excitation-contraction coupling in ventricular cardiomyocytes. Disruption of t-tubule structure during heart failure has been linked to dyssynchronous, slowed Ca(2+) release and reduced power of the heartbeat. The underlying mechanism is, however, unknown. We presently investigated whether elevated ventricular wall stress triggers remodelling of t-tubule structure and function. METHODS AND RESULTS MRI and blood pressure measurements were employed to examine regional wall stress across the left ventricle of sham-operated and failing, post-infarction rat hearts. In failing hearts, elevated left ventricular diastolic pressure and ventricular dilation resulted in markedly increased wall stress, particularly in the thin-walled region proximal to the infarct. High wall stress in this proximal zone was associated with reduced expression of the dyadic anchor junctophilin-2 and disrupted cardiomyocyte t-tubular structure. Indeed, local wall stress measurements predicted t-tubule density across sham and failing hearts. Elevated wall stress and disrupted cardiomyocyte structure in the proximal zone were also associated with desynchronized Ca(2+) release in cardiomyocytes and markedly reduced local contractility in vivo. A causative role of wall stress in promoting t-tubule remodelling was established by applying stretch to papillary muscles ex vivo under culture conditions. Loads comparable to wall stress levels observed in vivo in the proximal zone reduced expression of junctophilin-2 and promoted t-tubule loss. CONCLUSION Elevated wall stress reduces junctophilin-2 expression and disrupts t-tubule integrity, Ca(2+) release, and contractile function. These findings provide new insight into the role of wall stress in promoting heart failure progression.
Collapse
Affiliation(s)
- Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Marianne Ruud
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Emil K S Espe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | | | - Åsmund T Røe
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Lili Zhang
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Ole M Sejersted
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Geir A Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Ullevål, Kirkeveien 166, 0424 Oslo, Norway K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, 0316 Oslo, Norway
| |
Collapse
|
19
|
Lee JC, Kim KC, Choe SY, Hong YM. Reduced immunoreactivities of B-type natriuretic peptide in pulmonary arterial hypertension rats after ranolazine treatment. Anat Cell Biol 2016; 49:7-14. [PMID: 27051563 PMCID: PMC4819080 DOI: 10.5115/acb.2016.49.1.7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 01/25/2016] [Accepted: 03/07/2016] [Indexed: 02/05/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe pulmonary vascular disease characterized by sustained increase in the pulmonary arterial pressure and excessive thickening and remodeling of the distal small pulmonary arteries. During disease progression, structural remodeling of the right ventricular (RV) impairs pump function, creates pro-arrhythmic substrates and triggers for arrhythmias. Notably, RV failure and lethal arrhythmias are major contributors to cardiac death in PAH that are not directly addressed by currently available therapies. Ranolazine (RAN) is an anti-anginal, anti-ischemic drug that has cardioprotective effects of heart dysfunction. RAN also has anti-arrhythmic effects due to inhibition of the late sodium current in cardiomyocytes. Therefore, we hypothesized that RAN could reduce the mal-adaptive structural remodeling of the RV, and prevent triggered ventricular arrhythmias in the monocrotaline-induced rat model of PAH. RAN reduced ventricular hypertrophy, reduced levels of B-type natriuretic peptide, and decreased the expression of fibrosis. In addition, RAN prevented cardiovascular death in rat model of PAH. These results support the notion that RAN can improve the functional properties of the RV, highlighting its potential benefits in the setting of heart impairment.
