1
|
Lane KV, Dow LP, Castillo EA, Boros R, Feinstein SD, Pardon G, Pruitt BL. Cell Architecture and Dynamics of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPSC-CMs) on Hydrogels with Spatially Patterned Laminin and N-Cadherin. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39680735 DOI: 10.1021/acsami.4c11934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Controlling cellular shape with micropatterning extracellular matrix (ECM) proteins on hydrogels has been shown to improve the reproducibility of the cell structure, enhancing our ability to collect statistics on single-cell behaviors. Patterning methods have advanced efforts in developing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) as a promising human model for studies of the heart structure, function, and disease. Patterned single hiPSC-CMs have exhibited phenotypes closer to mature, primary CMs across several metrics, including sarcomere alignment and contractility, area and aspect ratio, and force production. Micropatterning of hiPSC-CM pairs has shown further improvement of hiPSC-CM contractility compared to patterning single cells, suggesting that CM-CM interactions improve hiPSC-CM function. However, whether patterning single hiPSC-CMs on a protein associated with CM-CM adhesion, like N-cadherin, can drive similar enhancement of the hiPSC-CM structure and function has not been tested. To address this, we developed a novel dual-protein patterning process featuring covalent binding of proteins at the hydrogel surface to ensure robust force transfer and force sensing. The patterns comprised rectangular laminin islands for attachment across the majority of the cell area, with N-cadherin "end caps" to imitate CM-CM adherens junctions. We used this method to geometrically control single-cell CMs on deformable hydrogels suitable for traction force microscopy (TFM) to observe cellular dynamics. We seeded α-actinin::GFP-tagged hiPSC-CMs on dual-protein patterned hydrogels and verified the interaction between hiPSC-CMs and N-cadherin end caps via immunofluorescent staining. We found that hiPSC-CMs on dual-protein patterns exhibited higher cell area and contractility in the direction of sarcomere organization than those on laminin-only patterns but no difference in sarcomere organization or total force production. This work demonstrates a method for covalent patterning of multiple proteins on polyacrylamide hydrogels for mechanobiological studies. However, we conclude that N-cadherin only modestly improves single-cell patterned hiPSC-CM models and is not sufficient to elicit increases in contractility observed in hiPSC-CM pairs.
Collapse
Affiliation(s)
- Kerry V Lane
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Liam P Dow
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Erica A Castillo
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305, United States
| | - Rémi Boros
- Department of Physics, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Samuel D Feinstein
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Gaspard Pardon
- AGORA Cancer Research Center, Swiss Federal Institute of Technology of Lausanne, Lausanne CH-1011, Switzerland
| | - Beth L Pruitt
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
2
|
DePalma SJ, Jilberto J, Stis AE, Huang DD, Lo J, Davidson CD, Chowdhury A, Kent RN, Jewett ME, Kobeissi H, Chen CS, Lejeune E, Helms AS, Nordsletten DA, Baker BM. Matrix Architecture and Mechanics Regulate Myofibril Organization, Costamere Assembly, and Contractility in Engineered Myocardial Microtissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309740. [PMID: 39558513 DOI: 10.1002/advs.202309740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/09/2024] [Indexed: 11/20/2024]
Abstract
The mechanical function of the myocardium is defined by cardiomyocyte contractility and the biomechanics of the extracellular matrix (ECM). Understanding this relationship remains an important unmet challenge due to limitations in existing approaches for engineering myocardial tissue. Here, they established arrays of cardiac microtissues with tunable mechanics and architecture by integrating ECM-mimetic synthetic, fiber matrices, and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), enabling real-time contractility readouts, in-depth structural assessment, and tissue-specific computational modeling. They found that the stiffness and alignment of matrix fibers distinctly affect the structural development and contractile function of pure iPSC-CM tissues. Further examination into the impact of fibrous matrix stiffness enabled by computational models and quantitative immunofluorescence implicates cell-ECM interactions in myofibril assembly, myofibril maturation, and notably costamere assembly, which correlates with improved contractile function of tissues. These results highlight how iPSC-CM tissue models with controllable architecture and mechanics can elucidate mechanisms of tissue maturation and disease.
