1
|
Ruud M, Frisk M, Melleby AO, Norseng PA, Mohamed BA, Li J, Aronsen JM, Setterberg IE, Jakubiczka J, van Hout I, Coffey S, Shen X, Nygård S, Lunde IG, Tønnessen T, Jones PP, Sjaastad I, Gullestad L, Toischer K, Dahl CP, Christensen G, Louch WE. Regulation of cardiomyocyte t-tubule structure by preload and afterload: Roles in cardiac compensation and decompensation. J Physiol 2024; 602:4487-4510. [PMID: 38686538 DOI: 10.1113/jp284566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Mechanical load is a potent regulator of cardiac structure and function. Although high workload during heart failure is associated with disruption of cardiomyocyte t-tubules and Ca2+ homeostasis, it remains unclear whether changes in preload and afterload may promote adaptive t-tubule remodelling. We examined this issue by first investigating isolated effects of stepwise increases in load in cultured rat papillary muscles. Both preload and afterload increases produced a biphasic response, with the highest t-tubule densities observed at moderate loads, whereas excessively low and high loads resulted in low t-tubule levels. To determine the baseline position of the heart on this bell-shaped curve, mice were subjected to mildly elevated preload or afterload (1 week of aortic shunt or banding). Both interventions resulted in compensated cardiac function linked to increased t-tubule density, consistent with ascension up the rising limb of the curve. Similar t-tubule proliferation was observed in human patients with moderately increased preload or afterload (mitral valve regurgitation, aortic stenosis). T-tubule growth was associated with larger Ca2+ transients, linked to upregulation of L-type Ca2+ channels, Na+-Ca2+ exchanger, mechanosensors and regulators of t-tubule structure. By contrast, marked elevation of cardiac load in rodents and patients advanced the heart down the declining limb of the t-tubule-load relationship. This bell-shaped relationship was lost in the absence of electrical stimulation, indicating a key role of systolic stress in controlling t-tubule plasticity. In conclusion, modest augmentation of workload promotes compensatory increases in t-tubule density and Ca2+ cycling, whereas this adaptation is reversed in overloaded hearts during heart failure progression. KEY POINTS: Excised papillary muscle experiments demonstrated a bell-shaped relationship between cardiomyocyte t-tubule density and workload (preload or afterload), which was only present when muscles were electrically stimulated. The in vivo heart at baseline is positioned on the rising phase of this curve because moderate increases in preload (mice with brief aortic shunt surgery, patients with mitral valve regurgitation) resulted in t-tubule growth. Moderate increases in afterload (mice and patients with mild aortic banding/stenosis) similarly increased t-tubule density. T-tubule proliferation was associated with larger Ca2+ transients, with upregulation of the L-type Ca2+ channel, Na+-Ca2+ exchanger, mechanosensors and regulators of t-tubule structure. By contrast, marked elevation of cardiac load in rodents and patients placed the heart on the declining phase of the t-tubule-load relationship, promoting heart failure progression. The dependence of t-tubule structure on preload and afterload thus enables both compensatory and maladaptive remodelling, in rodents and humans.
Collapse
Affiliation(s)
- Marianne Ruud
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Arne Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Per Andreas Norseng
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Belal A Mohamed
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jan Magnus Aronsen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Joanna Jakubiczka
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Isabelle van Hout
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Sean Coffey
- Department of Medicine and HeartOtago, Dunedin School of Medicine, Dunedin Hospital, Dunedin, New Zealand
| | - Xin Shen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Ståle Nygård
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
- Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway
| | - Peter P Jones
- Department of Physiology, School of Biomedical Sciences and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Lars Gullestad
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Karl Toischer
- Department of Cardiology and Pneumology, Georg-August-University, Göttingen, Germany
| | - Cristen P Dahl
- Department of Cardiology, Oslo University Hospital, Oslo, Norway
| | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- K.G. Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
2
|
Khomtchouk BB, Lee YS, Khan ML, Sun P, Mero D, Davidson MH. Targeting the cytoskeleton and extracellular matrix in cardiovascular disease drug discovery. Expert Opin Drug Discov 2022; 17:443-460. [PMID: 35258387 PMCID: PMC9050939 DOI: 10.1080/17460441.2022.2047645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/24/2022] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Currently, cardiovascular disease (CVD) drug discovery has focused primarily on addressing the inflammation and immunopathology aspects inherent to various CVD phenotypes such as cardiac fibrosis and coronary artery disease. However, recent findings suggest new biological pathways for cytoskeletal and extracellular matrix (ECM) regulation across diverse CVDs, such as the roles of matricellular proteins (e.g. tenascin-C) in regulating the cellular microenvironment. The success of anti-inflammatory drugs like colchicine, which targets microtubule polymerization, further suggests that the cardiac cytoskeleton and ECM provide prospective therapeutic opportunities. AREAS COVERED Potential therapeutic targets include proteins such as gelsolin and calponin 2, which play pivotal roles in plaque development. This review focuses on the dynamic role that the cytoskeleton and ECM play in CVD pathophysiology, highlighting how novel target discovery in cytoskeletal and ECM-related genes may enable therapeutics development to alter the regulation of cellular architecture in plaque formation and rupture, cardiac contractility, and other molecular mechanisms. EXPERT OPINION Further research into the cardiac cytoskeleton and its associated ECM proteins is an area ripe for novel target discovery. Furthermore, the structural connection between the cytoskeleton and the ECM provides an opportunity to evaluate both entities as sources of potential therapeutic targets for CVDs.
Collapse
Affiliation(s)
- Bohdan B. Khomtchouk
- University of Chicago, Department of Medicine, Section of Computational Biomedicine and Biomedical Data Science, Institute for Genomics and Systems Biology, Chicago, IL USA
| | - Yoon Seo Lee
- The College of the University of Chicago, Chicago, IL USA
| | - Maha L. Khan
- The College of the University of Chicago, Chicago, IL USA
| | - Patrick Sun
- The College of the University of Chicago, Chicago, IL USA
| | | | - Michael H. Davidson
- University of Chicago, Department of Medicine, Section of Cardiology, Chicago, IL USA
| |
Collapse
|
3
|
Meng Q, Zhou L, Liang H, Hu A, Zhou H, Zhou J, Zhou X, Lin H, Li X, Jiang L, Dong J. Spine‑specific downregulation of LAPTM5 expression promotes the progression and spinal metastasis of estrogen receptor‑positive breast cancer by activating glutamine‑dependent mTOR signaling. Int J Oncol 2022; 60:47. [PMID: 35294039 PMCID: PMC8923652 DOI: 10.3892/ijo.2022.5337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/24/2022] [Indexed: 11/05/2022] Open
Abstract
Estrogen receptor-positive (ER+) breast cancer (BC) is a malignancy that is prone to metastasis to the spine, which is difficult to treat and often results in poor prognosis. However, the mechanism underlying the tumorigenesis and spinal metastasis of ER+ BC remains unclear. Lysosomal protein transmembrane 5 (LAPTM5) has been reported as a tumor suppressor in several types of cancer, but its role in ER+ BC has not been described. Here, by analyzing a gene sequencing dataset and ER+ BC tissues, tumor-adjacent normal tissues and spinal metastatic tissues from patients and mouse models, we found that LAPTM5 expression is negatively related to the progression and spinal metastasis of ER+ BC. Subsequently, in vitro experiments demonstrated that downregulation of LAPTM5 expression promoted the proliferation, migration, and chemoresistance of ER+ BC cells by activating glutamine-dependent mTOR signaling. A high level of CX3CL1 could inhibit LAPTM5 expression, explaining how ER+ BC metastasized to the spine. Thus, we found that LAPTM5 functions as a tumor suppressor in ER+ BC and that the CX3CL/CX3CR1/LAPTM5/glutamine axis mediates the spinal metastasis of ER+ BC. This axis may be a promising therapeutic target for ER+ BC.
