1
|
Romanelli G, Villarreal L, Espasandín C, Benech JC. Diabetes induces modifications in costameric proteins and increases cardiomyocyte stiffness. Am J Physiol Cell Physiol 2024; 327:C1263-C1273. [PMID: 39374079 DOI: 10.1152/ajpcell.00273.2024] [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: 04/26/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 10/09/2024]
Abstract
Several studies have demonstrated that diabetes mellitus can increase the risk of cardiovascular disease and remains the principal cause of death in these patients. Costameres connect the sarcolemma with the cytoskeleton and extracellular matrix, facilitating the transmission of mechanical forces and cell signaling. They are related to cardiac physiology because individual cardiac cells are connected by intercalated discs that synchronize muscle contraction. Diabetes impacts the nanomechanical properties of cardiomyocytes, resulting in increased cellular and left ventricular stiffness, as evidenced in clinical studies of these patients. The question of whether costameric proteins are affected by diabetes in the heart has not been studied. This work analyzes whether type 1 diabetes mellitus (T1DM) modifies the costameric proteins and coincidentally changes the cellular mechanics in the same cardiomyocytes. The samples were analyzed by immunotechniques using laser confocal microscopy. Significant statistical differences were found in the spatial arrangement of the costameric proteins. However, these differences are not due to their expression. Atomic force microscopy was used to compare intrinsic cellular stiffness between diabetic and normal cardiomyocytes and obtain the first elasticity map sections of diabetic living cardiomyocytes. Data obtained demonstrated that diabetic cardiomyocytes had higher stiffness than control. The present work shows experimental evidence that intracellular changes related to cell-cell and cell-extracellular matrix communication occur, which could be related to cardiac pathogenic mechanisms. These changes could contribute to alterations in the mechanical and electrical properties of cardiomyocytes and, consequently, to diabetic cardiomyopathy.NEW & NOTEWORTHY The structural organization of cardiomyocyte proteins is critical for their efficient functioning as a contractile unit in the heart. This work shows that diabetes mellitus induces significant changes in the spatial organization of costamere proteins, t tubules, and intercalated discs. We obtained the first elasticity map sections of living diabetic cardiomyocytes. The results show statistical differences in the map sections of diabetic and control cardiomyocytes, with diabetic cardiomyocytes being stiffer than normal ones.
Collapse
Affiliation(s)
- Gerardo Romanelli
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Lihuén Villarreal
- Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Camila Espasandín
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Montevideo, Uruguay
| | - Juan Claudio Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
2
|
Shavlakadze T, Xiong K, Mishra S, McEwen C, Gadi A, Wakai M, Salmon H, Stec MJ, Negron N, Ni M, Wei Y, Atwal GS, Bai Y, Glass DJ. Age-related gene expression signatures from limb skeletal muscles and the diaphragm in mice and rats reveal common and species-specific changes. Skelet Muscle 2023; 13:11. [PMID: 37438807 DOI: 10.1186/s13395-023-00321-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND As a result of aging, skeletal muscle undergoes atrophy and a decrease in function. This age-related skeletal muscle weakness is known as "sarcopenia". Sarcopenia is part of the frailty observed in humans. In order to discover treatments for sarcopenia, it is necessary to determine appropriate preclinical models and the genes and signaling pathways that change with age in these models. METHODS AND RESULTS To understand the changes in gene expression that occur as a result of aging in skeletal muscles, we generated a multi-time-point gene expression signature throughout the lifespan of mice and rats, as these are the most commonly used species in preclinical research and intervention testing. Gastrocnemius, tibialis anterior, soleus, and diaphragm muscles from male and female C57Bl/6J mice and male Sprague Dawley rats were analyzed at ages 6, 12, 18, 21, 24, and 27 months, plus an additional 9-month group was used for rats. More age-related genes were identified in rat skeletal muscles compared with mice; this was consistent with the finding that rat muscles undergo more robust age-related decline in mass. In both species, pathways associated with innate immunity and inflammation linearly increased with age. Pathways linked with extracellular matrix remodeling were also universally downregulated. Interestingly, late downregulated pathways were exclusively found in the rat limb muscles and these were linked to metabolism and mitochondrial respiration; this was not seen in the mouse. CONCLUSIONS This extensive, side-by-side transcriptomic profiling shows that the skeletal muscle in rats is impacted more by aging compared with mice, and the pattern of decline in the rat may be more representative of the human. The observed changes point to potential therapeutic interventions to avoid age-related decline in skeletal muscle function.
Collapse
Affiliation(s)
- Tea Shavlakadze
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Kun Xiong
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Shawn Mishra
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Corissa McEwen
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Abhilash Gadi
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Matthew Wakai
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Hunter Salmon
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Michael J Stec
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Nicole Negron
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Min Ni
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Yi Wei
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Gurinder S Atwal
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Yu Bai
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - David J Glass
- Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA.
| |
Collapse
|
3
|
Ahmed RE, Tokuyama T, Anzai T, Chanthra N, Uosaki H. Sarcomere maturation: function acquisition, molecular mechanism, and interplay with other organelles. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210325. [PMID: 36189811 PMCID: PMC9527934 DOI: 10.1098/rstb.2021.0325] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/15/2022] [Indexed: 12/31/2022] Open
Abstract
During postnatal cardiac development, cardiomyocytes mature and turn into adult ones. Hence, all cellular properties, including morphology, structure, physiology and metabolism, are changed. One of the most important aspects is the contractile apparatus, of which the minimum unit is known as a sarcomere. Sarcomere maturation is evident by enhanced sarcomere alignment, ultrastructural organization and myofibrillar isoform switching. Any maturation process failure may result in cardiomyopathy. Sarcomere function is intricately related to other organelles, and the growing evidence suggests reciprocal regulation of sarcomere and mitochondria on their maturation. Herein, we summarize the molecular mechanism that regulates sarcomere maturation and the interplay between sarcomere and other organelles in cardiomyocyte maturation. 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)
- Razan E. Ahmed
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Takeshi Tokuyama
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Tatsuya Anzai
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Nawin Chanthra
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
4
|
Shi H, Wang C, Gao BZ, Henderson JH, Ma Z. Cooperation between myofibril growth and costamere maturation in human cardiomyocytes. Front Bioeng Biotechnol 2022; 10:1049523. [PMID: 36394013 PMCID: PMC9663467 DOI: 10.3389/fbioe.2022.1049523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022] Open
Abstract
Costameres, as striated muscle-specific cell adhesions, anchor both M-lines and Z-lines of the sarcomeres to the extracellular matrix. Previous studies have demonstrated that costameres intimately participate in the initial assembly of myofibrils. However, how costamere maturation cooperates with myofibril growth is still underexplored. In this work, we analyzed zyxin (costameres), α-actinin (Z-lines) and myomesin (M-lines) to track the behaviors of costameres and myofibrils within the cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs). We quantified the assembly and maturation of costameres associated with the process of myofibril growth within the hiPSC-CMs in a time-dependent manner. We found that asynchrony existed not only between the maturation of myofibrils and costameres, but also between the formation of Z-costameres and M-costameres that associated with different structural components of the sarcomeres. This study helps us gain more understanding of how costameres assemble and incorporate into the cardiomyocyte sarcomeres, which sheds a light on cardiomyocyte mechanobiology.
Collapse
Affiliation(s)
- Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States
| | - Bruce Z. Gao
- Department of Bioengineering, Clemson University, Clemson, SC, United States
| | - James H. Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, United States,BioInspired Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, United States,*Correspondence: Zhen Ma,
| |
Collapse
|
5
|
Ajayi PT, Katti P, Zhang Y, Willingham TB, Sun Y, Bleck CKE, Glancy B. Regulation of the evolutionarily conserved muscle myofibrillar matrix by cell type dependent and independent mechanisms. Nat Commun 2022; 13:2661. [PMID: 35562354 PMCID: PMC9106682 DOI: 10.1038/s41467-022-30401-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 04/29/2022] [Indexed: 12/29/2022] Open
Abstract
Skeletal muscles play a central role in human movement through forces transmitted by contraction of the sarcomere. We recently showed that mammalian sarcomeres are connected through frequent branches forming a singular, mesh-like myofibrillar matrix. However, the extent to which myofibrillar connectivity is evolutionarily conserved as well as mechanisms which regulate the specific architecture of sarcomere branching remain unclear. Here, we demonstrate the presence of a myofibrillar matrix in the tubular, but not indirect flight (IF) muscles within Drosophila melanogaster. Moreover, we find that loss of transcription factor H15 increases sarcomere branching frequency in the tubular jump muscles, and we show that sarcomere branching can be turned on in IF muscles by salm-mediated conversion to tubular muscles. Finally, we demonstrate that neurochondrin misexpression results in myofibrillar connectivity in IF muscles without conversion to tubular muscles. These data indicate an evolutionarily conserved myofibrillar matrix regulated by both cell-type dependent and independent mechanisms.
Collapse
Affiliation(s)
- Peter T Ajayi
- Muscle Energetics Laboratory, NHLBI, NIH, Bethesda, MD, 20892, USA
| | - Prasanna Katti
- Muscle Energetics Laboratory, NHLBI, NIH, Bethesda, MD, 20892, USA
| | - Yingfan Zhang
- Muscle Energetics Laboratory, NHLBI, NIH, Bethesda, MD, 20892, USA
| | | | - Ye Sun
- Electron Microscopy Core, NHLBI, NIH, Bethesda, MD, 20892, USA
| | | | - Brian Glancy
- Muscle Energetics Laboratory, NHLBI, NIH, Bethesda, MD, 20892, USA.
- NIAMS, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
6
|
Ma W, Gong H, Jani V, Lee KH, Landim-Vieira M, Papadaki M, Pinto JR, Aslam MI, Cammarato A, Irving T. Myofibril orientation as a metric for characterizing heart disease. Biophys J 2022; 121:565-574. [PMID: 35032456 PMCID: PMC8874025 DOI: 10.1016/j.bpj.2022.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
Myocyte disarray is a hallmark of many cardiac disorders. However, the relationship between alterations in the orientation of individual myofibrils and myofilaments to disease progression has been largely underexplored. This oversight has predominantly been because of a paucity of methods for objective and quantitative analysis. Here, we introduce a novel, less-biased approach to quantify myofibrillar and myofilament orientation in cardiac muscle under near-physiological conditions and demonstrate its superiority as compared with conventional histological assessments. Using small-angle x-ray diffraction, we first investigated changes in myofibrillar orientation at increasing sarcomere lengths in permeabilized, relaxed, wild-type mouse myocardium from the left ventricle by assessing the angular spread of the 1,0 equatorial reflection (angle σ). At a sarcomere length of 1.9 μm, the angle σ was 0.23 ± 0.01 rad, decreased to 0.19 ± 0.01 rad at a sarcomere length of 2.1 μm, and further decreased to 0.15 ± 0.01 rad at a sarcomere length of 2.3 μm (p < 0.0001). Angle σ was significantly larger in R403Q, a MYH7 hypertrophic cardiomyopathy model, porcine myocardium (0.24 ± 0.01 rad) compared with wild-type myocardium (0.14 ± 0.005 rad; p < 0.0001), as well as in human heart failure tissue (0.19 ± 0.006 rad) when compared with nonfailing samples (0.17 ± 0.007 rad; p = 0.01). These data indicate that diseased myocardium suffers from greater myofibrillar disorientation compared with healthy controls. Finally, we showed that conventional, histology-based analysis of disarray can be subject to user bias and/or sampling error and lead to false positives. Our method for directly assessing myofibrillar orientation avoids the artifacts introduced by conventional histological approaches that assess myocyte orientation and only indirectly evaluate myofibrillar orientation, and provides a precise and objective metric for phenotypically characterizing myocardium. The ability to obtain excellent x-ray diffraction patterns from frozen human myocardium provides a new tool for investigating structural anomalies associated with cardiac diseases.