Collapse
Affiliation(s)
- Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Javiera Jilberto
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Austin E Stis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Darcy D Huang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jason Lo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Aamilah Chowdhury
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Maggie E Jewett
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hiba Kobeissi
- Department of Mechanical Engineering, Boston University, Boston, MA, 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, 02215, USA
| | - Adam S Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David A Nordsletten
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, School of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, London, SE1 7EH, UK
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
3
|
DePalma SJ, Jillberto J, Stis AE, Huang DD, Lo J, Davidson CD, Chowdhury A, Jewett ME, Kobeissi H, Chen CS, Lejeune E, Helms AS, Nordsletten DA, Baker BM. Matrix architecture and mechanics regulate myofibril organization, costamere assembly, and contractility of engineered myocardial microtissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563346. [PMID: 37961415 PMCID: PMC10634701 DOI: 10.1101/2023.10.20.563346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The mechanical function of the myocardium is defined by cardiomyocyte contractility and the biomechanics of the extracellular matrix (ECM). Understanding this relationship remains an important unmet challenge due to limitations in existing approaches for engineering myocardial tissue. Here, we established arrays of cardiac microtissues with tunable mechanics and architecture by integrating ECM-mimetic synthetic, fiber matrices and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), enabling real-time contractility readouts, in-depth structural assessment, and tissue-specific computational modeling. We find that the stiffness and alignment of matrix fibers distinctly affect the structural development and contractile function of pure iPSC-CM tissues. Further examination into the impact of fibrous matrix stiffness enabled by computational models and quantitative immunofluorescence implicates cell-ECM interactions in myofibril assembly and notably costamere assembly, which correlates with improved contractile function of tissues. These results highlight how iPSC-CM tissue models with controllable architecture and mechanics can inform the design of translatable regenerative cardiac therapies.
Collapse
|
4
|
Fang Y, Sun W, Zhang T, Xiong Z. Recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps: A review. Biomaterials 2021; 280:121298. [PMID: 34864451 DOI: 10.1016/j.biomaterials.2021.121298] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022]
Abstract
The field of cardiac tissue engineering has advanced over the past decades; however, most research progress has been limited to engineered cardiac tissues (ECTs) at the microscale with minimal geometrical complexities such as 3D strips and patches. Although microscale ECTs are advantageous for drug screening applications because of their high-throughput and standardization characteristics, they have limited translational applications in heart repair and the in vitro modeling of cardiac function and diseases. Recently, researchers have made various attempts to construct engineered cardiac pumps (ECPs) such as chambered ventricles, recapitulating the geometrical complexity of the native heart. The transition from microscale ECTs to ECPs at a translatable scale would greatly accelerate their translational applications; however, researchers are confronted with several major hurdles, including geometrical reconstruction, vascularization, and functional maturation. Therefore, the objective of this paper is to review the recent advances on bioengineering approaches for fabrication of functional engineered cardiac pumps. We first review the bioengineering approaches to fabricate ECPs, and then emphasize the unmatched potential of 3D bioprinting techniques. We highlight key advances in bioprinting strategies with high cell density as researchers have begun to realize the critical role that the cell density of non-proliferative cardiomyocytes plays in the cell-cell interaction and functional contracting performance. We summarize the current approaches to engineering vasculatures both at micro- and meso-scales, crucial for the survival of thick cardiac tissues and ECPs. We showcase a variety of strategies developed to enable the functional maturation of cardiac tissues, mimicking the in vivo environment during cardiac development. By highlighting state-of-the-art research, this review offers personal perspectives on future opportunities and trends that may bring us closer to the promise of functional ECPs.
Collapse
Affiliation(s)
- Yongcong Fang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China
| | - Wei Sun
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA, 19104, USA
| | - Ting Zhang
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| | - Zhuo Xiong
- Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, PR China; Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Beijing, 100084, PR China; "Biomanufacturing and Engineering Living Systems" Innovation International Talents Base (111 Base), Beijing, 100084, PR China.