Collapse
Affiliation(s)
- Qingbing Meng
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Lei Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Haifeng Liang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Annan Hu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Hao Zhou
- Department of Orthopedic Surgery, Xuhui‑Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jian Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xiaogang Zhou
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Hong Lin
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Xilei Li
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Libo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Jian Dong
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
4
|
Liang X, Bai Z, Wang F, Han Y, Sun H, Xiaokereti J, Zhang L, Zhou X, Lu Y, Tang B. Full-Length Transcriptome Sequencing: An Insight Into the Dog Model of Heart Failure. Front Cardiovasc Med 2021; 8:712797. [PMID: 34977163 PMCID: PMC8716442 DOI: 10.3389/fcvm.2021.712797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Heart failure (HF) leads to a progressive increase in morbidity and mortality rates. This study aimed to explore the transcriptional landscape during HF and identify differentially expressed transcripts (DETs) and alternative splicing events associated with HF. We generated a dog model of HF (n = 3) using right ventricular pacemaker implantation. We performed full-length transcriptome sequencing (based on nanopore platform) on the myocardial tissues and analyzed the transcripts using differential expression analysis and functional annotation methods [Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses]. Additionally, we estimated the expression of the selected genes by quantitative real-time PCR (qRT-PCR) and detected the proportion of immune cells using flow cytometry. We found that increased B-type natriuretic peptide reduced ejection fraction, and apparent clinical signs were observed in the dog model of HF. We identified 67,458 transcripts using full-length transcriptome sequencing. A total of 785 DETs were obtained from the HF and control groups. These DETs were mainly enriched in the immune responses, especially Th1, Th2, and Th17 cell differentiation processes. Furthermore, flow cytometry results revealed that the proportion of Th1 and Th17 cells increased in patients with HF compared to controls, while the proportion of Th2 cells decreased. Differentially expressed genes in the HF and control groups associated with Th1, Th2, and Th17 cell differentiation were quantified using qRT-PCR. We also identified variable splicing events of sarcomere genes (e.g., MYBPC3, TNNT2, TTN, FLNC, and TTNI3). In addition, we detected 4,892 transcription factors and 406 lncRNAs associated with HF. Our analysis based on full-length transcript sequencing provided an analysis perspective in a dog model of HF, which is valuable for molecular research in an increasingly relevant large animal model of HF.
Collapse
Affiliation(s)
- Xiaoyan Liang
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Zechen Bai
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Feifei Wang
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang First Aid Center, People's Hospital of Xinjiang Uygur Autonomous Region, Ürümqi, China
| | - Yafan Han
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Huaxin Sun
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Jiasuoer Xiaokereti
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Ling Zhang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Xianhui Zhou
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Yanmei Lu
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- *Correspondence: Baopeng Tang
| | - Baopeng Tang
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Cardiac Remodeling, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
- Yanmei Lu
| |
Collapse
|
5
|
Setterberg IE, Le C, Frisk M, Li J, Louch WE. The Physiology and Pathophysiology of T-Tubules in the Heart. Front Physiol 2021; 12:718404. [PMID: 34566684 PMCID: PMC8458775 DOI: 10.3389/fphys.2021.718404] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/07/2021] [Indexed: 12/18/2022] Open
Abstract
In cardiomyocytes, invaginations of the sarcolemmal membrane called t-tubules are critically important for triggering contraction by excitation-contraction (EC) coupling. These structures form functional junctions with the sarcoplasmic reticulum (SR), and thereby enable close contact between L-type Ca2+ channels (LTCCs) and Ryanodine Receptors (RyRs). This arrangement in turn ensures efficient triggering of Ca2+ release, and contraction. While new data indicate that t-tubules are capable of exhibiting compensatory remodeling, they are also widely reported to be structurally and functionally compromised during disease, resulting in disrupted Ca2+ homeostasis, impaired systolic and/or diastolic function, and arrhythmogenesis. This review summarizes these findings, while highlighting an emerging appreciation of the distinct roles of t-tubules in the pathophysiology of heart failure with reduced and preserved ejection fraction (HFrEF and HFpEF). In this context, we review current understanding of the processes underlying t-tubule growth, maintenance, and degradation, underscoring the involvement of a variety of regulatory proteins, including junctophilin-2 (JPH2), amphiphysin-2 (BIN1), caveolin-3 (Cav3), and newer candidate proteins. Upstream regulation of t-tubule structure/function by cardiac workload and specifically ventricular wall stress is also discussed, alongside perspectives for novel strategies which may therapeutically target these mechanisms.
Collapse
Affiliation(s)
- Ingunn E Setterberg
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Christopher Le
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - Jia Li
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Centre for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
6
|
Clerx M, Mirams GR, Rogers AJ, Narayan SM, Giles WR. Immediate and Delayed Response of Simulated Human Atrial Myocytes to Clinically-Relevant Hypokalemia. Front Physiol 2021; 12:651162. [PMID: 34122128 PMCID: PMC8188899 DOI: 10.3389/fphys.2021.651162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/22/2021] [Indexed: 12/18/2022] Open
Abstract
Although plasma electrolyte levels are quickly and precisely regulated in the mammalian cardiovascular system, even small transient changes in K+, Na+, Ca2+, and/or Mg2+ can significantly alter physiological responses in the heart, blood vessels, and intrinsic (intracardiac) autonomic nervous system. We have used mathematical models of the human atrial action potential (AP) to explore the electrophysiological mechanisms that underlie changes in resting potential (Vr) and the AP following decreases in plasma K+, [K+]o, that were selected to mimic clinical hypokalemia. Such changes may be associated with arrhythmias and are commonly encountered in patients (i) in therapy for hypertension and heart failure; (ii) undergoing renal dialysis; (iii) with any disease with acid-base imbalance; or (iv) post-operatively. Our study emphasizes clinically-relevant hypokalemic conditions, corresponding to [K+]o reductions of approximately 1.5 mM from the normal value of 4 to 4.5 mM. We show how the resulting electrophysiological responses in human atrial myocytes progress within two distinct time frames: (i) Immediately after [K+]o is reduced, the K+-sensing mechanism of the background inward rectifier current (IK1) responds. Specifically, its highly non-linear current-voltage relationship changes significantly as judged by the voltage dependence of its region of outward current. This rapidly alters, and sometimes even depolarizes, Vr and can also markedly prolong the final repolarization phase of the AP, thus modulating excitability and refractoriness. (ii) A second much slower electrophysiological response (developing 5-10 minutes after [K+]o is reduced) results from alterations in the intracellular electrolyte balance. A progressive shift in intracellular [Na+]i causes a change in the outward electrogenic current generated by the Na+/K+ pump, thereby modifying Vr and AP repolarization and changing the human atrial electrophysiological substrate. In this study, these two effects were investigated quantitatively, using seven published models of the human atrial AP. This highlighted the important role of IK1 rectification when analyzing both the mechanisms by which [K+]o regulates Vr and how the AP waveform may contribute to "trigger" mechanisms within the proarrhythmic substrate. Our simulations complement and extend previous studies aimed at understanding key factors by which decreases in [K+]o can produce effects that are known to promote atrial arrhythmias in human hearts.