Collapse
Affiliation(s)
- Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, Illinois.
| | - Henry Gong
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, Illinois
| | - Vivek Jani
- Department of Biomedical Engineering, The Johns Hopkins School of Medicine, The Johns Hopkins University, Baltimore, Maryland; Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kyoung Hwan Lee
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - Maria Papadaki
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois
| | - Jose R Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, Florida
| | - M Imran Aslam
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anthony Cammarato
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Thomas Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, Illinois
| |
Collapse
|
7
|
Singh JP, Young JL. The cardiac nanoenvironment: form and function at the nanoscale. Biophys Rev 2021; 13:625-636. [PMID: 34765045 PMCID: PMC8555021 DOI: 10.1007/s12551-021-00834-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Mechanical forces in the cardiovascular system occur over a wide range of length scales. At the whole organ level, large scale forces drive the beating heart as a synergistic unit. On the microscale, individual cells and their surrounding extracellular matrix (ECM) exhibit dynamic reciprocity, with mechanical feedback moving bidirectionally. Finally, in the nanometer regime, molecular features of cells and the ECM show remarkable sensitivity to mechanical cues. While small, these nanoscale properties are in many cases directly responsible for the mechanosensitive signaling processes that elicit cellular outcomes. Given the inherent challenges in observing, quantifying, and reconstituting this nanoscale environment, it is not surprising that this landscape has been understudied compared to larger length scales. Here, we aim to shine light upon the cardiac nanoenvironment, which plays a crucial role in maintaining physiological homeostasis while also underlying pathological processes. Thus, we will highlight strategies aimed at (1) elucidating the nanoscale components of the cardiac matrix, and (2) designing new materials and biosystems capable of mimicking these features in vitro.
Collapse
Affiliation(s)
- Jashan P Singh
- Mechanobiology Institute, National University of Singapore, 117411 Singapore, Singapore
| | - Jennifer L Young
- Mechanobiology Institute, National University of Singapore, 117411 Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, 117575 Singapore, Singapore
| |
Collapse
|
8
|
Gómez-Oca R, Cowling BS, Laporte J. Common Pathogenic Mechanisms in Centronuclear and Myotubular Myopathies and Latest Treatment Advances. Int J Mol Sci 2021; 22:11377. [PMID: 34768808 PMCID: PMC8583656 DOI: 10.3390/ijms222111377] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023] Open
Abstract
Centronuclear myopathies (CNM) are rare congenital disorders characterized by muscle weakness and structural defects including fiber hypotrophy and organelle mispositioning. The main CNM forms are caused by mutations in: the MTM1 gene encoding the phosphoinositide phosphatase myotubularin (myotubular myopathy), the DNM2 gene encoding the mechanoenzyme dynamin 2, the BIN1 gene encoding the membrane curvature sensing amphiphysin 2, and the RYR1 gene encoding the skeletal muscle calcium release channel/ryanodine receptor. MTM1, BIN1, and DNM2 proteins are involved in membrane remodeling and trafficking, while RyR1 directly regulates excitation-contraction coupling (ECC). Several CNM animal models have been generated or identified, which confirm shared pathological anomalies in T-tubule remodeling, ECC, organelle mispositioning, protein homeostasis, neuromuscular junction, and muscle regeneration. Dynamin 2 plays a crucial role in CNM physiopathology and has been validated as a common therapeutic target for three CNM forms. Indeed, the promising results in preclinical models set up the basis for ongoing clinical trials. Another two clinical trials to treat myotubular myopathy by MTM1 gene therapy or tamoxifen repurposing are also ongoing. Here, we review the contribution of the different CNM models to understanding physiopathology and therapy development with a focus on the commonly dysregulated pathways and current therapeutic targets.
Collapse
Affiliation(s)
- Raquel Gómez-Oca
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
- Dynacure, 67400 Illkirch, France;
| | | | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
| |
Collapse
|
9
|
Allosteric Modulation of GSK-3β as a New Therapeutic Approach in Limb Girdle Muscular Dystrophy R1 Calpain 3-Related. Int J Mol Sci 2021; 22:ijms22147367. [PMID: 34298987 PMCID: PMC8308041 DOI: 10.3390/ijms22147367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy produced by mutations in the CAPN3 gene. It is a rare disease and there is no cure or treatment for the disease while the pathophysiological mechanism by which the absence of calpain 3 provokes the dystrophy in muscles is not clear. However, key proteins implicated in Wnt and mTOR signaling pathways, which regulate muscle homeostasis, showed a considerable reduction in their expression and in their phosphorylation in LGMDR1 patients' muscles. Finally, the administration of tideglusib and VP0.7, ATP non-competitive inhibitors of glycogen synthase kinase 3β (GSK-3β), restore the expression and phosphorylation of these proteins in LGMDR1 cells, opening the possibility of their use as therapeutic options.
Collapse
|
10
|
Izu L, Shimkunas R, Jian Z, Hegyi B, Kazemi-Lari M, Baker A, Shaw J, Banyasz T, Chen-Izu Y. Emergence of Mechano-Sensitive Contraction Autoregulation in Cardiomyocytes. Life (Basel) 2021; 11:503. [PMID: 34072584 PMCID: PMC8227646 DOI: 10.3390/life11060503] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
The heart has two intrinsic mechanisms to enhance contractile strength that compensate for increased mechanical load to help maintain cardiac output. When vascular resistance increases the ventricular chamber initially expands causing an immediate length-dependent increase of contraction force via the Frank-Starling mechanism. Additionally, the stress-dependent Anrep effect slowly increases contraction force that results in the recovery of the chamber volume towards its initial state. The Anrep effect poses a paradox: how can the cardiomyocyte maintain higher contractility even after the cell length has recovered its initial length? Here we propose a surface mechanosensor model that enables the cardiomyocyte to sense different mechanical stresses at the same mechanical strain. The cell-surface mechanosensor is coupled to a mechano-chemo-transduction feedback mechanism involving three elements: surface mechanosensor strain, intracellular Ca2+ transient, and cell strain. We show that in this simple yet general system, contractility autoregulation naturally emerges, enabling the cardiomyocyte to maintain contraction amplitude despite changes in a range of afterloads. These nontrivial model predictions have been experimentally confirmed. Hence, this model provides a new conceptual framework for understanding the contractility autoregulation in cardiomyocytes, which contributes to the heart's intrinsic adaptivity to mechanical load changes in health and diseases.
Collapse
Affiliation(s)
- Leighton Izu
- Department of Pharmacology, University of California, Davis, CA 95616, USA; (R.S.); (Z.J.); (B.H.); (M.K.-L.); (T.B.); (Y.C.-I.)
| | - Rafael Shimkunas
- Department of Pharmacology, University of California, Davis, CA 95616, USA; (R.S.); (Z.J.); (B.H.); (M.K.-L.); (T.B.); (Y.C.-I.)
| | - Zhong Jian
- Department of Pharmacology, University of California, Davis, CA 95616, USA; (R.S.); (Z.J.); (B.H.); (M.K.-L.); (T.B.); (Y.C.-I.)
| | - Bence Hegyi
- Department of Pharmacology, University of California, Davis, CA 95616, USA; (R.S.); (Z.J.); (B.H.); (M.K.-L.); (T.B.); (Y.C.-I.)
| | - Mohammad Kazemi-Lari
- Department of Pharmacology, University of California, Davis, CA 95616, USA; (R.S.); (Z.J.); (B.H.); (M.K.-L.); (T.B.); (Y.C.-I.)
| | - Anthony Baker
- Department of Medicine, University of California, San Francisco, CA 94121, USA;
| | - John Shaw
- Department of Aerospace Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Tamas Banyasz
- Department of Pharmacology, University of California, Davis, CA 95616, USA; (R.S.); (Z.J.); (B.H.); (M.K.-L.); (T.B.); (Y.C.-I.)
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary
| | - Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA 95616, USA; (R.S.); (Z.J.); (B.H.); (M.K.-L.); (T.B.); (Y.C.-I.)