| |
Collapse
|
5
|
Latham SL, Weiß N, Schwanke K, Thiel C, Croucher DR, Zweigerdt R, Manstein DJ, Taft MH. Myosin-18B Regulates Higher-Order Organization of the Cardiac Sarcomere through Thin Filament Cross-Linking and Thick Filament Dynamics. Cell Rep 2021; 32:108090. [PMID: 32877672 DOI: 10.1016/j.celrep.2020.108090] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/07/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022] Open
Abstract
MYO18B loss-of-function mutations and depletion significantly compromise the structural integrity of striated muscle sarcomeres. The molecular function of the encoded protein, myosin-18B (M18B), within the developing muscle is unknown. Here, we demonstrate that recombinant M18B lacks motor ATPase activity and harbors previously uncharacterized N-terminal actin-binding domains, properties that make M18B an efficient actin cross-linker and molecular brake capable of regulating muscle myosin-2 contractile forces. Spatiotemporal analysis of M18B throughout cardiomyogenesis and myofibrillogenesis reveals that this structural myosin undergoes nuclear-cytoplasmic redistribution during myogenic differentiation, where its incorporation within muscle stress fibers coincides with actin striation onset. Furthermore, this analysis shows that M18B is directly integrated within the muscle myosin thick filament during myofibril maturation. Altogether, our data suggest that M18B has evolved specific biochemical properties that allow it to define and maintain sarcomeric organization from within the thick filament via its dual actin cross-linking and motor modulating capabilities.
Collapse
Affiliation(s)
- Sharissa L Latham
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany; The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Hospital Clinical School, UNSW Sydney, NSW 2052, Australia
| | - Nadine Weiß
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany
| | - Kristin Schwanke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover 30625, Germany
| | - Claudia Thiel
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany
| | - David R Croucher
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Hospital Clinical School, UNSW Sydney, NSW 2052, Australia
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover 30625, Germany
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany
| | - Manuel H Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany.
| |
Collapse
|
6
|
Wang J, Fan Y, Dube S, Agassy NW, Dube DK, Sanger JM, Sanger JW. Myofibril assembly and the roles of the ubiquitin proteasome system. Cytoskeleton (Hoboken) 2020; 77:456-479. [PMID: 33124174 DOI: 10.1002/cm.21641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 10/07/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
De novo assembly of myofibrils in vertebrate cross-striated muscles progresses in three distinct steps, first from a minisarcomeric alignment of several nonmuscle and muscle proteins in premyofibrils, followed by insertions of additional proteins and increased organization in nascent myofibrils, ending with mature contractile myofibrils. In a search for controls of the process of myofibril assembly, we discovered that the transition from nascent to mature myofibrils could be halted by inhibitors of three distinct functions of the ubiquitin proteasome system (UPS). First, inhibition of pathway to E3 Cullin ligases that ubiquitinate proteins led to an arrest of myofibrillogenesis at the nascent myofibril stage. Second, inhibition of p97 protein extractions of ubiquitinated proteins led to a similar arrest of myofibrillogenesis at the nascent myofibril stage. Third, inhibitors of proteolytic action by proteasomes also blocked nascent myofibrils from transitioning to mature myofibrils. In contrast, inhibitors of autophagy or lysosomes did not affect myofibrillogenesis. To probe for differences in the effects of UPS inhibitors during myofibrillogenesis, we analyzed by fluorescence recovery after photobleaching the exchange rates of two selected sarcomeric proteins (muscle myosin II heavy chains and light chains). In the presence of p97 and proteasomal inhibitors, the dynamics of each of these two myosin proteins decreased in the nascent myofibril stage, but were unaffected in the mature myofibril stage. The increased stability of myofibrils occurring in the transition from nascent to mature myofibril assembly indicates the importance of dynamics and selective destruction in the muscle myosin II proteins for the remodeling of nascent to mature myofibrils.
Collapse
Affiliation(s)
- Jushuo Wang
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Yingli Fan
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Syamalima Dube
- Department of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Nicodeme Wanko Agassy
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Dipak K Dube
- Department of Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jean M Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Joseph W Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
7
|
Taneja N, Neininger AC, Burnette DT. Coupling to substrate adhesions drives the maturation of muscle stress fibers into myofibrils within cardiomyocytes. Mol Biol Cell 2020; 31:1273-1288. [PMID: 32267210 PMCID: PMC7353145 DOI: 10.1091/mbc.e19-11-0652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Forces generated by heart muscle contraction must be balanced by adhesion to the extracellular matrix (ECM) and to other cells for proper heart function. Decades of data have suggested that cell-ECM adhesions are important for sarcomere assembly. However, the relationship between cell-ECM adhesions and sarcomeres assembling de novo remains untested. Sarcomeres arise from muscle stress fibers (MSFs) that are translocating on the top (dorsal) surface of cultured cardiomyocytes. Using an array of tools to modulate cell-ECM adhesion, we established a strong positive correlation between the extent of cell-ECM adhesion and sarcomere assembly. On the other hand, we found a strong negative correlation between the extent of cell-ECM adhesion and the rate of MSF translocation, a phenomenon also observed in nonmuscle cells. We further find a conserved network architecture that also exists in nonmuscle cells. Taken together, our results show that cell-ECM adhesions mediate coupling between the substrate and MSFs, allowing their maturation into sarcomere-containing myofibrils.