Collapse
Affiliation(s)
- Jae Chul Lee
- Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea.; Department of Surgery, Brain Korea 21 PLUS Project for Medical Sciences and HBP Surgery and Liver Transplantation, Korea University College of Medicine, Seoul, Korea.; Department of Anatomy, Seoul National University College of Medicine, Seoul, Korea.; Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| | - Kwan Chang Kim
- Department of Thoracic and Cardiovascular Surgery, Ewha Womans University School of Medicine, Seoul, Korea
| | - Soo Young Choe
- Department of Biology, School of Life Sciences, Chungbuk National University, Cheongju, Korea
| | - Young Mi Hong
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| |
Collapse
|
20
|
Liles JT, Hoyer K, Oliver J, Chi L, Dhalla AK, Belardinelli L. Ranolazine reduces remodeling of the right ventricle and provoked arrhythmias in rats with pulmonary hypertension. J Pharmacol Exp Ther 2015; 353:480-9. [PMID: 25770134 DOI: 10.1124/jpet.114.221861] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 03/12/2015] [Indexed: 01/22/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease that often results in right ventricular (RV) failure and death. During disease progression, structural and electrical remodeling of the right ventricle impairs pump function, creates proarrhythmic substrates, and triggers for arrhythmias. Notably, RV failure and lethal arrhythmias are major contributors to cardiac death in patients with PAH that are not directly addressed by currently available therapies. Ranolazine (RAN) is an antianginal, anti-ischemic drug that has cardioprotective effects in experimental and clinical settings of left-sided heart dysfunction. RAN also has antiarrhythmic effects due to inhibition of the late sodium current in cardiomyocytes. We therefore hypothesized that RAN could reduce the maladaptive structural and electrical remodeling of the right ventricle and could prevent triggered ventricular arrhythmias in the monocrotaline rat model of PAH. Indeed, in both in vivo and ex vivo experimental settings, chronic RAN treatment reduced electrical heterogeneity (right ventricular-left ventricular action potential duration dispersion), shortened heart-rate corrected QT intervals in the right ventricle, and normalized RV dysfunction. Chronic RAN treatment also dose-dependently reduced ventricular hypertrophy, reduced circulating levels of B-type natriuretic peptide, and decreased the expression of fibrotic markers. In addition, the acute administration of RAN prevented isoproterenol-induced ventricular tachycardia/ventricular fibrillation and subsequent cardiovascular death in rats with established PAH. These results support the notion that RAN can improve the electrical and functional properties of the right ventricle, highlighting its potential benefits in the setting of RV impairment.
Collapse
Affiliation(s)
- John T Liles
- Department of Biology, Gilead Sciences, Inc., Fremont, California
| | - Kirsten Hoyer
- Department of Biology, Gilead Sciences, Inc., Fremont, California
| | - Jason Oliver
- Department of Biology, Gilead Sciences, Inc., Fremont, California
| | - Liguo Chi
- Department of Biology, Gilead Sciences, Inc., Fremont, California
| | | | | |
Collapse
|
21
|
Guo A, Song LS. AutoTT: automated detection and analysis of T-tubule architecture in cardiomyocytes. Biophys J 2015; 106:2729-36. [PMID: 24940790 DOI: 10.1016/j.bpj.2014.05.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/06/2014] [Accepted: 05/12/2014] [Indexed: 12/31/2022] Open
Abstract
Cardiac transverse (T)-tubules provide a specialized structure for synchronization and stabilization of sarcoplasmic reticulum Ca(2+) release in healthy cardiomyocytes. The application of laser scanning confocal microscopy and the use of fluorescent lipophilic membrane dyes have boosted the discoveries that T-tubule remodeling is a significant factor contributing to cardiac contractile dysfunction. However, the analysis and quantification of the remodeling of T-tubules have been a challenge and remain inconsistent among different research laboratories. Fast Fourier transformation (FFT) is the major analysis method applied to calculate the spatial frequency spectrum, which is used to represent the regularity of T-tubule systems. However, this approach is flawed because the density of T-tubules as well as non-T-tubule signals in the images influence the spectrum power generated by FFT. Preprocessing of images and topological architecture extracting is necessary to remove non-T-tubule noise from the analysis. In addition, manual analysis of images is time consuming and prone to errors and investigator bias. Therefore, we developed AutoTT, an automated analysis program that incorporates image processing, morphological feature extraction, and FFT analysis of spectrum power. The underlying algorithm is implemented in MATLAB (The MathWorks, Natick, MA). The program outputs the densities of transversely oriented T-tubules and longitudinally oriented T-tubules, power spectrum of the overall T-tubule systems, and averaged spacing of T-tubules. We also combined the density and regularity of T-tubules to give an index of T-tubule integrity (TTint), which provides a global evaluation of T-tubule alterations. In summary, AutoTT provides a reliable, easy to use, and fast approach for analyzing myocyte T-tubules. This program can also be applied to measure the density and integrity of other cellular structures.