Collapse
Affiliation(s)
- Michael Clerx
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Gary R Mirams
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Albert J Rogers
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Sanjiv M Narayan
- Department of Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Wayne R Giles
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
7
|
Caffarra Malvezzi C, Cabassi A, Miragoli M. Mitochondrial mechanosensor in cardiovascular diseases. VASCULAR BIOLOGY 2020; 2:R85-R92. [PMID: 32923977 PMCID: PMC7439846 DOI: 10.1530/vb-20-0002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/22/2020] [Indexed: 12/26/2022]
Abstract
The role of mitochondria in cardiac tissue is of utmost importance due to the dynamic nature of the heart and its energetic demands, necessary to assure its proper beating function. Recently, other important mitochondrial roles have been discovered, namely its contribution to intracellular calcium handling in normal and pathological myocardium. Novel investigations support the fact that during the progression toward heart failure, mitochondrial calcium machinery is compromised due to its morphological, structural and biochemical modifications resulting in facilitated arrhythmogenesis and heart failure development. The interaction between mitochondria and sarcomere directly affect cardiomyocyte excitation-contraction and is also involved in mechano-transduction through the cytoskeletal proteins that tether together the mitochondria and the sarcoplasmic reticulum. The focus of this review is to briefly elucidate the role of mitochondria as (mechano) sensors in the heart.
Collapse
Affiliation(s)
| | - Aderville Cabassi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Michele Miragoli
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,Center of Excellence for Toxicological Research, Department of Medicine and Surgery, University of Parma, Parma, Italy.,Department of Cardiovascular Medicine, Humanitas Clinical and Research Center - IRCCS, 20090 Rozzano, Milan, Italy
| |
Collapse
|
8
|
Pereira AHM, Cardoso AC, Consonni SR, Oliveira RR, Saito A, Vaggione MLB, Matos-Souza JR, Carazzolle MF, Gonçalves A, Fernandes JL, Ribeiro GCA, Lopes MM, Molkentin JD, Franchini KG. MEF2C repressor variant deregulation leads to cell cycle re-entry and development of heart failure. EBioMedicine 2020; 51:102571. [PMID: 31911274 PMCID: PMC6948164 DOI: 10.1016/j.ebiom.2019.11.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 11/07/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
Background A pathophysiological link exists between dysregulation of MEF2C transcription factors and heart failure (HF), but the underlying mechanisms remain elusive. Alternative splicing of MEF2C exons α, β and γ provides transcript diversity with gene activation or repression functionalities. Methods Neonatal and adult rat ventricular myocytes were used to overexpress MEF2C splicing variants γ+ (repressor) or γ-, or the inactive MEF2Cγ+23/24 (K23T/R24L). Phenotypic alterations in cardiomyocytes were determined by confocal and electron microscopy, flow cytometry and DNA microarray. We used transgenic mice with cardiac-specific overexpression of MEF2Cγ+ or MEF2Cγ− to explore the impact of MEF2C variants in cardiac phenotype. Samples of non-infarcted areas of the left ventricle from patients and mouse model of myocardial infarction were used to detect the expression of MEF2Cγ+ in failing hearts. Findings We demonstrate a previously unrealized upregulation of the transrepressor MEF2Cγ+ isoform in human and mouse failing hearts. We show that adenovirus-mediated overexpression of MEF2Cγ+ downregulates multiple MEF2-target genes, and drives incomplete cell-cycle reentry, partial dedifferentiation and apoptosis in the neonatal and adult rat. None of these changes was observed in cardiomyocytes overexpressing MEF2Cγ-. Transgenic mice overexpressing MEF2Cγ+, but not the MEF2Cγ-, developed dilated cardiomyopathy, correlated to cell-cycle reentry and apoptosis of cardiomyocytes. Interpretation Our results provide a mechanistic link between MEF2Cγ+ and deleterious abnormalities in cardiomyocytes, supporting the notion that splicing dysregulation in MEF2C towards the selection of the MEF2Cγ+ variant contributes to the pathogenesis of HF by promoting cardiomyocyte dropout. Funding São Paulo Research Foundation (FAPESP); Brazilian National Research Council (CNPq).
Collapse
Affiliation(s)
- Ana Helena M Pereira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, Sao Paulo, Brazil
| | - Alisson C Cardoso
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, Sao Paulo, Brazil
| | - Silvio R Consonni
- Department of Biochemistry and Tissue Biology, University of Campinas, Campinas, Brazil
| | - Renata R Oliveira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, Sao Paulo, Brazil
| | - Angela Saito
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, Sao Paulo, Brazil
| | - Maria Luisa B Vaggione
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, Sao Paulo, Brazil
| | - Jose R Matos-Souza
- Department of Internal Medicine, University of Campinas, Campinas, Brazil
| | | | - Anderson Gonçalves
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, Sao Paulo, Brazil
| | | | | | | | - Jeffery D Molkentin
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, USA
| | - Kleber G Franchini
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), 13083-970 Campinas, Sao Paulo, Brazil; Department of Internal Medicine, University of Campinas, Campinas, Brazil.
| |
Collapse
|
9
|
Martens P, Dupont M, Dauw J, Somers F, Herbots L, Timmermans P, Verwerft J, Mullens W. Rationale and design of the IRON-CRT trial: effect of intravenous ferric carboxymaltose on reverse remodelling following cardiac resynchronization therapy. ESC Heart Fail 2019; 6:1208-1215. [PMID: 31562751 PMCID: PMC6989286 DOI: 10.1002/ehf2.12503] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/14/2019] [Accepted: 07/01/2019] [Indexed: 12/28/2022] Open
Abstract
AIMS Iron deficiency is common in heart failure with reduced ejection fraction (HFrEF). In patients with cardiac resynchronization therapy (CRT), it is associated with a diminished reverse remodelling response and poor functional improvement. The latter is partially related to a loss in contractile force at higher heart rates (negative force-frequency relationship). METHODS AND RESULTS The effect of intravenous ferric carboxymaltose on reverse remodelling following cardiac resynchronization therapy (IRON-CRT) trial is a multicentre, prospective, randomized, double-blind controlled trial in HFrEF patients who experienced incomplete reverse remodelling (defined as a left ventricular ejection fraction below <45%) at least 6 months after CRT. Additionally, patients need to have iron deficiency defined as a ferritin below 100 μg/L irrespective of transferrin saturation or a ferritin between 100 and 300 μg/L with a transferrin saturation <20%. Patients will be randomized to either intravenous ferric carboxymaltose (dose based according to Summary of Product Characteristics) or intravenous placebo. The primary objective is to evaluate the effect of ferric carboxymaltose on metrics of cardiac reverse remodelling and contractility, measured by the primary endpoint, change in left ventricular ejection fraction assessed by three-dimensional (3D) echo from baseline to 3 month follow-up and the secondary endpoints change in left ventricular end-systolic and end-diastolic volume. The secondary objective is to determine if ferric carboxymaltose is capable of improving cardiac contractility in vivo, by assessing the force-frequency relationship through incremental biventricular pacing. A total of 100 patients will be randomized in a 1:1 fashion. CONCLUSIONS The IRON-CRT trial will determine the effect of ferric carboxymaltose on cardiac reverse remodelling and rate-dependent cardiac contractility in HFrEF patients.