Collapse
Affiliation(s)
- Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
| | - Abigail C Neininger
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232
| |
Collapse
|
8
|
Rasmussen ML, Taneja N, Neininger AC, Wang L, Robertson GL, Riffle SN, Shi L, Knollmann BC, Burnette DT, Gama V. MCL-1 Inhibition by Selective BH3 Mimetics Disrupts Mitochondrial Dynamics Causing Loss of Viability and Functionality of Human Cardiomyocytes. iScience 2020; 23:101015. [PMID: 32283523 PMCID: PMC7155208 DOI: 10.1016/j.isci.2020.101015] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 02/25/2020] [Accepted: 03/24/2020] [Indexed: 12/17/2022] Open
Abstract
MCL-1 is a well-characterized inhibitor of cell death that has also been shown to be a regulator of mitochondrial dynamics in human pluripotent stem cells. We used cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs) to uncover whether MCL-1 is crucial for cardiac function and survival. Inhibition of MCL-1 by BH3 mimetics resulted in the disruption of mitochondrial morphology and dynamics as well as disorganization of the actin cytoskeleton. Interfering with MCL-1 function affects the homeostatic proximity of DRP-1 and MCL-1 at the outer mitochondrial membrane, resulting in decreased functionality of hiPSC-CMs. Cardiomyocytes display abnormal cardiac performance even after caspase inhibition, supporting a nonapoptotic activity of MCL-1 in hiPSC-CMs. BH3 mimetics targeting MCL-1 are promising anti-tumor therapeutics. Progression toward using BCL-2 family inhibitors, especially targeting MCL-1, depends on understanding its canonical function not only in preventing apoptosis but also in the maintenance of mitochondrial dynamics and function. BH3 mimetics targeting MCL-1 disrupt the mitochondrial network of human iPSC-CMs The BH3-mimetic-mediated effects on mitochondrial dynamics are DRP-1-dependent Targeting MCL-1 affects the survival and function of human cardiomyocytes Human iPSC-derived cardiomyocytes can be used to reveal toxicity of MCL-1 inhibitors
Collapse
Affiliation(s)
- Megan L Rasmussen
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Nilay Taneja
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Abigail C Neininger
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Lili Wang
- Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Nashville, TN 37232, USA
| | - Gabriella L Robertson
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Stellan N Riffle
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Linzheng Shi
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Bjorn C Knollmann
- Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Center for Arrhythmia Research and Therapeutics, Department of Medicine, Nashville, TN 37232, USA
| | - Dylan T Burnette
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Vivian Gama
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
9
|
Mechanical Forces Regulate Cardiomyocyte Myofilament Maturation via the VCL-SSH1-CFL Axis. Dev Cell 2019; 51:62-77.e5. [PMID: 31495694 DOI: 10.1016/j.devcel.2019.08.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/02/2019] [Accepted: 08/07/2019] [Indexed: 01/07/2023]
Abstract
Mechanical forces regulate cell behavior and tissue morphogenesis. During cardiac development, mechanical stimuli from the heartbeat are required for cardiomyocyte maturation, but the underlying molecular mechanisms remain unclear. Here, we first show that the forces of the contracting heart regulate the localization and activation of the cytoskeletal protein vinculin (VCL), which we find to be essential for myofilament maturation. To further analyze the role of VCL in this process, we examined its interactome in contracting versus non-contracting cardiomyocytes and, in addition to several known interactors, including actin regulators, identified the slingshot protein phosphatase SSH1. We show how VCL recruits SSH1 and its effector, the actin depolymerizing factor cofilin (CFL), to regulate F-actin rearrangement and promote cardiomyocyte myofilament maturation. Overall, our results reveal that mechanical forces generated by cardiac contractility regulate cardiomyocyte maturation through the VCL-SSH1-CFL axis, providing further insight into how mechanical forces are transmitted intracellularly to regulate myofilament maturation.