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
- Department of Internal Medicine, Division of Cardiology, University of California, Davis, CA 95616, USA
| |
Collapse
|
11
|
Imanaka-Yoshida K. Tenascin-C in Heart Diseases-The Role of Inflammation. Int J Mol Sci 2021; 22:ijms22115828. [PMID: 34072423 PMCID: PMC8198581 DOI: 10.3390/ijms22115828] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/25/2021] [Accepted: 05/27/2021] [Indexed: 12/20/2022] Open
Abstract
Tenascin-C (TNC) is a large extracellular matrix (ECM) glycoprotein and an original member of the matricellular protein family. TNC is transiently expressed in the heart during embryonic development, but is rarely detected in normal adults; however, its expression is strongly up-regulated with inflammation. Although neither TNC-knockout nor -overexpressing mice show a distinct phenotype, disease models using genetically engineered mice combined with in vitro experiments have revealed multiple significant roles for TNC in responses to injury and myocardial repair, particularly in the regulation of inflammation. In most cases, TNC appears to deteriorate adverse ventricular remodeling by aggravating inflammation/fibrosis. Furthermore, accumulating clinical evidence has shown that high TNC levels predict adverse ventricular remodeling and a poor prognosis in patients with various heart diseases. Since the importance of inflammation has attracted attention in the pathophysiology of heart diseases, this review will focus on the roles of TNC in various types of inflammatory reactions, such as myocardial infarction, hypertensive fibrosis, myocarditis caused by viral infection or autoimmunity, and dilated cardiomyopathy. The utility of TNC as a biomarker for the stratification of myocardial disease conditions and the selection of appropriate therapies will also be discussed from a clinical viewpoint.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan;
- Mie University Research Center for Matrix Biology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| |
Collapse
|
12
|
Imanaka-Yoshida K, Tawara I, Yoshida T. Tenascin-C in cardiac disease: a sophisticated controller of inflammation, repair, and fibrosis. Am J Physiol Cell Physiol 2020; 319:C781-C796. [PMID: 32845719 DOI: 10.1152/ajpcell.00353.2020] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tenascin-C (TNC) is a large extracellular matrix glycoprotein classified as a matricellular protein that is generally upregulated at high levels during physiological and pathological tissue remodeling and is involved in important biological signaling pathways. In the heart, TNC is transiently expressed at several important steps during embryonic development and is sparsely detected in normal adult heart but is re-expressed in a spatiotemporally restricted manner under pathological conditions associated with inflammation, such as myocardial infarction, hypertensive cardiac fibrosis, myocarditis, dilated cardiomyopathy, and Kawasaki disease. Despite its characteristic and spatiotemporally restricted expression, TNC knockout mice develop a grossly normal phenotype. However, various disease models using TNC null mice combined with in vitro experiments have revealed many important functions for TNC and multiple molecular cascades that control cellular responses in inflammation, tissue repair, and even myocardial regeneration. TNC has context-dependent diverse functions and, thus, may exert both harmful and beneficial effects in damaged hearts. However, TNC appears to deteriorate adverse ventricular remodeling by proinflammatory and profibrotic effects in most cases. Its specific expression also makes TNC a feasible diagnostic biomarker and target for molecular imaging to assess inflammation in the heart. Several preclinical studies have shown the utility of TNC as a biomarker for assessing the prognosis of patients and selecting appropriate therapy, particularly for inflammatory heart diseases.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| | - Toshimichi Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Mie University Research Center for Matrix Biology, Tsu, Japan
| |
Collapse
|
13
|
Unconventional roles for membrane traffic proteins in response to muscle membrane stress. Curr Opin Cell Biol 2020; 65:42-49. [DOI: 10.1016/j.ceb.2020.02.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/10/2020] [Accepted: 02/15/2020] [Indexed: 12/19/2022]
|
14
|
Pamuk U, Cankaya AO, Yucesoy CA. Principles of the Mechanism for Epimuscular Myofascial Loads Leading to Non-uniform Strain Distributions Along Muscle Fiber Direction: Finite Element Modeling. Front Physiol 2020; 11:789. [PMID: 32714211 PMCID: PMC7351515 DOI: 10.3389/fphys.2020.00789] [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: 01/31/2020] [Accepted: 06/15/2020] [Indexed: 01/13/2023] Open
Abstract
Sarcomere lengths and their changes are key determinants of muscle active force production. Recent studies indicate inhomogeneity of sarcomere lengths within the muscle. Studies utilizing magnetic resonance imaging (MRI) analyses for quantifying local muscle tissue strains and diffusion tensor imaging (DTI) analyses allowing for determination of their components along muscle fascicles show that those length changes can be non-uniform. Specifically, two questions arise regarding the muscle’s length change heterogeneities along the muscle fiber direction: (1) How can a passively lengthened muscle show shortened regions? (2) How can an isometric contracting muscle show lengthened parts? Using finite element modeling and studying principles of the mechanism of strain heterogeneity along the muscle fiber direction, the aim was to test the following hypothesis: epimuscular myofascial loads can lead locally to strains opposing those elsewhere within the muscle that are determined by the globally imposed conditions. The geometry of the model was defined by the contour of a longitudinal slice of the rat extensor digitorum longus (EDL) muscle belly. Three models were studied: (1) isolated muscle (muscle modeled fully isolated from its surroundings) and models aiming at representing the principles of a muscle in its in vivo context including (2) extramuscularly connected muscle (muscle’s connections to non-muscular structures are modeled exclusively) and (3) epimuscularly connected muscle (additionally muscle’s connections to neighboring muscle are modeled). Three cases were studied: passive isometric muscle with imposed relative position change (Case I), passive lengthened muscle (Case II), and active isometric muscle with imposed relative position change (Case III). The findings indicated non-uniform strains for all models except for zero strain in model (1) in Case I, but models (2) and (3) also showed strains opposing the imposed effect. Case I: model (3) showed shortened and lengthened sections (up to 35.3%), caused exclusively by imposed relative position change. Case II: models (2) and (3) showed shortened sections (up to 12.7 and 19.5%, respectively) in addition to lengthened sections. Case III: models (2) and (3) showed lengthened sections (up to 5 and 23.4%, respectively) in addition to shortened sections. These effects get more pronounced with stiffer epimuscular connections. Assessments of forces exerted on the muscle by the epimuscular connections showed that such strain heterogeneities are ascribed to epimuscular myofascial loads determined by muscle relative position changes.
Collapse
Affiliation(s)
- Uluç Pamuk
- Biomechanica Laboratory, Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Alican Onur Cankaya
- Biomechanica Laboratory, Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Can A Yucesoy
- Biomechanica Laboratory, Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey
| |
Collapse
|
15
|
Casas-Fraile L, Cornelis FM, Costamagna D, Rico A, Duelen R, Sampaolesi MM, López de Munain A, Lories RJ, Sáenz A. Frizzled related protein deficiency impairs muscle strength, gait and calpain 3 levels. Orphanet J Rare Dis 2020; 15:119. [PMID: 32448375 PMCID: PMC7245871 DOI: 10.1186/s13023-020-01372-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/31/2020] [Indexed: 01/24/2023] Open
Abstract
Background Limb-girdle muscular dystrophy recessive 1 calpain3-related (LGMDR1), previously known as LGMD2A, is a disease caused by mutations in the CAPN3 gene. It is characterized by progressive weakness and muscle degeneration. Frizzled related protein (FRZB), upregulated in LGMDR1, was identified as a key regulator of the crosstalk between Wnt and integrin signalling pathways. FRZB gene silencing showed a recovery in the expression of some of the costamere protein levels in myotubes. Results Here, we performed a comprehensive characterization of Frzb−/− mice muscles to study the absence of Frzb in skeletal muscle and eventual links with the molecular characteristics of LGMDR1 patient muscles. Frzb−/− mice showed reduced muscle size and strength. Gait analysis showed that Frzb−/− mice moved more slowly but no impaired regeneration capacity was observed after muscle injury. Additionally, Frzb−/− mice muscle showed an increased number of mesoangioblasts. Lack of Frzb gene in Frzb−/− mice and its increased expression in LGMDR1 patients, showed contrary regulation of Rora, Slc16a1, Tfrc and Capn3 genes. The reciprocal regulation of Frzb and Capn3 genes further supports this axis as a potential target for LGMDR1 patients. Conclusions Our data confirm a role for Frzb in the regulation of Rora, Slc16a1, Tfrc, and Capn3 genes in muscle cells. In vivo, reduced muscle strength and gait in the Frzb−/− mice are intriguing features. The reciprocal relationship between FRZB and CAPN3 further supports a key role for this axis in patients with LGMDR1.
Collapse
Affiliation(s)
- Leire Casas-Fraile
- Biodonostia Health Research Institute, Neurosciences Area, San Sebastian, Spain.,Spanish Ministry of Economy & Competitiveness, Carlos III Health Institute, CIBER, Madrid, Spain.,Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Frederique M Cornelis
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium
| | - Domiziana Costamagna
- Department of Development and Regeneration, Stem Cell Institute, Laboratory of Translational Cardiomyology, KU Leuven, Leuven, Belgium
| | - Anabel Rico
- Biodonostia Health Research Institute, Neurosciences Area, San Sebastian, Spain
| | - Robin Duelen
- Department of Development and Regeneration, Stem Cell Institute, Laboratory of Translational Cardiomyology, KU Leuven, Leuven, Belgium
| | - Maurilio M Sampaolesi
- Department of Development and Regeneration, Stem Cell Institute, Laboratory of Translational Cardiomyology, KU Leuven, Leuven, Belgium.,Department of Public Health, Experimental and Forensic Medicine, Human Anatomy Unit, University of Pavia, Pavia, Italy
| | - Adolfo López de Munain
- Biodonostia Health Research Institute, Neurosciences Area, San Sebastian, Spain.,Spanish Ministry of Economy & Competitiveness, Carlos III Health Institute, CIBER, Madrid, Spain.,Department of Neurology, Donostia University Hospital, Donostia, Spain.,Department of Neurosciences, University of the Basque Country, Leioa, Spain
| | - Rik J Lories
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, Laboratory of Tissue Homeostasis and Disease, KU Leuven, Leuven, Belgium.,Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Amets Sáenz
- Biodonostia Health Research Institute, Neurosciences Area, San Sebastian, Spain. .,Spanish Ministry of Economy & Competitiveness, Carlos III Health Institute, CIBER, Madrid, Spain.
| |
Collapse
|
16
|
Sun S, Shi H, Moore S, Wang C, Ash-Shakoor A, Mather PT, Henderson JH, Ma Z. Progressive Myofibril Reorganization of Human Cardiomyocytes on a Dynamic Nanotopographic Substrate. ACS APPLIED MATERIALS & INTERFACES 2020; 12:21450-21462. [PMID: 32326701 DOI: 10.1021/acsami.0c03464] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cardiomyocyte (CM) alignment with striated myofibril organization is developed during early cardiac organogenesis. Previous work has successfully achieved in vitro CM alignment using a variety of biomaterial scaffolds and substrates with static topographic features. However, the cellular processes that occur during the response of CMs to dynamic surface topographic changes, which may provide a model of in vivo developmental progress of CM alignment within embryonic myocardium, remains poorly understood. To gain insights into these cellular processes involved in the response of CMs to dynamic topographic changes, we developed a dynamic topographic substrate that employs a shape memory polymer coated with polyelectrolyte multilayers to produce a flat-to-wrinkle surface transition when triggered by a change in incubation temperature. Using this system, we investigated cellular morphological alignment and intracellular myofibril reorganization in response to the dynamic wrinkle formation. Hence, we identified the progressive cellular processes of human-induced pluripotent stem cell-CMs in a time-dependent manner, which could provide a foundation for a mechanistic model of cardiac myofibril reorganization in response to extracellular microenvironment changes.
Collapse
Affiliation(s)
- Shiyang Sun
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Sarah Moore
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Ariel Ash-Shakoor
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
| | - Patrick T Mather
- Department of Chemical Engineering, Bucknell University, Lewisburg, Pennsylvania 17837, United States
| | - James H Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, New York 13244, United States
- Syracuse Biomaterials Institute, Syracuse University, Syracuse, New York 13244, United States
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
17
|
Lindsay A, Southern WM, McCourt PM, Larson AA, Hodges JS, Lowe DA, Ervasti JM. Variable cytoplasmic actin expression impacts the sensitivity of different dystrophin-deficient mdx skeletal muscles to eccentric contraction. FEBS J 2019; 286:2562-2576. [PMID: 30942954 PMCID: PMC6613979 DOI: 10.1111/febs.14831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/24/2019] [Accepted: 04/01/2019] [Indexed: 11/29/2022]
Abstract
Eccentric contractions (ECCs) induce force loss in several skeletal muscles of dystrophin-deficient mice (mdx), with the exception of the soleus (Sol). The eccentric force : isometric force (ECC : ISO), expression level of utrophin, fiber type distribution, and sarcoendoplasmic reticulum calcium ATPase expression are factors that differ between muscles and may contribute to the sensitivity of mdx skeletal muscle to ECC. Here, we confirm that the Sol of mdx mice loses only 13% force compared to 87% in the extensor digitorum longus (EDL) following 10 ECC of isolated muscles. The Sol has a greater proportion of fibers expressing Type I myosin heavy chain (MHC) and expresses 2.3-fold more utrophin compared to the EDL. To examine the effect of ECC : ISO, we show that the mdx Sol is insensitive to ECC at ECC : ISO up to 230 ± 15%. We show that the peroneus longus (PL) muscle presents with similar ECC : ISO compared to the EDL, intermediate force loss (68%) following 10 ECC, and intermediate fiber type distribution and utrophin expression relative to EDL and Sol. The combined absence of utrophin and dystrophin in mdx/utrophin-/- mice rendered the Sol only partially susceptible to ECC and exacerbated force loss in the EDL and PL. Most interestingly, the expression levels of cytoplasmic β- and γ-actins correlate inversely with a given muscle's sensitivity to ECC; EDL < PL < Sol. Our data indicate that fiber type, utrophin, and cytoplasmic actin expression all contribute to the differential sensitivities of mdxEDL, PL, and Sol muscles to ECC.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - William M. Southern
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - Preston M. McCourt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - Alexie A. Larson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, USA
| | - James S. Hodges
- Division of Biostatistics, University of Minnesota, Minneapolis, USA
| | - Dawn A. Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, USA
| | - James M. Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| |
Collapse
|
18
|
Watson SA, Duff J, Bardi I, Zabielska M, Atanur SS, Jabbour RJ, Simon A, Tomas A, Smolenski RT, Harding SE, Perbellini F, Terracciano CM. Biomimetic electromechanical stimulation to maintain adult myocardial slices in vitro. Nat Commun 2019; 10:2168. [PMID: 31092830 PMCID: PMC6520377 DOI: 10.1038/s41467-019-10175-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 04/24/2019] [Indexed: 01/01/2023] Open
Abstract
Adult cardiac tissue undergoes a rapid process of dedifferentiation when cultured outside the body. The in vivo environment, particularly constant electromechanical stimulation, is fundamental to the regulation of cardiac structure and function. We investigated the role of electromechanical stimulation in preventing culture-induced dedifferentiation of adult cardiac tissue using rat, rabbit and human heart failure myocardial slices. Here we report that the application of a preload equivalent to sarcomere length (SL) = 2.2 μm is optimal for the maintenance of rat myocardial slice structural, functional and transcriptional properties at 24 h. Gene sets associated with the preservation of structure and function are activated, while gene sets involved in dedifferentiation are suppressed. The maximum contractility of human heart failure myocardial slices at 24 h is also optimally maintained at SL = 2.2 μm. Rabbit myocardial slices cultured at SL = 2.2 μm remain stable for 5 days. This approach substantially prolongs the culture of adult cardiac tissue in vitro.