Collapse
Affiliation(s)
- Pieter Martens
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium.,Doctoral School for Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Matthias Dupont
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Jeroen Dauw
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Frauke Somers
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - Lieven Herbots
- Department of Cardiology, Jessa Ziekenhuis, Hasselt, Belgium
| | | | - Jan Verwerft
- Department of Cardiology, Jessa Ziekenhuis, Hasselt, Belgium
| | - Wilfried Mullens
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium.,Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
10
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
11
|
Antoniou CK, Manolakou P, Magkas N, Konstantinou K, Chrysohoou C, Dilaveris P, Gatzoulis KA, Tousoulis D. Cardiac Resynchronisation Therapy and Cellular Bioenergetics: Effects Beyond Chamber Mechanics. Eur Cardiol 2019; 14:33-44. [PMID: 31131035 PMCID: PMC6523053 DOI: 10.15420/ecr.2019.2.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cardiac resynchronisation therapy is a cornerstone in the treatment of advanced dyssynchronous heart failure. However, despite its widespread clinical application, precise mechanisms through which it exerts its beneficial effects remain elusive. Several studies have pointed to a metabolic component suggesting that, both in concert with alterations in chamber mechanics and independently of them, resynchronisation reverses detrimental changes to cellular metabolism, increasing energy efficiency and metabolic reserve. These actions could partially account for the existence of responders that improve functionally but not echocardiographically. This article will attempt to summarise key components of cardiomyocyte metabolism in health and heart failure, with a focus on the dyssynchronous variant. Both chamber mechanics-related and -unrelated pathways of resynchronisation effects on bioenergetics – stemming from the ultramicroscopic level – and a possible common underlying mechanism relating mechanosensing to metabolism through the cytoskeleton will be presented. Improved insights regarding the cellular and molecular effects of resynchronisation on bioenergetics will promote our understanding of non-response, optimal device programming and lead to better patient care.
Collapse
Affiliation(s)
| | - Panagiota Manolakou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Nikolaos Magkas
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Konstantinos Konstantinou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Christina Chrysohoou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Polychronis Dilaveris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Konstantinos A Gatzoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| | - Dimitrios Tousoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens Athens, Greece
| |
Collapse
|
12
|
Colli DF, Blood SR, Sankarankutty AC, Sachse FB, Frisk M, Louch WE, Kekenes-Huskey PM. A Matched-Filter-Based Algorithm for Subcellular Classification of T-System in Cardiac Tissues. Biophys J 2019; 116:1386-1393. [PMID: 30979553 PMCID: PMC6486484 DOI: 10.1016/j.bpj.2019.03.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 11/26/2022] Open
Abstract
In mammalian ventricular cardiomyocytes, invaginations of the surface membrane form the transverse tubular system (T-system), which consists of transverse tubules (TTs) that align with sarcomeres and Z-lines as well as longitudinal tubules (LTs) that are present between Z-lines in some species. In many cardiac disease etiologies, the T-system is perturbed, which is believed to promote spatially heterogeneous, dyssynchronous Ca2+ release and inefficient contraction. In general, T-system characterization approaches have been directed primarily at isolated cells and do not detect subcellular T-system heterogeneity. Here, we present MatchedMyo, a matched-filter-based algorithm for subcellular T-system characterization in isolated cardiomyocytes and millimeter-scale myocardial sections. The algorithm utilizes "filters" representative of TTs, LTs, and T-system absence. Application of the algorithm to cardiomyocytes isolated from rat disease models of myocardial infarction (MI), dilated cardiomyopathy induced via aortic banding, and sham surgery confirmed and quantified heterogeneous T-system structure and remodeling. Cardiomyocytes from post-MI hearts exhibited increasing T-system disarray as proximity to the infarct increased. We found significant (p < 0.05, Welch's t-test) increases in LT density within cardiomyocytes proximal to the infarct (12 ± 3%, data reported as mean ± SD, n = 3) versus sham (4 ± 2%, n = 5), but not distal to the infarct (7 ± 1%, n = 3). The algorithm also detected decreases in TTs within 5° of the myocyte minor axis for isolated aortic banding (36 ± 9%, n = 3) and MI cardiomyocytes located intermediate (37 ± 4%, n = 3) and proximal (34 ± 4%, n = 3) to the infarct versus sham (57 ± 12%, n = 5). Application of bootstrapping to rabbit MI tissue revealed distal sections comprised 18.9 ± 1.0% TTs, whereas proximal sections comprised 10.1 ± 0.8% TTs (p < 0.05), a 46.6% decrease. The matched-filter approach therefore provides a robust and scalable technique for T-system characterization from isolated cells through millimeter-scale myocardial sections.
Collapse
Affiliation(s)
- Dylan F Colli
- Department of Chemistry, University of Kentucky, Lexington, Kentucky; Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky.
| | - S Ryan Blood
- Department of Chemistry, University of Kentucky, Lexington, Kentucky; Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky
| | - Aparna C Sankarankutty
- Nora Eccles Harrison Cardiovascular Research and Training Institute & Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute & Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Michael Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - William E Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; K.G. Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Peter M Kekenes-Huskey
- Department of Chemistry, University of Kentucky, Lexington, Kentucky; Department of Chemical and Materials Engineering, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
13
|
Jones PP, MacQuaide N, Louch WE. Dyadic Plasticity in Cardiomyocytes. Front Physiol 2018; 9:1773. [PMID: 30618792 PMCID: PMC6298195 DOI: 10.3389/fphys.2018.01773] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 11/23/2018] [Indexed: 11/13/2022] Open
Abstract
Contraction of cardiomyocytes is dependent on sub-cellular structures called dyads, where invaginations of the surface membrane (t-tubules) form functional junctions with the sarcoplasmic reticulum (SR). Within each dyad, Ca2+ entry through t-tubular L-type Ca2+ channels (LTCCs) elicits Ca2+ release from closely apposed Ryanodine Receptors (RyRs) in the SR membrane. The efficiency of this process is dependent on the density and macroscale arrangement of dyads, but also on the nanoscale organization of LTCCs and RyRs within them. We presently review accumulating data demonstrating the remarkable plasticity of these structures. Dyads are known to form gradually during development, with progressive assembly of both t-tubules and junctional SR terminals, and precise trafficking of LTCCs and RyRs. While dyads can exhibit compensatory remodeling when required, dyadic degradation is believed to promote impaired contractility and arrythmogenesis in cardiac disease. Recent data indicate that this plasticity of dyadic structure/function is dependent on the regulatory proteins junctophilin-2, amphiphysin-2 (BIN1), and caveolin-3, which critically arrange dyadic membranes while stabilizing the position and activity of LTCCs and RyRs. Indeed, emerging evidence indicates that clustering of both channels enables "coupled gating", implying that nanoscale localization and function are intimately linked, and may allow fine-tuning of LTCC-RyR crosstalk. We anticipate that improved understanding of dyadic plasticity will provide greater insight into the processes of cardiac compensation and decompensation, and new opportunities to target the basic mechanisms underlying heart disease.