Collapse
|
10
|
Xiao J, Li F, Yang Q, Zeng X, Ke Z. Co‐expression analysis provides important module and pathways of human dilated cardiomyopathy. J Cell Physiol 2019; 235:494-503. [PMID: 31236962 DOI: 10.1002/jcp.28989] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/24/2019] [Accepted: 03/05/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Junhui Xiao
- Department of Cardiology, Huadu District People's Hospital Southern Medical University Guangzhou China
| | - Fang Li
- Department of Cardiology, Huadu District People's Hospital Southern Medical University Guangzhou China
| | - Qianzhao Yang
- Department of Cardiology, Huadu District People's Hospital Southern Medical University Guangzhou China
| | | | - Zun‐Ping Ke
- Department of Cardiology, The Fifth People's Hospital of Shanghai Fudan University Shanghai China
| |
Collapse
|
11
|
Besser RR, Ishahak M, Mayo V, Carbonero D, Claure I, Agarwal A. Engineered Microenvironments for Maturation of Stem Cell Derived Cardiac Myocytes. Am J Cancer Res 2018; 8:124-140. [PMID: 29290797 PMCID: PMC5743464 DOI: 10.7150/thno.19441] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 10/19/2017] [Indexed: 01/11/2023] Open
Abstract
Through the use of stem cell-derived cardiac myocytes, tissue-engineered human myocardial constructs are poised for modeling normal and diseased physiology of the heart, as well as discovery of novel drugs and therapeutic targets in a human relevant manner. This review highlights the recent bioengineering efforts to recapitulate microenvironmental cues to further the maturation state of newly differentiated cardiac myocytes. These techniques include long-term culture, co-culture, exposure to mechanical stimuli, 3D culture, cell-matrix interactions, and electrical stimulation. Each of these methods has produced various degrees of maturation; however, a standardized measure for cardiomyocyte maturation is not yet widely accepted by the scientific community.
Collapse
|
12
|
Liu H, Qin W, Wang Z, Shao Y, Wang J, Borg TK, Gao BZ, Xu M. Disassembly of myofibrils and potential imbalanced forces on Z-discs in cultured adult cardiomyocytes. Cytoskeleton (Hoboken) 2016; 73:246-57. [PMID: 27072949 DOI: 10.1002/cm.21298] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 11/08/2022]
Abstract
Myofibrils are the main protein structures that generate force in the beating heart. Myofibril disassembly is related to many physiological and pathological processes. This study investigated, in a cultured rat adult cardiomyocyte model, the effect of force imbalance on myofibril disassembly. The imbalance of forces that were exerted on Z-discs was induced by the synergistic effect of broken intercalated discs and actin-myosin interaction. Cardiomyocytes with well-preserved intercalated discs were isolated from adult rat ventricles. The ultrastructure of cardiomyocyte was observed using a customized two-photon excitation fluorescence and second harmonic generation imaging system. The contraction of cardiomyocytes was recorded with a high-speed CCD camera, and the movement of cellular components was analyzed using a contractile imaging assay technique. The cardiomyocyte dynamic remodeling process was recorded using a time-lapse imaging system. The role of actin-myosin interaction in myofibril disassembly was investigated by incubating cardiomyocytes with blebbistatin (25 μM). Results demonstrated that the hierarchical disassembly process of myofibrils was initiated from cardiomyocyte free ends where intercalated discs had broken, during which the desmin network near the free cell ends was destroyed to release single myofibrils. Analysis of force (based on a schematic model of cardiomyocytes connected at intercalated discs) suggests that breaking of intercalated discs caused force imbalance on both sides of the Z-discs adjacent to the cell ends due to actin-myosin interaction. The damaged intercalated discs and actin-myosin interaction induced force imbalance on both sides of the Z-discs, which played an important role in the hierarchical disassembly of myofibrils. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Honghai Liu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267
| | - Wan Qin
- Department of Bioengineering and COMSET, Clemson University, Clemson, South Carolina, 29634
| | - Zhonghai Wang
- Department of Bioengineering and COMSET, Clemson University, Clemson, South Carolina, 29634
| | - Yonghong Shao
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Jingcai Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267
| | - Thomas K Borg
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, 29425