Collapse
Affiliation(s)
- Samuel A Watson
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - James Duff
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Ifigeneia Bardi
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Magdalena Zabielska
- Department of Biochemistry, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Santosh S Atanur
- Faculty of Medicine, Department of Medicine, Imperial College London, London, SW7 2AZ, UK
| | - Richard J Jabbour
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - André Simon
- Department of Cardiothoracic Transplantation & Mechanical Circulatory Support, Royal Brompton & Harefield NHS Foundation Trust, Harefield, UB9 6JH, UK
| | - Alejandra Tomas
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London, London, W12 0NN, UK
| | - Ryszard T Smolenski
- Department of Biochemistry, Medical University of Gdańsk, Gdańsk, 80-210, Poland
| | - Sian E Harding
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Filippo Perbellini
- National Heart & Lung Institute, Imperial College London, London, W12 0NN, UK.
| | | |
Collapse
|
19
|
Solís C, Russell B. CapZ integrates several signaling pathways in response to mechanical stiffness. J Gen Physiol 2019; 151:660-669. [PMID: 30808692 PMCID: PMC6504289 DOI: 10.1085/jgp.201812199] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/13/2018] [Accepted: 02/06/2019] [Indexed: 12/22/2022] Open
Abstract
Changes in mechanical load, hormones, or metabolic stress provoke remodeling of the actin-based thin filaments within muscle fibers. Solís and Russell show that several signaling pathways converge at the actin-capping protein CapZ to regulate muscle fiber growth in response to mechanical stiffness and neurohumoral signaling. Muscle adaptation is a response to physiological demand elicited by changes in mechanical load, hormones, or metabolic stress. Cytoskeletal remodeling processes in many cell types are thought to be primarily regulated by thin filament formation due to actin-binding accessory proteins, such as the actin-capping protein. Here, we hypothesize that in muscle, the actin-capping protein (named CapZ) integrates signaling by a variety of pathways, including phosphorylation and phosphatidylinositol 4,5-bisphosphate (PIP2) binding, to regulate muscle fiber growth in response to mechanical load. To test this hypothesis, we assess mechanotransduction signaling that regulates muscle growth using neonatal rat ventricular myocytes cultured on substrates with the stiffness of the healthy myocardium (10 kPa), fibrotic myocardium (100 kPa), or glass. We investigate how PIP2 signaling affects CapZ using the PIP2 sequestering agent neomycin and the effect of PKC-mediated CapZ phosphorylation using the PKC-activating drug phorbol 12-myristate 13-acetate (PMA). Molecular simulations suggest that close interactions between PIP2 and the β-tentacle of CapZ are modified by phosphorylation at T267. Fluorescence recovery after photobleaching (FRAP) demonstrates that the kinetic binding constant of CapZ to sarcomeric thin filaments in living muscle cells increases with stiffness or PMA treatment but is diminished by PIP2 reduction. Furthermore, CapZ with a deletion of the β-tentacle that lacks the phosphorylation site T267 shows increased FRAP kinetics with lack of sensitivity to PMA treatment or PIP2 reduction. Förster resonance energy transfer (FRET) probes the molecular interactions between PIP2 and CapZ, which are decreased by PIP2 availability or by the β-tentacle truncation. These data suggest that CapZ is bound to actin tightly in the idle, locked state, with little phosphorylation or PIP2 binding. However, this tight binding is loosened in growth states triggered by mechanical stimuli such as substrate stiffness, which may have relevance to fibrotic heart disease.
Collapse
Affiliation(s)
- Christopher Solís
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL
| | - Brenda Russell
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL
| |
Collapse
|
20
|
Babačić H, Mehta A, Merkel O, Schoser B. CRISPR-cas gene-editing as plausible treatment of neuromuscular and nucleotide-repeat-expansion diseases: A systematic review. PLoS One 2019; 14:e0212198. [PMID: 30794581 PMCID: PMC6386526 DOI: 10.1371/journal.pone.0212198] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/29/2019] [Indexed: 12/26/2022] Open
Abstract
INTRODUCTION The system of clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated proteins (cas) is a new technology that allows easier manipulation of the genome. Its potential to edit genes opened a new door in treatment development for incurable neurological monogenic diseases (NMGDs). The aim of this systematic review was to summarise the findings on the current development of CRISPR-cas for therapeutic purposes in the most frequent NMGDs and provide critical assessment. METHODS AND DATA ACQUISITION We searched the MEDLINE and EMBASE databases, looking for original studies on the use of CRISPR-cas to edit pathogenic variants in models of the most frequent NMGDs, until end of 2017. We included all the studies that met the following criteria: 1. Peer-reviewed study report with explicitly described experimental designs; 2. In vitro, ex vivo, or in vivo study using human or other animal biological systems (including cells, tissues, organs, organisms); 3. focusing on CRISPR as the gene-editing method of choice; and 5. featured at least one NMGD. RESULTS We obtained 404 papers from MEDLINE and 513 from EMBASE. After removing the duplicates, we screened 490 papers by title and abstract and assessed them for eligibility. After reading 50 full-text papers, we finally selected 42 for the review. DISCUSSION Here we give a systematic summary on the preclinical development of CRISPR-cas for therapeutic purposes in NMGDs. Furthermore, we address the clinical interpretability of the findings, giving a comprehensive overview of the current state of the art. Duchenne's muscular dystrophy (DMD) paves the way forward, with 26 out of 42 studies reporting different strategies on DMD gene editing in different models of the disease. Most of the strategies aimed for permanent exon skipping by deletion with CRISPR-cas. Successful silencing of the mHTT gene with CRISPR-cas led to successful reversal of the neurotoxic effects in the striatum of mouse models of Huntington's disease. Many other strategies have been explored, including epigenetic regulation of gene expression, in cellular and animal models of: myotonic dystrophy, Fraxile X syndrome, ataxias, and other less frequent dystrophies. Still, before even considering the clinical application of CRISPR-cas, three major bottlenecks need to be addressed: efficacy, safety, and delivery of the systems. This requires a collaborative approach in the research community, while having ethical considerations in mind.
Collapse
Affiliation(s)
- Haris Babačić
- Friedrich Baur Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
- * E-mail: (BS); (HB)
| | - Aditi Mehta
- Faculty of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Olivia Merkel
- Faculty of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Benedikt Schoser
- Friedrich Baur Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
- * E-mail: (BS); (HB)
| |
Collapse
|
21
|
Franck A, Lainé J, Moulay G, Lemerle E, Trichet M, Gentil C, Benkhelifa-Ziyyat S, Lacène E, Bui MT, Brochier G, Guicheney P, Romero N, Bitoun M, Vassilopoulos S. Clathrin plaques and associated actin anchor intermediate filaments in skeletal muscle. Mol Biol Cell 2019; 30:579-590. [PMID: 30601711 PMCID: PMC6589689 DOI: 10.1091/mbc.e18-11-0718] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Clathrin plaques are stable features of the plasma membrane observed in several cell types. They are abundant in muscle, where they localize at costameres that link the contractile apparatus to the sarcolemma and connect the sarcolemma to the basal lamina. Here, we show that clathrin plaques and surrounding branched actin filaments form microdomains that anchor a three-dimensional desmin intermediate filament (IF) web. Depletion of clathrin plaque and branched actin components causes accumulation of desmin tangles in the cytoplasm. We show that dynamin 2, whose mutations cause centronuclear myopathy (CNM), regulates both clathrin plaques and surrounding branched actin filaments, while CNM-causing mutations lead to desmin disorganization in a CNM mouse model and patient biopsies. Our results suggest a novel paradigm in cell biology, wherein clathrin plaques act as platforms capable of recruiting branched cortical actin, which in turn anchors IFs, both essential for striated muscle formation and function.
Collapse
Affiliation(s)
- Agathe Franck
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Jeanne Lainé
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France.,Department of Physiology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Gilles Moulay
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Eline Lemerle
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Michaël Trichet
- Institut de Biologie Paris-Seine, Sorbonne Université, CNRS, FR3631 Paris, France
| | - Christel Gentil
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Sofia Benkhelifa-Ziyyat
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Emmanuelle Lacène
- Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Mai Thao Bui
- Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Guy Brochier
- Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Pascale Guicheney
- Institute of Cardiometabolism and Nutrition, UMRS 1166, INSERM , Sorbonne Université, F-75013 Paris, France
| | - Norma Romero
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France.,Neuromuscular Morphology Unit, Institute of Myology, Pitié-Salpêtrière Hospital, Sorbonne Université, F-75013 Paris, France
| | - Marc Bitoun
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| | - Stéphane Vassilopoulos
- Center of Research in Myology, Institute of Myology, UMRS 974, INSERM, Sorbonne Université, F-75013 Paris, France
| |
Collapse
|
22
|
Abstract
Activation of the electrical signal and its transmission as a depolarizing wave in the whole heart requires highly organized myocyte architecture and cell-cell contacts. In addition, complex trafficking and anchoring intracellular machineries regulate the proper surface expression of channels and their targeting to distinct membrane domains. An increasing list of proteins, lipids, and second messengers can contribute to the normal targeting of ion channels in cardiac myocytes. However, their precise roles in the electrophysiology of the heart are far from been extensively understood. Nowadays, much effort in the field focuses on understanding the mechanisms that regulate ion channel targeting to sarcolemma microdomains and their organization into macromolecular complexes. The purpose of the present section is to provide an overview of the characterized partners of the main cardiac sodium channel, NaV1.5, involved in regulating the functional expression of this channel both in terms of trafficking and targeting into microdomains.