Collapse
Affiliation(s)
- Peter P. Jones
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- HeartOtago, University of Otago, Dunedin, New Zealand
| | - Niall MacQuaide
- Institute of Cardiovascular Sciences, University of Glasgow, Glasgow, United Kingdom
- Clyde Biosciences, Glasgow, United Kingdom
| | - William E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital, University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
Radwański PB, Johnson CN, Györke S, Veeraraghavan R. Cardiac Arrhythmias as Manifestations of Nanopathies: An Emerging View. Front Physiol 2018; 9:1228. [PMID: 30233404 PMCID: PMC6131669 DOI: 10.3389/fphys.2018.01228] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
A nanodomain is a collection of proteins localized within a specialized, nanoscale structural environment, which can serve as the functional unit of macroscopic physiologic processes. We are beginning to recognize the key roles of cardiomyocyte nanodomains in essential processes of cardiac physiology such as electrical impulse propagation and excitation–contraction coupling (ECC). There is growing appreciation of nanodomain dysfunction, i.e., nanopathy, as a mechanistic driver of life-threatening arrhythmias in a variety of pathologies. Here, we offer an overview of current research on the role of nanodomains in cardiac physiology with particular emphasis on: (1) sodium channel-rich nanodomains within the intercalated disk that participate in cell-to-cell electrical coupling and (2) dyadic nanodomains located along transverse tubules that participate in ECC. The beat to beat function of cardiomyocytes involves three phases: the action potential, the calcium transient, and mechanical contraction/relaxation. In all these phases, cell-wide function results from the aggregation of the stochastic function of individual proteins. While it has long been known that proteins that exist in close proximity influence each other’s function, it is increasingly appreciated that there exist nanoscale structures that act as functional units of cardiac biophysical phenomena. Termed nanodomains, these structures are collections of proteins, localized within specialized nanoscale structural environments. The nano-environments enable the generation of localized electrical and/or chemical gradients, thereby conferring unique functional properties to these units. Thus, the function of a nanodomain is determined by its protein constituents as well as their local structural environment, adding an additional layer of complexity to cardiac biology and biophysics. However, with the emergence of experimental techniques that allow direct investigation of structure and function at the nanoscale, our understanding of cardiac physiology and pathophysiology at these scales is rapidly advancing. Here, we will discuss the structure and functions of multiple cardiomyocyte nanodomains, and novel strategies that target them for the treatment of cardiac arrhythmias.
Collapse
Affiliation(s)
- Przemysław B Radwański
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States.,Division of Pharmacy Practice and Science, College of Pharmacy, The Ohio State University, Columbus, OH, United States
| | - Christopher N Johnson
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Vanderbilt Center for Arrhythmia Research and Therapeutics, Nashville, TN, United States
| | - Sándor Györke
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Rengasayee Veeraraghavan
- Bob and Corinne Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH, United States.,Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
15
|
Rao P, Faddis M. Cardiac resynchronisation therapy: current indications, management and basic troubleshooting. Heart 2017; 103:2000-2007. [DOI: 10.1136/heartjnl-2016-310656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/11/2017] [Accepted: 06/26/2017] [Indexed: 01/14/2023] Open
|
16
|
Liu Y, Xia T, Wei J, Liu Q, Li X. Micropatterned co-culture of cardiac myocytes on fibrous scaffolds for predictive screening of drug cardiotoxicities. NANOSCALE 2017; 9:4950-4962. [PMID: 28382363 DOI: 10.1039/c7nr00001d] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The spatial arrangement of cardiac myocytes (CMs) and other non-myocytes scaffolds, closely resembling native tissue, is essential to control the CM morphology and function for cardiac tissue regeneration. In the current study, micropatterned fibrous scaffolds were developed to establish a CM co-culture system with cardiac fibroblasts (CFs) and endothelial cells (ECs) as a potential in vitro drug screening model. To pursue a biomimetic approach to influence CM behaviors, strip, oval and wave-patterned mats were constructed by deposition of electrospun fibers on lithographic collectors, followed by precise stacking for cell co-cultures. CMs, CFs, and ECs were located on the patterned scaffolds with controlled cellular distribution in the respective regions and no across condition was found. Compared with those after strip and oval-patterned co-culture, CMs co-cultured on wave-patterned scaffolds displayed significantly greater cell viabilities, larger cell elongation ratios, stronger expressions of cardiac-specific Troponin I, connexin 43 and sarcomeric α-actinin and higher beating rates during 15 days of incubation. The responses of co-cultured CMs to quinidine, erythromycin and sotalol show good correlations with clinical observations in the beating rate and the prolongation of the contraction and relaxation time. The in vivo safety data reflected well with the concentrations for 50% of maximal effect (EC50) after drug treatment on co-cultured CMs, which was determined from the changes in the corrected field potential duration (FPDc) against the drug concentrations. During 15 days of patterned co-culture, the interbeat intervals and fluctuations of the CMs indicated quick changes in response to haloperidol treatment and sufficient restoration of the original beating profiles after drug removal. This study demonstrates the capabilities of micropatterned co-culture of CMs to establish the cardiac function as a reproducible and reliable platform for screening cardiac side effects of drugs.
Collapse
Affiliation(s)
- Yaowen Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China.
| | | | | | | | | |
Collapse
|
17
|
Abstract
Dyssynchronous contraction of the ventricle significantly worsens morbidity and mortality in patients with heart failure (HF). Approximately one-third of patients with HF have cardiac dyssynchrony and are candidates for cardiac resynchronization therapy (CRT). The initial understanding of dyssynchrony and CRT was in terms of global mechanics and hemodynamics, but lack of clinical benefit in a sizable subgroup of recipients who appear otherwise appropriate has challenged this paradigm. This article reviews current understanding of these cellular and subcellular mechanisms, arguing that these aspects are key to improving CRT use, as well as translating its benefits to a wider HF population.
Collapse
Affiliation(s)
- Jonathan A Kirk
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 858, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 858, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
Martens P, Verbrugge F, Nijst P, Dupont M, Tang WHW, Mullens W. Impact of Iron Deficiency on Response to and Remodeling After Cardiac Resynchronization Therapy. Am J Cardiol 2017; 119:65-70. [PMID: 27780556 DOI: 10.1016/j.amjcard.2016.09.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/01/2016] [Accepted: 09/01/2016] [Indexed: 11/19/2022]
Abstract
Iron deficiency is prevalent in heart failure with reduced ejection fraction and relates to symptomatic status, readmission, and all-cause mortality. The relation between iron status and response to cardiac resynchronization therapy (CRT) remains insufficiently elucidated. This study assesses the impact of iron deficiency on clinical response and reverse cardiac remodeling and outcome after CRT. Baseline characteristics, change in New York Heart Association functional class, reverse cardiac remodeling on echocardiography, and clinical outcome (i.e., all-cause mortality and heart failure readmissions) were retrospectively evaluated in consecutive CRT patients who had full iron status and complete blood count available at implantation, implanted at a single tertiary care center with identical dedicated multidisciplinary CRT follow-up from October 2008 to August 2015. A total of 541 patients were included with mean follow-up of 38 ± 22 months. Prevalence of iron deficiency was 56% at implantation. Patients with iron deficiency exhibited less symptomatic improvement 6 months after implantation (p value <0.001). In addition, both the decrease in left ventricular end-diastolic diameter (-3.1 vs -6.2 mm; p value = 0.011) and improvement in ejection fraction (+11% vs +15%, p value = 0.001) were significantly lower in patients with iron deficiency. Iron deficiency was significantly associated with an increased risk for heart failure admission or all-cause mortality (adjusted hazard ratio 1.718, 95% confidence interval 1.178 to 2.506), irrespectively of the presence of anemia (Hemoglobin <12 g/dl in women and <13 g/dl in men). In conclusion, iron deficiency is prevalent and affects both clinical response and reverse cardiac remodeling after CRT implantation. Moreover, it is a powerful predictor of adverse clinical outcomes in CRT.