| | - Bruce Z Gao
- Department of Bioengineering and COMSET, Clemson University, Clemson, South Carolina, 29634
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, 45267
| |
Collapse
|
13
|
Myhre JL, Hills JA, Jean F, Pilgrim DB. Unc45b is essential for early myofibrillogenesis and costamere formation in zebrafish. Dev Biol 2014; 390:26-40. [DOI: 10.1016/j.ydbio.2014.02.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 02/25/2014] [Indexed: 01/16/2023]
|
14
|
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: 8.2] [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
|
15
|
Nguyen PD, Hsiao ST, Sivakumaran P, Lim SY, Dilley RJ. Enrichment of neonatal rat cardiomyocytes in primary culture facilitates long-term maintenance of contractility in vitro. Am J Physiol Cell Physiol 2012; 303:C1220-8. [PMID: 22932682 DOI: 10.1152/ajpcell.00449.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Long-term culture of primary neonatal rat cardiomyocytes is limited by the loss of spontaneous contractile phenotype within weeks in culture. This may be due to loss of contractile cardiomyocytes from the culture or overgrowth of the non-cardiomyocyte population. Using the mitochondria specific fluorescent dye, tetramethylrhodamine methyl ester perchlorate (TMRM), we showed that neonatal rat cardiomyocytes enriched by fluorescence-activated cell sorting can be maintained as contractile cultures for long periods (24-wk culture vs. 2 wk for unsorted cardiomyocytes). Long-term culture of this purified cardiomyocyte (TMRM high) population retained the expression of cardiomyocyte markers, continued calcium cycling, and displayed cyclic electrical activity that could be regulated pharmacologically. These findings suggest that non-cardiomyocyte populations can negatively influence contractility of cardiomyocytes in culture and that by purifying cardiomyocytes, the cultures retain potential as an experimental model for longitudinal studies of cardiomyocyte biology in vitro.
Collapse
Affiliation(s)
- Phong D Nguyen
- O’Brien Institute and University of Melbourne, Department of Surgery, St Vincent’s Hospital, Victoria, Australia
| | | | | | | | | |
Collapse
|
16
|
Sheehy SP, Grosberg A, Parker KK. The contribution of cellular mechanotransduction to cardiomyocyte form and function. Biomech Model Mechanobiol 2012; 11:1227-39. [PMID: 22772714 DOI: 10.1007/s10237-012-0419-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 06/25/2012] [Indexed: 01/07/2023]
Abstract
Myocardial development is regulated by an elegantly choreographed ensemble of signaling events mediated by a multitude of intermediates that take a variety of forms. Cellular differentiation and maturation are a subset of vertically integrated processes that extend over several spatial and temporal scales to create a well-defined collective of cells that are able to function cooperatively and reliably at the organ level. Early efforts to understand the molecular mechanisms of cardiomyocyte fate determination focused primarily on genetic and chemical mediators of this process. However, increasing evidence suggests that mechanical interactions between the extracellular matrix (ECM) and cell surface receptors as well as physical interactions between neighboring cells play important roles in regulating the signaling pathways controlling the developmental processes of the heart. Interdisciplinary efforts have made it apparent that the influence of the ECM on cellular behavior occurs through a multitude of physical mechanisms, such as ECM boundary conditions, elasticity, and the propagation of mechanical signals to intracellular compartments, such as the nucleus. In addition to experimental studies, a number of mathematical models have been developed that attempt to capture the interplay between cells and their local microenvironment and the influence these interactions have on cellular self-assembly and functional behavior. Nevertheless, many questions remain unanswered concerning the mechanism through which physical interactions between cardiomyocytes and their environment are translated into biochemical cellular responses and how these signaling modalities can be utilized in vitro to fabricate myocardial tissue constructs from stem cell-derived cardiomyocytes that more faithfully represent their in vivo counterpart. These studies represent a broad effort to characterize biological form as a conduit for information transfer that spans the nanometer length scale of proteins to the meter length scale of the patient and may yield new insights into the contribution of mechanotransduction into heart development and disease.