Collapse
Affiliation(s)
- Ang Guo
- Division of Cardiovascular Medicine, Department of Internal Medicine and Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Long-Sheng Song
- Division of Cardiovascular Medicine, Department of Internal Medicine and Francois M. Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
22
|
Nivala M, Song Z, Weiss JN, Qu Z. T-tubule disruption promotes calcium alternans in failing ventricular myocytes: mechanistic insights from computational modeling. J Mol Cell Cardiol 2014; 79:32-41. [PMID: 25450613 DOI: 10.1016/j.yjmcc.2014.10.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/25/2014] [Accepted: 10/28/2014] [Indexed: 10/24/2022]
Abstract
In heart failure (HF), T-tubule (TT) disruption contributes to dyssynchronous calcium (Ca) release and impaired contraction, but its role in arrhythmogenesis remains unclear. In this study, we investigate the effects of TT disruption and other HF remodeling factors on Ca alternans in ventricular myocytes using computer modeling. A ventricular myocyte model with detailed spatiotemporal Ca cycling modeled by a coupled Ca release unit (CRU) network was used, in which the L-type Ca channels and the ryanodine receptor (RyR) channels were simulated by random Markov transitions. TT disruption, which removes the L-type Ca channels from the associated CRUs, results in "orphaned" RyR clusters and thus provides increased opportunity for spark-induced Ca sparks to occur. This effect combined with other HF remodeling factors promoted alternans by two distinct mechanisms: 1) for normal sarco-endoplasmic reticulum Ca ATPase (SERCA) activity, alternans was caused by both CRU refractoriness and coupling. The increased opportunity for spark-induced sparks by TT disruption combined with the enhanced CRU coupling by Ca elevation in the presence or absence of increased RyR leakiness facilitated spark synchronization on alternate beats to promote Ca alternans; 2) for down-regulated SERCA, alternans was caused by the sarcoplasmic reticulum (SR) Ca load-dependent mechanism, independent of CRU refractoriness. TT disruption and increased RyR leakiness shifted and steepened the SR Ca release-load relationship, which combines with down-regulated SERCA to promote Ca alternans. In conclusion, the mechanisms of Ca alternans for normal and down-regulated SERCA are different, and TT disruption promotes Ca alternans by both mechanisms, which may contribute to alternans at different stages of HF.