Collapse
|
23
|
Filipova D, Henry M, Rotshteyn T, Brunn A, Carstov M, Deckert M, Hescheler J, Sachinidis A, Pfitzer G, Papadopoulos S. Distinct transcriptomic changes in E14.5 mouse skeletal muscle lacking RYR1 or Cav1.1 converge at E18.5. PLoS One 2018; 13:e0194428. [PMID: 29543863 PMCID: PMC5854361 DOI: 10.1371/journal.pone.0194428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/04/2018] [Indexed: 12/20/2022] Open
Abstract
In skeletal muscle the coordinated actions of two mechanically coupled Ca2+ channels-the 1,4-dihydropyridine receptor (Cav1.1) and the type 1 ryanodine receptor (RYR1)-underlie the molecular mechanism of rapid cytosolic [Ca2+] increase leading to contraction. While both [Ca2+]i and contractile activity have been implicated in the regulation of myogenesis, less is known about potential specific roles of Cav1.1 and RYR1 in skeletal muscle development. In this study, we analyzed the histology and the transcriptomic changes occurring at E14.5 -the end of primary myogenesis and around the onset of intrauterine limb movement, and at E18.5 -the end of secondary myogenesis, in WT, RYR1-/-, and Cav1.1-/- murine limb skeletal muscle. At E14.5 the muscle histology of both mutants exhibited initial alterations, which became much more severe at E18.5. Immunohistological analysis also revealed higher levels of activated caspase-3 in the Cav1.1-/- muscles at E14.5, indicating an increase in apoptosis. With WT littermates as controls, microarray analyses identified 61 and 97 differentially regulated genes (DEGs) at E14.5, and 493 and 1047 DEGs at E18.5, in RYR1-/- and Cav1.1-/- samples, respectively. Gene enrichment analysis detected no overlap in the affected biological processes and pathways in the two mutants at E14.5, whereas at E18.5 there was a significant overlap of DEGs in both mutants, affecting predominantly processes linked to muscle contraction. Moreover, the E18.5 vs. E14.5 comparison revealed multiple genotype-specific DEGs involved in contraction, cell cycle and miRNA-mediated signaling in WT, neuronal and bone development in RYR1-/-, and lipid metabolism in Cav1.1-/- samples. Taken together, our study reveals discrete changes in the global transcriptome occurring in limb skeletal muscle from E14.5 to E18.5 in WT, RYR1-/- and Cav1.1-/- mice. Our results suggest distinct functional roles for RYR1 and Cav1.1 in skeletal primary and secondary myogenesis.
Collapse
Affiliation(s)
- Dilyana Filipova
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | - Margit Henry
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Tamara Rotshteyn
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Anna Brunn
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Mariana Carstov
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Martina Deckert
- Department of Neuropathology, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Gabriele Pfitzer
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| | - Symeon Papadopoulos
- Institute of Vegetative Physiology, Center of Physiology and Pathophysiology, University of Cologne, Cologne, Germany
| |
Collapse
|
24
|
Roy S, Mathew MK. Fluid flow modulates electrical activity in cardiac hERG potassium channels. J Biol Chem 2018; 293:4289-4303. [PMID: 29305421 DOI: 10.1074/jbc.ra117.000432] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/28/2017] [Indexed: 01/01/2023] Open
Abstract
Fluid movement within the heart generates substantial shear forces, but the effect of this mechanical stress on the electrical activity of the human heart has not been examined. The fast component of the delayed rectifier potassium currents responsible for repolarization of the cardiac action potential, Ikr, is encoded by the human ether-a-go-go related gene (hERG) channel. Here, we exposed hERG1a channel-expressing HEK293T cells to laminar shear stress (LSS) and observed that this mechanical stress increased the whole-cell current by 30-40%. LSS shifted the voltage dependence of steady-state activation of the hERG channel to the hyperpolarizing direction, accelerated the time course of activation and recovery from inactivation, slowed down deactivation, and shifted the steady-state inactivation to the positive direction, all of which favored the hERG open state. In contrast, the time course of inactivation was faster, favoring the closed state. Using specific inhibitors of focal adhesion kinase, a regulator of mechano-transduction via the integrin pathway, we also found that the LSS-induced modulation of the whole-cell current depended on the integrin pathway. The hERG1b channel variant, which lacks the Per-Arnt-Sim (PAS) domain, and long QT syndrome-associated variants having point mutations in the PAS domain were unaffected by LSS, suggesting that the PAS domain in hERG1a channel may be involved in sensing mechanical shear stress. We conclude that a mechano-electric feedback pathway modulates hERG channel activity through the integrin pathway, indicating that mechanical forces in the heart influence its electrical activity.
Collapse
Affiliation(s)
- Samrat Roy
- From the National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru 560065.,the Biocon Bristol Myers Squibb Research Center, Bengaluru 560099, and.,the School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar 751024, India
| | - M K Mathew
- From the National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru 560065,
| |
Collapse
|
25
|
Chen-Izu Y, Izu LT. Mechano-chemo-transduction in cardiac myocytes. J Physiol 2017; 595:3949-3958. [PMID: 28098356 DOI: 10.1113/jp273101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 12/15/2016] [Indexed: 12/31/2022] Open
Abstract
The heart has the ability to adjust to changing mechanical loads. The Frank-Starling law and the Anrep effect describe exquisite intrinsic mechanisms the heart has for autoregulating the force of contraction to maintain cardiac output under changes of preload and afterload. Although these mechanisms have been known for more than a century, their cellular and molecular underpinnings are still debated. How does the cardiac myocyte sense changes in preload or afterload? How does the myocyte adjust its response to compensate for such changes? In cardiac myocytes Ca2+ is a crucial regulator of contractile force and in this review we compare and contrast recent studies from different labs that address these two important questions. The 'dimensionality' of the mechanical milieu under which experiments are carried out provide important clues to the location of the mechanosensors and the kinds of mechanical forces they can sense and respond to. As a first approximation, sensors inside the myocyte appear to modulate reactive oxygen species while sensors on the cell surface appear to also modulate nitric oxide signalling; both signalling pathways affect Ca2+ handling. Undoubtedly, further studies will add layers to this simplified picture. Clarifying the intimate links from cellular mechanics to reactive oxygen species and nitric oxide signalling and to Ca2+ handling will deepen our understanding of the Frank-Starling law and the Anrep effect, and also provide a unified view on how arrhythmias may arise in seemingly disparate diseases that have in common altered myocyte mechanics.
Collapse
Affiliation(s)
- Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, CA, 95616, USA.,Department of Biomedical Engineering, University of California, Davis, CA, 95616, USA.,Department of Internal Medicine/Division of Cardiology, University of California, Davis, CA, 95616, USA
| | - Leighton T Izu
- Department of Pharmacology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
26
|
Atmanli A, Domian IJ. Recreating the Cardiac Microenvironment in Pluripotent Stem Cell Models of Human Physiology and Disease. Trends Cell Biol 2016; 27:352-364. [PMID: 28007424 DOI: 10.1016/j.tcb.2016.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/18/2016] [Accepted: 11/28/2016] [Indexed: 12/20/2022]
Abstract
The advent of human pluripotent stem cell (hPSC) biology has opened unprecedented opportunities for the use of tissue engineering to generate human cardiac tissue for in vitro study. Engineering cardiac constructs that recapitulate human development and disease requires faithful recreation of the cardiac niche in vitro. Here we discuss recent progress in translating the in vivo cardiac microenvironment into PSC models of the human heart. We review three key physiologic features required to recreate the cardiac niche and facilitate normal cardiac differentiation and maturation: the biochemical, biophysical, and bioelectrical signaling cues. Finally, we discuss key barriers that must be overcome to fulfill the promise of stem cell biology in preclinical applications and ultimately in clinical practice.
Collapse
Affiliation(s)
- Ayhan Atmanli
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Ibrahim John Domian
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
27
|
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: 2.9] [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
|
28
|
Eftestøl E, Egner IM, Lunde IG, Ellefsen S, Andersen T, Sjåland C, Gundersen K, Bruusgaard JC. Increased hypertrophic response with increased mechanical load in skeletal muscles receiving identical activity patterns. Am J Physiol Cell Physiol 2016; 311:C616-C629. [PMID: 27488660 DOI: 10.1152/ajpcell.00016.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/26/2016] [Indexed: 11/22/2022]
Abstract
It is often assumed that mechanical factors are important for effects of exercise on muscle, but during voluntary training and most experimental conditions the effects could solely be attributed to differences in electrical activity, and direct evidence for a mechanosensory pathway has been scarce. We here show that, in rat muscles stimulated in vivo under deep anesthesia with identical electrical activity patterns, isometric contractions induced twofold more hypertrophy than contractions with 50-60% of the isometric force. The number of myonuclei and the RNA levels of myogenin and myogenic regulatory factor 4 were increased with high load, suggesting that activation of satellite cells is mechano dependent. On the other hand, training induced a major shift in fiber type distribution from type 2b to 2x that was load independent, indicating that the electrical signaling rather than mechanosignaling controls fiber type. RAC-α serine/threonine-protein kinase (Akt) and ribosomal protein S6 kinase β-1 (S6K1) were not significantly differentially activated by load, suggesting that the differences in mechanical factors were not important for activating the Akt/mammalian target of rapamycin/S6K1 pathway. The transmembrane molecule syndecan-4 implied in overload hypertrophy in cardiac muscle was not load dependent, suggesting that mechanosignaling in skeletal muscle is different.
Collapse
Affiliation(s)
- Einar Eftestøl
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ingrid M Egner
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Ida G Lunde
- Department of Genetics, Harvard Medical School, Boston, Massachusetts; Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway; KG Jebsen Cardiac Research Center and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Stian Ellefsen
- Section for Sport Sciences, Lillehammer University College, Lillehammer, Norway; and
| | - Tom Andersen
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | | | - Jo C Bruusgaard
- Department of Biosciences, University of Oslo, Oslo, Norway; Department of Health Sciences, Kristiania University College, Oslo, Norway
| |
Collapse
|
29
|
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
|
30
|
Capulli AK, MacQueen LA, Sheehy SP, Parker KK. Fibrous scaffolds for building hearts and heart parts. Adv Drug Deliv Rev 2016; 96:83-102. [PMID: 26656602 PMCID: PMC4807693 DOI: 10.1016/j.addr.2015.11.020] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/24/2015] [Accepted: 11/26/2015] [Indexed: 12/14/2022]
Abstract
Extracellular matrix (ECM) structure and biochemistry provide cell-instructive cues that promote and regulate tissue growth, function, and repair. From a structural perspective, the ECM is a scaffold that guides the self-assembly of cells into distinct functional tissues. The ECM promotes the interaction between individual cells and between different cell types, and increases the strength and resilience of the tissue in mechanically dynamic environments. From a biochemical perspective, factors regulating cell-ECM adhesion have been described and diverse aspects of cell-ECM interactions in health and disease continue to be clarified. Natural ECMs therefore provide excellent design rules for tissue engineering scaffolds. The design of regenerative three-dimensional (3D) engineered scaffolds is informed by the target ECM structure, chemistry, and mechanics, to encourage cell infiltration and tissue genesis. This can be achieved using nanofibrous scaffolds composed of polymers that simultaneously recapitulate 3D ECM architecture, high-fidelity nanoscale topography, and bio-activity. Their high porosity, structural anisotropy, and bio-activity present unique advantages for engineering 3D anisotropic tissues. Here, we use the heart as a case study and examine the potential of ECM-inspired nanofibrous scaffolds for cardiac tissue engineering. We asked: Do we know enough to build a heart? To answer this question, we tabulated structural and functional properties of myocardial and valvular tissues for use as design criteria, reviewed nanofiber manufacturing platforms and assessed their capabilities to produce scaffolds that meet our design criteria. Our knowledge of the anatomy and physiology of the heart, as well as our ability to create synthetic ECM scaffolds have advanced to the point that valve replacement with nanofibrous scaffolds may be achieved in the short term, while myocardial repair requires further study in vitro and in vivo.