Collapse
Affiliation(s)
- Pieter Martens
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium; Doctoral School for Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | | | - Petra Nijst
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium; Doctoral School for Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Matthias Dupont
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium
| | - W H Wilson Tang
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio
| | - Wilfried Mullens
- Department of Cardiology, Ziekenhuis Oost-Limburg, Genk, Belgium; Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
19
|
Pasqualini FS, Nesmith AP, Horton RE, Sheehy SP, Parker KK. Mechanotransduction and Metabolism in Cardiomyocyte Microdomains. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4081638. [PMID: 28044126 PMCID: PMC5164897 DOI: 10.1155/2016/4081638] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 01/11/2023]
Abstract
Efficient contractions of the left ventricle are ensured by the continuous transfer of adenosine triphosphate (ATP) from energy production sites, the mitochondria, to energy utilization sites, such as ionic pumps and the force-generating sarcomeres. To minimize the impact of intracellular ATP trafficking, sarcomeres and mitochondria are closely packed together and in proximity with other ultrastructures involved in excitation-contraction coupling, such as t-tubules and sarcoplasmic reticulum junctions. This complex microdomain has been referred to as the intracellular energetic unit. Here, we review the literature in support of the notion that cardiac homeostasis and disease are emergent properties of the hierarchical organization of these units. Specifically, we will focus on pathological alterations of this microdomain that result in cardiac diseases through energy imbalance and posttranslational modifications of the cytoskeletal proteins involved in mechanosensing and transduction.
Collapse
Affiliation(s)
- Francesco S. Pasqualini
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Institute for Regenerative Medicine (IREM), Wyss Translational Center, University and ETH Zurich, Zurich, Switzerland
| | - Alexander P. Nesmith
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Renita E. Horton
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- James Worth Bagley College of Engineering and College of Agriculture and Life Sciences, Mississippi State University, Starkville, MS, USA
| | - Sean P. Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| |
Collapse
|
20
|
Finkenstaedt-Quinn SA, Qiu TA, Shin K, Haynes CL. Super-resolution imaging for monitoring cytoskeleton dynamics. Analyst 2016; 141:5674-5688. [PMID: 27549146 DOI: 10.1039/c6an00731g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The cytoskeleton is a key cellular structure that is important in the control of cellular movement, structure, and sensing. To successfully image the individual cytoskeleton components, high resolution and super-resolution fluorescence imaging methods are needed. This review covers the three basic cytoskeletal elements and the relative benefits and drawbacks of fixed versus live cell imaging before moving on to recent studies using high resolution and super-resolution techniques. The techniques covered include the near-diffraction limited imaging methods of confocal microscopy and TIRF microscopy and the super-resolution fluorescence imaging methods of STORM, PALM, and STED.
Collapse
|
21
|
Zhang L, Zhang N, Tang X, Liu F, Luo S, Xiao H. Increased α-Actinin-2 Expression in the Atrial Myocardium of Patients with Atrial Fibrillation Related to Rheumatic Heart Disease. Cardiology 2016; 135:151-159. [PMID: 27344599 DOI: 10.1159/000446362] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/22/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVES Atrial fibrosis, a marker of atrial structural remodeling, plays a critical role in atrial fibrillation (AF). α- Actinin-2 is associated with structural remodeling related to stretching. The transforming growth factor-β1 (TGF-β1)/Smad pathway plays an important role in atrial fibrosis. We investigated the effects of the TGF-β1/Smad signaling pathway on α-actinin-2 in atrial fibrosis in patients with AF. METHODS Forty-one right atrial specimens obtained from patients with rheumatic heart disease (RHD) were divided into a chronic (c)AF group, i.e. RHD + cAF (n = 29), and a sinus rhythm group, i.e. RHD + sinus rhythm (n = 12). Patients with congenital heart disease (CHD) and sinus rhythm who underwent heart surgery served as controls (n = 10). Fibrosis was assessed by histological examination, and expression of α-actinin-2, TGF-β1 and Smad2/phosphorylated Smad2 (p-Smad2) was evaluated by immunohistochemistry, quantitative real-time PCR and Western blotting. In rat atrial fibroblasts treated with TGF-β1, the collagen content was measured using hydroxyproline detection, and α-actinin-2 and p-Smad2 were evaluated by semiquantitative reverse-transcription PCR and Western blotting. RESULTS The histology results revealed a significant increase in atrial fibrosis in AF patients. The collagen content, mRNA and protein expression levels of α-actinin-2 and the components of the TGF-β1/Smad signaling pathway were significantly gradually increased in the CHD + sinus rhythm, RHD + sinus rhythm and RHD + cAF groups (p < 0.05). The mRNA and protein levels of α-actinin-2 and TGF-β1 in RHD patients were positively correlated with the collagen volume fraction. A positive correlation between the expression of α-actinin-2 and TGF-β1 was also observed. In rat atrial fibroblasts treated with TGF-β1, the collagen content was greater than that in the control group (p < 0.05), and the expression levels of α- actinin-2 and p-Smad2 were also upregulated (p < 0.05). CONCLUSIONS α-Actinin-2 expression was increased in the atrial tissues of patients with AF secondary to RHD. α-Actinin-2 was upregulated via the TGF-β1/Smad pathway in atrial fibroblasts, which suggests that it may be involved in TGF-β1/Smad pathway-induced atrial fibrosis in patients with AF.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
22
|
Abstract
Unlike diet and exercise, which individuals can modulate according to their lifestyle, aging is unavoidable. With normal or healthy aging, the heart undergoes extensive vascular, cellular, and interstitial molecular changes that result in stiffer less compliant hearts that experience a general decline in organ function. Although these molecular changes deemed cardiac remodeling were once thought to be concomitant with advanced cardiovascular disease, they can be found in patients without manifestation of clinical disease. It is now mostly acknowledged that these age-related mechanical changes confer vulnerability of the heart to cardiovascular stresses associated with disease, such as hypertension and atherosclerosis. However, recent studies have aimed at differentiating the initial compensatory changes that occur within the heart with age to maintain contractile function from the maladaptive responses associated with disease. This work has identified new targets to improve cardiac function during aging. Spanning invertebrate to vertebrate models, we use this review to delineate some hallmarks of physiological versus pathological remodeling that occur in the cardiomyocyte and its microenvironment, focusing especially on the mechanical changes that occur within the sarcomere, intercalated disc, costamere, and extracellular matrix.