Collapse
Affiliation(s)
- Michael Nivala
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Zhen Song
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - James N Weiss
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Zhilin Qu
- Department of Medicine (Cardiology), David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
23
|
Pourrier M, Williams S, McAfee D, Belardinelli L, Fedida D. CrossTalk proposal: The late sodium current is an important player in the development of diastolic heart failure (heart failure with a preserved ejection fraction). J Physiol 2014; 592:411-4. [PMID: 24488066 DOI: 10.1113/jphysiol.2013.262261] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
24
|
Rocchetti M, Sala L, Rizzetto R, Staszewsky LI, Alemanni M, Zambelli V, Russo I, Barile L, Cornaghi L, Altomare C, Ronchi C, Mostacciuolo G, Lucchetti J, Gobbi M, Latini R, Zaza A. Ranolazine prevents INaL enhancement and blunts myocardial remodelling in a model of pulmonary hypertension. Cardiovasc Res 2014; 104:37-48. [DOI: 10.1093/cvr/cvu188] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
25
|
Williams S, Pourrier M, McAfee D, Lin S, Fedida D. Ranolazine improves diastolic function in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol 2014; 306:H867-81. [PMID: 24464752 DOI: 10.1152/ajpheart.00704.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diastolic dysfunction can lead to heart failure with preserved ejection fraction, for which there is no effective therapeutic. Ranolazine has been reported to reduce diastolic dysfunction, but the specific mechanisms of action are unclear. The effect of ranolazine on diastolic function was examined in spontaneously hypertensive rats (SHRs), where left ventricular relaxation is impaired and stiffness increased. The objective of this study was to determine whether ranolazine improves diastolic function in SHRs and identify the mechanism(s) by which improvement is achieved. Specifically, to test the hypothesis that ranolazine, by inhibiting late sodium current, reduces Ca(2+) overload and promotes ventricular relaxation and reduction in diastolic stiffness, the effects of ranolazine or vehicle on heart function and the response to dobutamine challenge were evaluated in aged male SHRs and Wistar-Kyoto rats by echocardiography and pressure-volume loop analysis. The effects of ranolazine and the more specific sodium channel inhibitor tetrodotoxin were determined on the late sodium current, sarcomere length, and intracellular calcium in isolated cardiomyocytes. Ranolazine reduced the end-diastolic pressure-volume relationship slope and improved diastolic function during dobutamine challenge in the SHR. Ranolazine and tetrodotoxin also enhanced cardiomyocyte relaxation and reduced myoplasmic free Ca(2+) during diastole at high-stimulus rates in the SHR. The density of the late sodium current was elevated in SHRs. In conclusion, ranolazine was effective in reducing diastolic dysfunction in the SHR. Its mechanism of action, at least in part, is consistent with inhibition of the increased late sodium current in the SHR leading to reduced Ca(2+) overload.
Collapse
Affiliation(s)
- Sarah Williams
- Department of Anesthesiology, Pharmacology, and Therapeutics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | | | | | | | | |
Collapse
|
26
|
Luo A, Ma J, Song Y, Qian C, Wu Y, Zhang P, Wang L, Fu C, Cao Z, Shryock JC. Larger late sodium current density as well as greater sensitivities to ATX II and ranolazine in rabbit left atrial than left ventricular myocytes. Am J Physiol Heart Circ Physiol 2013; 306:H455-61. [PMID: 24322614 DOI: 10.1152/ajpheart.00727.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
An increase of cardiac late sodium current (INa.L) is arrhythmogenic in atrial and ventricular tissues, but the densities of INa.L and thus the potential relative contributions of this current to sodium ion (Na(+)) influx and arrhythmogenesis in atria and ventricles are unclear. In this study, whole-cell and cell-attached patch-clamp techniques were used to measure INa.L in rabbit left atrial and ventricular myocytes under identical conditions. The density of INa.L was 67% greater in left atrial (0.50 ± 0.09 pA/pF, n = 20) than in left ventricular cells (0.30 ± 0.07 pA/pF, n = 27, P < 0.01) when elicited by step pulses from -120 to -20 mV at a rate of 0.2 Hz. Similar results were obtained using step pulses from -90 to -20 mV. Anemone toxin II (ATX II) increased INa.L with an EC50 value of 14 ± 2 nM and a Hill slope of 1.4 ± 0.1 (n = 9) in atrial myocytes and with an EC50 of 21 ± 5 nM and a Hill slope of 1.2 ± 0.1 (n = 12) in ventricular myocytes. Na(+) channel open probability (but not mean open time) was greater in atrial than in ventricular cells in the absence and presence of ATX II. The INa.L inhibitor ranolazine (3, 6, and 9 μM) reduced INa.L more in atrial than ventricular myocytes in the presence of 40 nM ATX II. In summary, rabbit left atrial myocytes have a greater density of INa.L and higher sensitivities to ATX II and ranolazine than rabbit left ventricular myocytes.
Collapse
Affiliation(s)
- Antao Luo
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|