Collapse
Affiliation(s)
- A K Capulli
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - L A MacQueen
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Sean P Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
| | - K K Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
31
|
Hughes DC, Wallace MA, Baar K. Effects of aging, exercise, and disease on force transfer in skeletal muscle. Am J Physiol Endocrinol Metab 2015; 309:E1-E10. [PMID: 25968577 PMCID: PMC4490334 DOI: 10.1152/ajpendo.00095.2015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/08/2015] [Indexed: 11/22/2022]
Abstract
The loss of muscle strength and increased injury rate in aging skeletal muscle has previously been attributed to loss of muscle protein (cross-sectional area) and/or decreased neural activation. However, it is becoming clear that force transfer within and between fibers plays a significant role in this process as well. Force transfer involves a secondary matrix of proteins that align and transmit the force produced by the thick and thin filaments along muscle fibers and out to the extracellular matrix. These specialized networks of cytoskeletal proteins aid in passing force through the muscle and also serve to protect individual fibers from injury. This review discusses the cytoskeleton proteins that have been identified as playing a role in muscle force transmission, both longitudinally and laterally, and where possible highlights how disease, aging, and exercise influence the expression and function of these proteins.
Collapse
Affiliation(s)
- David C Hughes
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | - Marita A Wallace
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| | - Keith Baar
- Department of Neurobiology, Physiology and Behavior, University of California Davis, Davis, California
| |
Collapse
|
32
|
Abstract
Muscle fibres are very specialised cells with a complex structure that requires a high level of organisation of the constituent proteins. For muscle contraction to function properly, there is a need for not only sarcomeres, the contractile structures of the muscle fibre, but also costameres. These are supramolecular structures associated with the sarcolemma that allow muscle adhesion to the extracellular matrix. They are composed of protein complexes that interact and whose functions include maintaining cell structure and signal transduction mediated by their constituent proteins. It is important to improve our understanding of these structures, as mutations in various genes that code for costamere proteins cause many types of muscular dystrophy. In this review, we provide a description of costameres detailing each of their constituent proteins, such as dystrophin, dystrobrevin, syntrophin, sarcoglycans, dystroglycans, vinculin, talin, integrins, desmin, plectin, etc. We describe as well the diseases associated with deficiency thereof, providing a general overview of their importance.
Collapse
|
33
|
Kaushik G, Spenlehauer A, Sessions AO, Trujillo AS, Fuhrmann A, Fu Z, Venkatraman V, Pohl D, Tuler J, Wang M, Lakatta EG, Ocorr K, Bodmer R, Bernstein SI, Van Eyk JE, Cammarato A, Engler AJ. Vinculin network-mediated cytoskeletal remodeling regulates contractile function in the aging heart. Sci Transl Med 2015; 7:292ra99. [PMID: 26084806 PMCID: PMC4507505 DOI: 10.1126/scitranslmed.aaa5843] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The human heart is capable of functioning for decades despite minimal cell turnover or regeneration, suggesting that molecular alterations help sustain heart function with age. However, identification of compensatory remodeling events in the aging heart remains elusive. We present the cardiac proteomes of young and old rhesus monkeys and rats, from which we show that certain age-associated remodeling events within the cardiomyocyte cytoskeleton are highly conserved and beneficial rather than deleterious. Targeted transcriptomic analysis in Drosophila confirmed conservation and implicated vinculin as a unique molecular regulator of cardiac function during aging. Cardiac-restricted vinculin overexpression reinforced the cortical cytoskeleton and enhanced myofilament organization, leading to improved contractility and hemodynamic stress tolerance in healthy and myosin-deficient fly hearts. Moreover, cardiac-specific vinculin overexpression increased median life span by more than 150% in flies. A broad array of potential therapeutic targets and regulators of age-associated modifications, specifically for vinculin, are presented. These findings suggest that the heart has molecular mechanisms to sustain performance and promote longevity, which may be assisted by therapeutic intervention to ameliorate the decline of function in aging patient hearts.
Collapse
Affiliation(s)
- Gaurav Kaushik
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Alice Spenlehauer
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ayla O Sessions
- Biomedical Sciences Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Adriana S Trujillo
- Department of Biology, Heart Institute, and Molecular Biology Institute, San Diego State University, San Diego, CA 92182, USA
| | - Alexander Fuhrmann
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Zongming Fu
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Danielle Pohl
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jeremy Tuler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Edward G Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Karen Ocorr
- Development and Aging Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Rolf Bodmer
- Development and Aging Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037, USA
| | - Sanford I Bernstein
- Department of Biology, Heart Institute, and Molecular Biology Institute, San Diego State University, San Diego, CA 92182, USA
| | - Jennifer E Van Eyk
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA. Advanced Clinical Biosystems Research Institute, Barbra Streisand Women's Heart Center, Cedars-Sinai Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anthony Cammarato
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA. Biomedical Sciences Program, University of California, San Diego, La Jolla, CA 92093, USA. Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
34
|
EGR1 Functions as a Potent Repressor of MEF2 Transcriptional Activity. PLoS One 2015; 10:e0127641. [PMID: 26011708 PMCID: PMC4444265 DOI: 10.1371/journal.pone.0127641] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 04/17/2015] [Indexed: 11/19/2022] Open
Abstract
The myocyte enhancer factor 2 (MEF2) transcription factor requires interactions with co-factors for precise regulation of its target genes. Our lab previously reported that the mammalian MEF2A isoform regulates the cardiomyocyte costamere, a critical muscle-specific focal adhesion complex involved in contractility, through its transcriptional control of genes encoding proteins localized to this cytoskeletal structure. To further dissect the transcriptional mechanisms of costamere gene regulation and identify potential co-regulators of MEF2A, a bioinformatics analysis of transcription factor binding sites was performed using the proximal promoter regions of selected costamere genes. One of these predicted sites belongs to the early growth response (EGR) transcription factor family. The EGR1 isoform has been shown to be involved in a number of pathways in cardiovascular homeostasis and disease, making it an intriguing candidate MEF2 coregulator to further characterize. Here, we demonstrate that EGR1 interacts with MEF2A and is a potent and specific repressor of MEF2 transcriptional activity. Furthermore, we show that costamere gene expression in cardiomyocytes is dependent on EGR1 transcriptional activity. This study identifies a mechanism by which MEF2 activity can be modulated to ensure that costamere gene expression is maintained at levels commensurate with cardiomyocyte contractile activity.
Collapse
|
35
|
Geach TJ, Hirst EMA, Zimmerman LB. Contractile activity is required for Z-disc sarcomere maturation in vivo. Genesis 2015; 53:299-307. [PMID: 25845369 PMCID: PMC4676352 DOI: 10.1002/dvg.22851] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/30/2015] [Accepted: 03/31/2015] [Indexed: 01/16/2023]
Abstract
Sarcomere structure underpins structural integrity, signaling, and force transmission in the muscle. In embryos of the frog Xenopus tropicalis, muscle contraction begins even while sarcomerogenesis is ongoing. To determine whether contractile activity plays a role in sarcomere formation in vivo, chemical tools were used to block acto-myosin contraction in embryos of the frog X. tropicalis, and Z-disc assembly was characterized in the paralyzed dicky ticker mutant. Confocal and ultrastructure analysis of paralyzed embryos showed delayed Z-disc formation and defects in thick filament organization. These results suggest a previously undescribed role for contractility in sarcomere maturation in vivo. genesis 53:299–307, 2015. © 2015 The Authors. Genesis Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Timothy J Geach
- Division of Developmental Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Elizabeth M A Hirst
- Division of Developmental Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| | - Lyle B Zimmerman
- Division of Developmental Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, United Kingdom
| |
Collapse
|
36
|
Lyon RC, Zanella F, Omens JH, Sheikh F. Mechanotransduction in cardiac hypertrophy and failure. Circ Res 2015; 116:1462-1476. [PMID: 25858069 PMCID: PMC4394185 DOI: 10.1161/circresaha.116.304937] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/13/2015] [Indexed: 01/10/2023]
Abstract
Cardiac muscle cells have an intrinsic ability to sense and respond to mechanical load through a process known as mechanotransduction. In the heart, this process involves the conversion of mechanical stimuli into biochemical events that induce changes in myocardial structure and function. Mechanotransduction and its downstream effects function initially as adaptive responses that serve as compensatory mechanisms during adaptation to the initial load. However, under prolonged and abnormal loading conditions, the remodeling processes can become maladaptive, leading to altered physiological function and the development of pathological cardiac hypertrophy and heart failure. Although the mechanisms underlying mechanotransduction are far from being fully elucidated, human and mouse genetic studies have highlighted various cytoskeletal and sarcolemmal structures in cardiac myocytes as the likely candidates for load transducers, based on their link to signaling molecules and architectural components important in disease pathogenesis. In this review, we summarize recent developments that have uncovered specific protein complexes linked to mechanotransduction and mechanotransmission within the sarcomere, the intercalated disc, and at the sarcolemma. The protein structures acting as mechanotransducers are the first step in the process that drives physiological and pathological cardiac hypertrophy and remodeling, as well as the transition to heart failure, and may provide better insights into mechanisms driving mechanotransduction-based diseases.
Collapse
Affiliation(s)
- Robert C. Lyon
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Fabian Zanella
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Jeffrey H. Omens
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Farah Sheikh
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| |
Collapse
|
37
|
Imanaka-Yoshida K, Aoki H. Tenascin-C and mechanotransduction in the development and diseases of cardiovascular system. Front Physiol 2014; 5:283. [PMID: 25120494 PMCID: PMC4114189 DOI: 10.3389/fphys.2014.00283] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 07/10/2014] [Indexed: 12/14/2022] Open
Abstract
Living tissue is composed of cells and extracellular matrix (ECM). In the heart and blood vessels, which are constantly subjected to mechanical stress, ECM molecules form well-developed fibrous frameworks to maintain tissue structure. ECM is also important for biological signaling, which influences various cellular functions in embryonic development, and physiological/pathological responses to extrinsic stimuli. Among ECM molecules, increased attention has been focused on matricellular proteins. Matricellular proteins are a growing group of non-structural ECM proteins highly up-regulated at active tissue remodeling, serving as biological mediators. Tenascin-C (TNC) is a typical matricellular protein, which is highly expressed during embryonic development, wound healing, inflammation, and cancer invasion. The expression is tightly regulated, dependent on the microenvironment, including various growth factors, cytokines, and mechanical stress. In the heart, TNC appears in a spatiotemporal-restricted manner during early stages of development, sparsely detected in normal adults, but transiently re-expressed at restricted sites associated with tissue injury and inflammation. Similarly, in the vascular system, TNC is strongly up-regulated during embryonic development and under pathological conditions with an increase in hemodynamic stress. Despite its intriguing expression pattern, cardiovascular system develops normally in TNC knockout mice. However, deletion of TNC causes acute aortic dissection (AAD) under strong mechanical and humoral stress. Accumulating reports suggest that TNC may modulate the inflammatory response and contribute to elasticity of the tissue, so that it may protect cardiovascular tissue from destructive stress responses. TNC may be a key molecule to control cellular activity during development, adaptation, or pathological tissue remodeling.