Collapse
Affiliation(s)
- Ayla O Sessions
- From the Biomedical Sciences Program (A.O.S., A.J.E.) and Department of Bioengineering, University of California, San Diego, La Jolla (A.J.E.); and Sanford Consortium for Regenerative Medicine, La Jolla, CA (A.J.E.)
| | - Adam J Engler
- From the Biomedical Sciences Program (A.O.S., A.J.E.) and Department of Bioengineering, University of California, San Diego, La Jolla (A.J.E.); and Sanford Consortium for Regenerative Medicine, La Jolla, CA (A.J.E.).
| |
Collapse
|
23
|
Lichter J, Li H, Sachse FB. Measurement of Strain in Cardiac Myocytes at Micrometer Scale Based on Rapid Scanning Confocal Microscopy and Non-Rigid Image Registration. Ann Biomed Eng 2016; 44:3020-3031. [PMID: 27001400 DOI: 10.1007/s10439-016-1593-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/12/2016] [Indexed: 12/17/2022]
Abstract
Measurement of cell shortening is an important technique for assessment of physiology and pathophysiology of cardiac myocytes. Many types of heart disease are associated with decreased myocyte shortening, which is commonly caused by structural and functional remodeling. Here, we present a new approach for local measurement of 2-dimensional strain within cells at high spatial resolution. The approach applies non-rigid image registration to quantify local displacements and Cauchy strain in images of cells undergoing contraction. We extensively evaluated the approach using synthetic cell images and image sequences from rapid scanning confocal microscopy of fluorescently labeled isolated myocytes from the left ventricle of normal and diseased canine heart. Application of the approach yielded a comprehensive description of cellular strain including novel measurements of transverse strain and spatial heterogeneity of strain. Quantitative comparison with manual measurements of strain in image sequences indicated reliability of the developed approach. We suggest that the developed approach provides researchers with a novel tool to investigate contractility of cardiac myocytes at subcellular scale. In contrast to previously introduced methods for measuring cell shorting, the developed approach provides comprehensive information on the spatio-temporal distribution of 2-dimensional strain at micrometer scale.
Collapse
Affiliation(s)
- J Lichter
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT, 84112-5000, USA.,Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Hui Li
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT, 84112-5000, USA
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT, 84112-5000, USA. .,Department of Bioengineering, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
24
|
Abstract
Dyssynchronous contraction of the ventricle significantly worsens morbidity and mortality in patients with heart failure (HF). Approximately one-third of patients with HF have cardiac dyssynchrony and are candidates for cardiac resynchronization therapy (CRT). The initial understanding of dyssynchrony and CRT was in terms of global mechanics and hemodynamics, but lack of clinical benefit in a sizable subgroup of recipients who appear otherwise appropriate has challenged this paradigm. This article reviews current understanding of these cellular and subcellular mechanisms, arguing that these aspects are key to improving CRT use, as well as translating its benefits to a wider HF population.
Collapse
Affiliation(s)
- Jonathan A Kirk
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 858, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 858, 720 Rutland Avenue, Baltimore, MD 21205, USA
| |
Collapse
|
25
|
Katz AM, Rolett EL. Heart failure: when form fails to follow function. Eur Heart J 2015; 37:449-54. [PMID: 26497163 DOI: 10.1093/eurheartj/ehv548] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 09/23/2015] [Indexed: 12/16/2022] Open
Abstract
Cardiac performance is normally determined by architectural, cellular, and molecular structures that determine the heart's form, and by physiological and biochemical mechanisms that regulate the function of these structures. Impaired adaptation of form to function in failing hearts contributes to two syndromes initially called systolic heart failure (SHF) and diastolic heart failure (DHF). In SHF, characterized by high end-diastolic volume (EDV), the left ventricle (LV) cannot eject a normal stroke volume (SV); in DHF, with normal or low EDV, the LV cannot accept a normal venous return. These syndromes are now generally defined in terms of ejection fraction (EF): SHF became 'heart failure with reduced ejection fraction' (HFrEF) while DHF became 'heart failure with normal or preserved ejection fraction' (HFnEF or HFpEF). However, EF is a chimeric index because it is the ratio between SV--which measures function, and EDV--which measures form. In SHF the LV dilates when sarcomere addition in series increases cardiac myocyte length, whereas sarcomere addition in parallel can cause concentric hypertrophy in DHF by increasing myocyte thickness. Although dilatation in SHF allows the LV to accept a greater venous return, it increases the energy cost of ejection and initiates a vicious cycle that contributes to progressive dilatation. In contrast, concentric hypertrophy in DHF facilitates ejection but impairs filling and can cause heart muscle to deteriorate. Differences in the molecular signals that initiate dilatation and concentric hypertrophy can explain why many drugs that improve prognosis in SHF have little if any benefit in DHF.
Collapse
Affiliation(s)
- Arnold M Katz
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA University of Connecticut School of Medicine, Farmington, CT, USA
| | - Ellis L Rolett
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA Section of Cardiology, Dartmouth-Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA
| |
Collapse
|
26
|
Li H, Lichter JG, Seidel T, Tomaselli GF, Bridge JHB, Sachse FB. Cardiac Resynchronization Therapy Reduces Subcellular Heterogeneity of Ryanodine Receptors, T-Tubules, and Ca2+ Sparks Produced by Dyssynchronous Heart Failure. Circ Heart Fail 2015; 8:1105-14. [PMID: 26294422 DOI: 10.1161/circheartfailure.115.002352] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/17/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND Cardiac resynchronization therapy (CRT) is a major advance for treatment of patients with dyssynchronous heart failure (DHF). However, our understanding of DHF-associated remodeling of subcellular structure and function and their restoration after CRT remains incomplete. METHODS AND RESULTS We investigated subcellular heterogeneity of remodeling of structures and proteins associated with excitation-contraction coupling in cardiomyocytes in DHF and after CRT. Three-dimensional confocal microscopy revealed subcellular heterogeneity of ryanodine receptor (RyR) density and the transverse tubular system (t-system) in a canine model of DHF. RyR density at the ends of lateral left ventricular cardiomyocytes was higher than that in cell centers, whereas the t-system was depleted at cell ends. In anterior left ventricular cardiomyocytes, however, we found a similar degree of heterogeneous RyR remodeling, despite preserved t-system. Synchronous heart failure was associated with marginal heterogeneity of RyR density. We used rapid scanning confocal microscopy to investigate effects of heterogeneous structural remodeling on calcium signaling. In DHF, diastolic Ca(2+) spark density was smaller at cell ends versus centers. After CRT, subcellular heterogeneity of structures and function was reduced. CONCLUSIONS RyR density exhibits remarkable subcellular heterogeneity in DHF. RyR remodeling occurred in lateral and anterior cardiomyocytes, but remodeling of t-system was confined to lateral myocytes. These findings indicate that different mechanisms underlie remodeling of RyRs and t-system. Furthermore, we suggest that ventricular dyssynchrony exacerbates subcellular remodeling in heart failure. CRT efficiently reduced subcellular heterogeneity. These results will help to explain remodeling of excitation-contraction coupling in disease and restoration after CRT.
Collapse
Affiliation(s)
- Hui Li
- From the Nora Eccles Harrison Cardiovascular Research and Training Institute (H.L., J.G.L., T.S., J.H.B., F.B.S.), and Department of Bioengineering (J.G.L., F.B.S.), University of Utah, Salt Lake City; and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (G.F.T.)
| | - Justin G Lichter
- From the Nora Eccles Harrison Cardiovascular Research and Training Institute (H.L., J.G.L., T.S., J.H.B., F.B.S.), and Department of Bioengineering (J.G.L., F.B.S.), University of Utah, Salt Lake City; and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (G.F.T.)
| | - Thomas Seidel
- From the Nora Eccles Harrison Cardiovascular Research and Training Institute (H.L., J.G.L., T.S., J.H.B., F.B.S.), and Department of Bioengineering (J.G.L., F.B.S.), University of Utah, Salt Lake City; and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (G.F.T.)
| | - Gordon F Tomaselli
- From the Nora Eccles Harrison Cardiovascular Research and Training Institute (H.L., J.G.L., T.S., J.H.B., F.B.S.), and Department of Bioengineering (J.G.L., F.B.S.), University of Utah, Salt Lake City; and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (G.F.T.)
| | - John H B Bridge
- From the Nora Eccles Harrison Cardiovascular Research and Training Institute (H.L., J.G.L., T.S., J.H.B., F.B.S.), and Department of Bioengineering (J.G.L., F.B.S.), University of Utah, Salt Lake City; and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (G.F.T.)
| | - Frank B Sachse
- From the Nora Eccles Harrison Cardiovascular Research and Training Institute (H.L., J.G.L., T.S., J.H.B., F.B.S.), and Department of Bioengineering (J.G.L., F.B.S.), University of Utah, Salt Lake City; and Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD (G.F.T.).
| |
Collapse
|
27
|
Affiliation(s)
- Stanley F Fernandez
- From the Departments of Medicine, Biomedical Engineering, Physiology and Biophysics, VA WNY Health Care System, and Clinical and Translational Research Center, University at Buffalo, NY
| | - John M Canty
- From the Departments of Medicine, Biomedical Engineering, Physiology and Biophysics, VA WNY Health Care System, and Clinical and Translational Research Center, University at Buffalo, NY.
| |
Collapse
|
28
|
Nemutlu E, Zhang S, Xu YZ, Terzic A, Zhong L, Dzeja PD, Cha YM. Cardiac resynchronization therapy induces adaptive metabolic transitions in the metabolomic profile of heart failure. J Card Fail 2015; 21:460-9. [PMID: 25911126 DOI: 10.1016/j.cardfail.2015.04.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 01/20/2015] [Accepted: 04/10/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Heart failure (HF) is associated with ventricular dyssynchrony and energetic inefficiency, which can be alleviated by cardiac resynchronization therapy (CRT). The aim of this study was to determine the metabolomic signature in HF and its prognostic value regarding the response to CRT. METHODS AND RESULTS This prospective study consisted of 24 patients undergoing CRT for advanced HF and 10 control patients who underwent catheter ablation for supraventricular arrhythmia but not CRT. Blood samples were collected before and 3 months after CRT. Metabolomic profiling of plasma samples was performed with the use of gas chromatography-mass spectrometry and nuclear magnetic resonance. The plasma metabolomic profile was altered in the HF patients, with a distinct panel of metabolites, including Krebs cycle and lipid, amino acid, and nucleotide metabolism. CRT improved the metabolomic profile. The succinate-glutamate ratio, an index of Krebs cycle activity, improved from 0.58 ± 0.13 to 2.84 ± 0.60 (P < .05). The glucose-palmitate ratio, an indicator of the balance between glycolytic and fatty acid metabolism, increased from 0.96 ± 0.05 to 1.54 ± 0.09 (P < .01). Compared with nonresponders to CRT, responders had a distinct baseline plasma metabolomic profile, including higher isoleucine, phenylalanine, leucine, glucose, and valine levels and lower glutamate levels at baseline (P < .05). CONCLUSIONS CRT improves the plasma metabolomic profile of HF patients, indicating harmonization of myocardial energy substrate metabolism. CRT responders may have a favorable metabolomic profile as a potential biomarker for predicting CRT outcome.
Collapse
Affiliation(s)
- Emirhan Nemutlu
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota; Departments of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota; Department of Analytical Chemistry, Faculty of Pharmacy, University of Hacettepe, Ankara, Turkey
| | - Song Zhang
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota; Departments of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Yi-Zhou Xu
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Andre Terzic
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota; Departments of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Li Zhong
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Petras D Dzeja
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota; Departments of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Yong-Mei Cha
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
29
|
Shibayama J, Yuzyuk TN, Cox J, Makaju A, Miller M, Lichter J, Li H, Leavy JD, Franklin S, Zaitsev AV. Metabolic remodeling in moderate synchronous versus dyssynchronous pacing-induced heart failure: integrated metabolomics and proteomics study. PLoS One 2015; 10:e0118974. [PMID: 25790351 PMCID: PMC4366225 DOI: 10.1371/journal.pone.0118974] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 01/08/2015] [Indexed: 01/08/2023] Open
Abstract
Heart failure (HF) is accompanied by complex alterations in myocardial energy metabolism. Up to 40% of HF patients have dyssynchronous ventricular contraction, which is an independent indicator of mortality. We hypothesized that electromechanical dyssynchrony significantly affects metabolic remodeling in the course of HF. We used a canine model of tachypacing-induced HF. Animals were paced at 200 bpm for 6 weeks either in the right atrium (synchronous HF, SHF) or in the right ventricle (dyssynchronous HF, DHF). We collected biopsies from left ventricular apex and performed comprehensive metabolic pathway analysis using multi-platform metabolomics (GC/MS; MS/MS; HPLC) and LC-MS/MS label-free proteomics. We found important differences in metabolic remodeling between SHF and DHF. As compared to Control, ATP, phosphocreatine (PCr), creatine, and PCr/ATP (prognostic indicator of mortality in HF patients) were all significantly reduced in DHF, but not SHF. In addition, the myocardial levels of carnitine (mitochondrial fatty acid carrier) and fatty acids (12:0, 14:0) were significantly reduced in DHF, but not SHF. Carnitine parmitoyltransferase I, a key regulatory enzyme of fatty acid ß-oxidation, was significantly upregulated in SHF but was not different in DHF, as compared to Control. Both SHF and DHF exhibited a reduction, but to a different degree, in creatine and the intermediates of glycolysis and the TCA cycle. In contrast to this, the enzymes of creatine kinase shuttle were upregulated, and the enzymes of glycolysis and the TCA cycle were predominantly upregulated or unchanged in both SHF and DHF. These data suggest a systemic mismatch between substrate supply and demand in pacing-induced HF. The energy deficit observed in DHF, but not in SHF, may be associated with a critical decrease in fatty acid delivery to the ß-oxidation pipeline, primarily due to a reduction in myocardial carnitine content.
Collapse
Affiliation(s)
- Junko Shibayama
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Tatiana N. Yuzyuk
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- ARUP Laboratories, Salt Lake City, Utah, United States of America
| | - James Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Aman Makaju
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Mickey Miller
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Justin Lichter
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Hui Li
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Jane D. Leavy
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Sarah Franklin
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Alexey V. Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
30
|
Taking a peek at the border of the sarcomere in heart failure and cardiac resynchronization therapy. J Mol Cell Cardiol 2014; 74:1-3. [PMID: 24792363 DOI: 10.1016/j.yjmcc.2014.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 11/20/2022]
|