Collapse
Affiliation(s)
- Kyoko Imanaka-Yoshida
- Department of Pathology and Matrix Biology, Mie University Graduate School of Medicine Tsu, Japan ; Mie University Research Center for Matrix Biology Tsu, Japan
| | - Hiroki Aoki
- Cardiovascular Research Institute, Kurume University Kurume, Japan
| |
Collapse
|
38
|
Abstract
Dystrophin and utrophin are highly similar proteins that both link cortical actin filaments with a complex of sarcolemmal glycoproteins, yet localize to different subcellular domains within normal muscle cells. In mdx mice and Duchenne muscular dystrophy patients, dystrophin is lacking and utrophin is consequently up-regulated and redistributed to locations normally occupied by dystrophin. Transgenic overexpression of utrophin has been shown to significantly improve aspects of the disease phenotype in the mdx mouse; therefore, utrophin up-regulation is under intense investigation as a potential therapy for Duchenne muscular dystrophy. Here we biochemically compared the previously documented microtubule binding activity of dystrophin with utrophin and analyzed several transgenic mouse models to identify phenotypes of the mdx mouse that remain despite transgenic utrophin overexpression. Our in vitro analyses revealed that dystrophin binds microtubules with high affinity and pauses microtubule polymerization, whereas utrophin has no activity in either assay. We also found that transgenic utrophin overexpression does not correct subsarcolemmal microtubule lattice disorganization, loss of torque production after in vivo eccentric contractions, or physical inactivity after mild exercise. Finally, our data suggest that exercise-induced inactivity correlates with loss of sarcolemmal neuronal NOS localization in mdx muscle, whereas loss of in vivo torque production after eccentric contraction-induced injury is associated with microtubule lattice disorganization.
Collapse
|
39
|
Rosado M, Barber CF, Berciu C, Feldman S, Birren SJ, Nicastro D, Goode BL. Critical roles for multiple formins during cardiac myofibril development and repair. Mol Biol Cell 2014; 25:811-27. [PMID: 24430873 PMCID: PMC3952851 DOI: 10.1091/mbc.e13-08-0443] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 11/13/2013] [Accepted: 01/09/2014] [Indexed: 12/31/2022] Open
Abstract
Cardiac and skeletal muscle function depends on the proper formation of myofibrils, which are tandem arrays of highly organized actomyosin contractile units called sarcomeres. How the architecture of these colossal molecular assemblages is established during development and maintained over the lifetime of an animal is poorly understood. We investigate the potential roles in myofibril formation and repair of formin proteins, which are encoded by 15 different genes in mammals. Using quantitative real-time PCR analysis, we find that 13 formins are differentially expressed in mouse hearts during postnatal development. Seven formins immunolocalize to sarcomeres in diverse patterns, suggesting that they have a variety of functional roles. Using RNA interference silencing, we find that the formins mDia2, DAAM1, FMNL1, and FMNL2 are required nonredundantly for myofibrillogenesis. Knockdown phenotypes include global loss of myofibril organization and defective sarcomeric ultrastructure. Finally, our analysis reveals an unanticipated requirement specifically for FMNL1 and FMNL2 in the repair of damaged myofibrils. Together our data reveal an unexpectedly large number of formins, with diverse localization patterns and nonredundant roles, functioning in myofibril development and maintenance, and provide the first evidence of actin assembly factors being required to repair myofibrils.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Animals
- Animals, Newborn
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Cell Differentiation
- Formins
- Gene Expression Regulation, Developmental
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Microfilament Proteins/antagonists & inhibitors
- Microfilament Proteins/genetics
- Microfilament Proteins/metabolism
- Microtubule-Associated Proteins/antagonists & inhibitors
- Microtubule-Associated Proteins/genetics
- Microtubule-Associated Proteins/metabolism
- Muscle Development/genetics
- Myocardium/cytology
- Myocardium/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/ultrastructure
- NADPH Dehydrogenase/antagonists & inhibitors
- NADPH Dehydrogenase/genetics
- NADPH Dehydrogenase/metabolism
- Primary Cell Culture
- Protein Isoforms/antagonists & inhibitors
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Sarcomeres/metabolism
- Sarcomeres/ultrastructure
- Thiazolidines/pharmacology
- Wound Healing/genetics
- rho GTP-Binding Proteins/antagonists & inhibitors
- rho GTP-Binding Proteins/genetics
- rho GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
| | | | - Cristina Berciu
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Steven Feldman
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Susan J. Birren
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Daniela Nicastro
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| | - Bruce L. Goode
- Biology Department and Rosenstiel Basic Medical Sciences Research Center, Brandeis University, Waltham, MA 02454
| |
Collapse
|
40
|
Zhou J, Shu Y, Lü SH, Li JJ, Sun HY, Tang RY, Duan CM, Wang Y, Lin QX, Mou YC, Li X, Wang CY. The spatiotemporal development of intercalated disk in three-dimensional engineered heart tissues based on collagen/matrigel matrix. PLoS One 2013; 8:e81420. [PMID: 24260578 PMCID: PMC3829928 DOI: 10.1371/journal.pone.0081420] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 10/12/2013] [Indexed: 01/09/2023] Open
Abstract
Intercalated disk (ID), which electromechanically couples cardiomyocytes into a functional syncitium, is closely related to normal morphology and function of engineered heart tissues (EHTs), but the development mode of ID in the three-dimensional (3D) EHTs is still unclear. In this study, we focused on the spatiotemporal development of the ID in the EHTs constructed by mixing neonatal rat cardiomyocytes with collagen/Matrigel, and investigated the effect of 3D microenvironment provided by collagen/Matrigel matrix on the formation of ID. By histological and immmunofluorescent staining, the spatiotemporal distribution of ID-related junctions was detected. Furthermore, the ultra-structures of the ID in different developmental stages were observed under transmission electron microscope. In addition, the expression of the related proteins was quantitatively analyzed. The results indicate that accompanying the re-organization of cardiomyocytes in collagen/Matrigel matrix, the proteins of adherens junctions, desmosomes and gap junctions redistributed from diffused distribution to intercellular regions to form an integrated ID. The adherens junction and desmosome which are related with mechanical connection appeared earlier than gap junction which is essential for electrochemical coupling. These findings suggest that the 3D microenvironment based on collagen/Matrigel matrix could support the ordered assembly of the ID in EHTs and have implications for comprehending the ordered and coordinated development of ID during the functional organization of EHTs.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Yao Shu
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Shuang-Hong Lü
- Laboratory of Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Jun-Jie Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Hong-Yu Sun
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Rong-Yu Tang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Cui-Mi Duan
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Yan Wang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Qiu-Xia Lin
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Yong-Chao Mou
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Xia Li
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
| | - Chang-Yong Wang
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences and Tissue Engineering Research Center, Academy of Military Medical Sciences, Beijing, China
- * E-mail:
| |
Collapse
|
41
|
Transcriptional networks regulating the costamere, sarcomere, and other cytoskeletal structures in striated muscle. Cell Mol Life Sci 2013; 71:1641-56. [PMID: 24218011 DOI: 10.1007/s00018-013-1512-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/27/2013] [Accepted: 10/30/2013] [Indexed: 10/26/2022]
Abstract
Structural abnormalities in striated muscle have been observed in numerous transcription factor gain- and loss-of-function phenotypes in animal and cell culture model systems, indicating that transcription is important in regulating the cytoarchitecture. While most characterized cytoarchitectural defects are largely indistinguishable by histological and ultrastructural criteria, analysis of dysregulated gene expression in each mutant phenotype has yielded valuable information regarding specific structural gene programs that may be uniquely controlled by each of these transcription factors. Linking the formation and maintenance of each subcellular structure or subset of proteins within a cytoskeletal compartment to an overlapping but distinct transcription factor cohort may enable striated muscle to control cytoarchitectural function in an efficient and specific manner. Here we summarize the available evidence that connects transcription factors, those with established roles in striated muscle such as MEF2 and SRF, as well as other non-muscle transcription factors, to the regulation of a defined cytoskeletal structure. The notion that genes encoding proteins localized to the same subcellular compartment are coordinately transcriptionally regulated may prompt rationally designed approaches that target specific transcription factor pathways to correct structural defects in muscle disease.
Collapse
|
42
|
Sequeira V, Nijenkamp LLAM, Regan JA, van der Velden J. The physiological role of cardiac cytoskeleton and its alterations in heart failure. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1838:700-22. [PMID: 23860255 DOI: 10.1016/j.bbamem.2013.07.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 12/11/2022]
Abstract
Cardiac muscle cells are equipped with specialized biochemical machineries for the rapid generation of force and movement central to the work generated by the heart. During each heart beat cardiac muscle cells perceive and experience changes in length and load, which reflect one of the fundamental principles of physiology known as the Frank-Starling law of the heart. Cardiac muscle cells are unique mechanical stretch sensors that allow the heart to increase cardiac output, and adjust it to new physiological and pathological situations. In the present review we discuss the mechano-sensory role of the cytoskeletal proteins with respect to their tight interaction with the sarcolemma and extracellular matrix. The role of contractile thick and thin filament proteins, the elastic protein titin, and their anchorage at the Z-disc and M-band, with associated proteins are reviewed in physiologic and pathologic conditions leading to heart failure. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Hervé
Collapse
Affiliation(s)
- Vasco Sequeira
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Louise L A M Nijenkamp
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | - Jessica A Regan
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; Department of Physiology, Molecular Cardiovascular Research Program, Sarver Heart Center, University of Arizona, Tucson, AZ 85724, USA
| | - Jolanda van der Velden
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands; ICIN-Netherlands Heart Institute, The Netherlands.
| |
Collapse
|
43
|
Mosqueira M, Zeiger U, Förderer M, Brinkmeier H, Fink RHA. Cardiac and respiratory dysfunction in Duchenne muscular dystrophy and the role of second messengers. Med Res Rev 2013; 33:1174-213. [PMID: 23633235 DOI: 10.1002/med.21279] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Duchenne muscular dystrophy (DMD) affects young boys and is characterized by the absence of dystrophin, a large cytoskeletal protein present in skeletal and cardiac muscle cells and neurons. The heart and diaphragm become necrotic in DMD patients and animal models of DMD, resulting in cardiorespiratory failure as the leading cause of death. The major consequences of the absence of dystrophin are high levels of intracellular Ca(2+) and the unbalanced production of NO that can finally trigger protein degradation and cell death. Cytoplasmic increase in Ca(2+) concentration directly and indirectly triggers different processes such as necrosis, fibrosis, and activation of macrophages. The absence of the neuronal isoform of nitric oxide synthase (nNOS) and the overproduction of NO by the inducible isoform (iNOS) further increase the intracellular Ca(2+) via a hypernitrosylation of the ryanodine receptor. NO overproduction, which further induces the expression of iNOS but decreases the expression of the endothelial isoform (eNOS), deregulates the muscle tissue blood flow creating an ischemic situation. The high levels of Ca(2+) in dystrophic muscles and the ischemic state of the muscle tissue would culminate in a positive feedback loop. While efforts continue toward optimizing cardiac and respiratory care of DMD patients, both Ca(2+) and NO in cardiac and respiratory muscle pathways have been shown to be important to the etiology of the disease. Understanding the mechanisms behind the fine regulation of Ca(2+) -NO may be important for a noninterventional and noninvasive supportive approach to treat DMD patients, improving the quality of life and natural history of DMD patients.
Collapse
Affiliation(s)
- Matias Mosqueira
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, INF326, Heidelberg University, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
44
|
Randazzo D, Giacomello E, Lorenzini S, Rossi D, Pierantozzi E, Blaauw B, Reggiani C, Lange S, Peter AK, Chen J, Sorrentino V. Obscurin is required for ankyrinB-dependent dystrophin localization and sarcolemma integrity. ACTA ACUST UNITED AC 2013; 200:523-36. [PMID: 23420875 PMCID: PMC3575540 DOI: 10.1083/jcb.201205118] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Obscurin contributes to the organization of subsarcolemma microtubules, localization of dystrophin at costameres, and maintenance of sarcolemmal integrity in skeletal muscle fibers. Obscurin is a large myofibrillar protein that contains several interacting modules, one of which mediates binding to muscle-specific ankyrins. Interaction between obscurin and the muscle-specific ankyrin sAnk1.5 regulates the organization of the sarcoplasmic reticulum in striated muscles. Additional muscle-specific ankyrin isoforms, ankB and ankG, are localized at the subsarcolemma level, at which they contribute to the organization of dystrophin and β-dystroglycan at costameres. In this paper, we report that in mice deficient for obscurin, ankB was displaced from its localization at the M band, whereas localization of ankG at the Z disk was not affected. In obscurin KO mice, localization at costameres of dystrophin, but not of β-dystroglycan, was altered, and the subsarcolemma microtubule cytoskeleton was disrupted. In addition, these mutant mice displayed marked sarcolemmal fragility and reduced muscle exercise tolerance. Altogether, the results support a model in which obscurin, by targeting ankB at the M band, contributes to the organization of subsarcolemma microtubules, localization of dystrophin at costameres, and maintenance of sarcolemmal integrity.
Collapse
Affiliation(s)
- Davide Randazzo
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Di Felice V, Serradifalco C, Rizzuto L, De Luca A, Rappa F, Barone R, Di Marco P, Cassata G, Puleio R, Verin L, Motta A, Migliaresi C, Guercio A, Zummo G. Silk fibroin scaffolds enhance cell commitment of adult rat cardiac progenitor cells. J Tissue Eng Regen Med 2013; 9:E51-64. [PMID: 23592297 DOI: 10.1002/term.1739] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Revised: 01/18/2013] [Accepted: 02/05/2013] [Indexed: 01/30/2023]
Abstract
The use of three-dimensional (3D) cultures may induce cardiac progenitor cells to synthesize their own extracellular matrix (ECM) and sarcomeric proteins to initiate cardiac differentiation. 3D cultures grown on synthetic scaffolds may favour the implantation and survival of stem cells for cell therapy when pharmacological therapies are not efficient in curing cardiovascular diseases and when organ transplantation remains the only treatment able to rescue the patient's life. Silk fibroin-based scaffolds may be used to increase cell affinity to biomaterials and may be chemically modified to improve cell adhesion. In the present study, porous, partially orientated and electrospun nanometric nets were used. Cardiac progenitor cells isolated from adult rats were seeded by capillarity in the 3D structures and cultured inside inserts for 21 days. Under this condition, the cells expressed a high level of sarcomeric and cardiac proteins and synthesized a great quantity of ECM. In particular, partially orientated scaffolds induced the synthesis of titin, which is a fundamental protein in sarcomere assembly.
Collapse
Affiliation(s)
- Valentina Di Felice
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| | - Claudia Serradifalco
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| | - Luigi Rizzuto
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| | - Angela De Luca
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| | - Francesca Rappa
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| | - Rosario Barone
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| | | | - Giovanni Cassata
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Roberto Puleio
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Lucia Verin
- Department of Materials Engineering and Industrial Technologies and Biotech Research Centre, University of Trento, Italy
- European Institute of Excellence in Tissue Engineering and Regenerative Medicine and INSTM Research Unit, Trento, Italy
| | - Antonella Motta
- Department of Materials Engineering and Industrial Technologies and Biotech Research Centre, University of Trento, Italy
- European Institute of Excellence in Tissue Engineering and Regenerative Medicine and INSTM Research Unit, Trento, Italy
| | - Claudio Migliaresi
- Department of Materials Engineering and Industrial Technologies and Biotech Research Centre, University of Trento, Italy
- European Institute of Excellence in Tissue Engineering and Regenerative Medicine and INSTM Research Unit, Trento, Italy
| | - Annalisa Guercio
- Istituto Zooprofilattico Sperimentale della Sicilia, Palermo, Italy
| | - Giovanni Zummo
- Department of Experimental Biomedicine and Clinical Neurosciences, University of Palermo, Italy
| |
Collapse
|
46
|
Hersch N, Wolters B, Dreissen G, Springer R, Kirchgeßner N, Merkel R, Hoffmann B. The constant beat: cardiomyocytes adapt their forces by equal contraction upon environmental stiffening. Biol Open 2013; 2:351-61. [PMID: 23519595 PMCID: PMC3603417 DOI: 10.1242/bio.20133830] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 12/23/2012] [Indexed: 12/25/2022] Open
Abstract
Cardiomyocytes are responsible for the permanent blood flow by coordinated heart contractions. This vital function is accomplished over a long period of time with almost the same performance, although heart properties, as its elasticity, change drastically upon aging or as a result of diseases like myocardial infarction. In this paper we have analyzed late rat embryonic heart muscle cells' morphology, sarcomere/costamere formation and force generation patterns on substrates of various elasticities ranging from ∼1 to 500 kPa, which covers physiological and pathological heart stiffnesses. Furthermore, adhesion behaviour, as well as single myofibril/sarcomere contraction patterns, was characterized with high spatial resolution in the range of physiological stiffnesses (15 kPa to 90 kPa). Here, sarcomere units generate an almost stable contraction of ∼4%. On stiffened substrates the contraction amplitude remains stable, which in turn leads to increased force levels allowing cells to adapt almost instantaneously to changing environmental stiffness. Furthermore, our data strongly indicate specific adhesion to flat substrates via both costameric and focal adhesions. The general appearance of the contractile and adhesion apparatus remains almost unaffected by substrate stiffness.
Collapse
Affiliation(s)
- Nils Hersch
- Institute of Complex Systems, ICS-7: Biomechanics, Forschungszentrum Jülich GmbH , 52425 Jülich , Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Biophysical Forces Modulate the Costamere and Z-Disc for Sarcomere Remodeling in Heart Failure. BIOPHYSICS OF THE FAILING HEART 2013. [DOI: 10.1007/978-1-4614-7678-8_7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Thoss F, Dietrich F, Punkt K, Illenberger S, Rottner K, Himmel M, Ziegler WH. Metavinculin: New insights into functional properties of a muscle adhesion protein. Biochem Biophys Res Commun 2012; 430:7-13. [PMID: 23159629 DOI: 10.1016/j.bbrc.2012.11.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 11/04/2012] [Indexed: 11/16/2022]
Abstract
Metavinculin is a muscle-specific splice variant of the ubiquitously expressed cytoskeletal adaptor protein vinculin. Both proteins are thought to be co-expressed in all muscle types where they co-localize to microfilament-associated adhesion sites. It has been shown that a metavinculin-specific insertion of 68 amino acids alters the biochemical properties of the five-helix bundle in the tail domain. Here, we demonstrate that the metavinculin-specific helix H1' plays an important role for protein stability of the tail domain, since a point mutation in this helix, R975W, which is associated with the occurrence of dilated cardiomyopathy in man, further decreases thermal stability of the metavinculin tail domain. In striated muscle progenitor cells (myoblasts), both, metavinculin and the R975W mutant show significantly reduced, albeit distinctive residency and exchange rates in adhesion sites as compared to vinculin. In contrast to previous studies, we show that metavinculin is localized in a muscle fiber type-dependent fashion to the costameres of striated muscle, reflecting the individual metabolic and physiological status of a given muscle fiber. Metavinculin expression is highest in fast, glycolytic muscle fibers and virtually absent in M. diaphragmaticus, a skeletal muscle entirely lacking fast, glycolytic fibers. In summary, our data suggest that metavinculin enrichment in attachment sites of muscle cells leads to higher mechanical stability of adhesion complexes allowing for greater shear force resistance.
Collapse
Affiliation(s)
- Florian Thoss
- Interdisciplinary Center for Clinical Research (IZKF) Leipzig, Faculty of Medicine, University of Leipzig, D-04103 Leipzig, Germany
| | | | | | | | | | | | | |
Collapse
|
49
|
Heavy and light roles: myosin in the morphogenesis of the heart. Cell Mol Life Sci 2012; 70:1221-39. [PMID: 22955375 PMCID: PMC3602621 DOI: 10.1007/s00018-012-1131-1] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 08/08/2012] [Accepted: 08/13/2012] [Indexed: 01/10/2023]
Abstract
Myosin is an essential component of cardiac muscle, from the onset of cardiogenesis through to the adult heart. Although traditionally known for its role in energy transduction and force development, recent studies suggest that both myosin heavy-chain and myosin light-chain proteins are required for a correctly formed heart. Myosins are structural proteins that are not only expressed from early stages of heart development, but when mutated in humans they may give rise to congenital heart defects. This review will discuss the roles of myosin, specifically with regards to the developing heart. The expression of each myosin protein will be described, and the effects that altering expression has on the heart in embryogenesis in different animal models will be discussed. The human molecular genetics of the myosins will also be reviewed.
Collapse
|
50
|
Okina E, Grossi A, Gopal S, Multhaupt HAB, Couchman JR. Alpha-actinin interactions with syndecan-4 are integral to fibroblast-matrix adhesion and regulate cytoskeletal architecture. Int J Biochem Cell Biol 2012; 44:2161-74. [PMID: 22940199 DOI: 10.1016/j.biocel.2012.08.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 08/01/2012] [Accepted: 08/15/2012] [Indexed: 11/26/2022]
Abstract
All cells of the musculoskeletal system possess transmembrane syndecan proteoglycans, notably syndecan-4. In fibroblasts it regulates integrin-mediated adhesion to the extracellular matrix. Syndecan-4 null mice have a complex wound repair phenotype while their fibroblasts have reduced focal adhesions and matrix contraction abilities. Signalling through syndecan-4 core protein to the actin cytoskeleton involves protein kinase Cα and Rho family G proteins but also direct interactions with α-actinin. The contribution of the latter interaction to cell-matrix adhesion is not defined but investigated here since manipulation of Rho GTPase and its downstream targets could not restore a wild type microfilament organisation to syndecan-4 null cells. Microarray and protein analysis revealed no significant alterations in mRNA or protein levels for actin- or α-actinin associated proteins when wild type and syndecan-4 knockout fibroblasts were compared. The binding site for syndecan-4 cytoplasmic domain was identified as spectrin repeat 4 of α-actinin while further experiments confirmed the importance of this interaction in stabilising cell-matrix junctions. However, α-actinin is also present in adherens junctions, these organelles not being disrupted in the absence of syndecan-4. Indeed, co-culture of wild type and knockout cells led to adherens junction-associated stress fibre formation in cells lacking syndecan-4, supporting the hypothesis that the proteoglycan regulates cell-matrix adhesion and its associated microfilament bundles at a post-translational level. These data provide an additional dimension to syndecan function related to tension at the cell-matrix interface, wound healing and potentially fibrosis.
Collapse
Affiliation(s)
- E Okina
- Department of Biomedical Sciences, University of Copenhagen, Denmark
| | | | | | | | | |
Collapse
|