Collapse
Affiliation(s)
- Sean P Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Pierce Hall Rm. 321, 29 Oxford St., Cambridge, MA 02138, USA
| | | | | |
Collapse
|
17
|
Zebda N, Dubrovskyi O, Birukov KG. Focal adhesion kinase regulation of mechanotransduction and its impact on endothelial cell functions. Microvasc Res 2011; 83:71-81. [PMID: 21741394 DOI: 10.1016/j.mvr.2011.06.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 06/19/2011] [Accepted: 06/20/2011] [Indexed: 01/06/2023]
Abstract
Vascular endothelial cells lining the blood vessels form the interface between the bloodstream and the vessel wall and as such they are continuously subjected to shear and cyclic stress from the flowing blood in the lumen. Additional mechanical stimuli are also imposed on these cells in the form of substrate stiffness transmitted from the extracellular matrix components in the basement membrane, and additional mechanical loads imposed on the lung endothelium as the result of respiration or mechanical ventilation in clinical settings. Focal adhesions (FAs) are complex structures assembled at the abluminal endothelial plasma membrane which connect the extracellular filamentous meshwork to the intracellular cytoskeleton and hence constitute the ideal checkpoint capable of controlling or mediating transduction of bidirectional mechanical signals. In this review we focus on focal adhesion kinase (FAK), a component of FAs, which has been studied for a number of years with regards to its involvement in mechanotransduction. We analyzed the recent advances in the understanding of the role of FAK in the signaling cascade(s) initiated by various mechanical stimuli with particular emphasis on potential implications on endothelial cell functions.
Collapse
Affiliation(s)
- Noureddine Zebda
- Section of Pulmonary and Critical Care, Lung Injury Center, Department of Medicine, The University of Chicago, IL 60637, USA
| | | | | |
Collapse
|
18
|
Mechanotransduction: the role of mechanical stress, myocyte shape, and cytoskeletal architecture on cardiac function. Pflugers Arch 2011; 462:89-104. [PMID: 21499986 DOI: 10.1007/s00424-011-0951-4] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2011] [Accepted: 02/27/2011] [Indexed: 12/16/2022]
Abstract
Mechanotransduction refers to the conversion of mechanical forces into biochemical or electrical signals that initiate structural and functional remodeling in cells and tissues. The heart is a kinetic organ whose form changes considerably during development and disease, requiring cardiac myocytes to be mechanically durable and capable of fusing a variety of environmental signals on different time scales. During physiological growth, myocytes adaptively remodel to mechanical loads. Pathological stimuli can induce maladaptive remodeling. In both of these conditions, the cytoskeleton plays a pivotal role in both sensing mechanical stress and mediating structural remodeling and functional responses within the myocyte.
Collapse
|
19
|
Sanger JM, Wang J, Gleason LM, Chowrashi P, Dube DK, Mittal B, Zhukareva V, Sanger JW. Arg/Abl-binding protein, a Z-body and Z-band protein, binds sarcomeric, costameric, and signaling molecules. Cytoskeleton (Hoboken) 2010; 67:808-23. [PMID: 20886612 DOI: 10.1002/cm.20490] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2010] [Revised: 09/22/2010] [Accepted: 09/23/2010] [Indexed: 12/14/2022]
Abstract
ArgBP2 (Arg/Abl-Binding Protein) is expressed at high levels in the heart and is localized in the Z-bands of mature myofibrils. ArgBP2 is a member of a small family of proteins that also includes vinexin and CAP (c-Cbl-associated protein), all characterized by having one sorbin homology (SOHO) domain and three C-terminal SH3 domains. Antibodies directed against ArgBP2 also react with the Z-bodies of myofibril precursors: premyofibrils and nascent myofibrils. Expression in cardiomyocytes of plasmids encoding Yellow Fluorescent Protein (YFP) fused to either full length ArgBP2, the SOHO, mid-ArgBP or the SH3 domains of ArgBP2 led to Z-band targeting of the fusion proteins, whereas an N-terminal fragment lacking these domains did not target to Z-bands. Although ArgBP2 is not found in skeletal muscle cells, YFP-ArgBP2 did target to Z-bodies and Z-bands in cultured myotubes. GST-ArgBP2-SH3 bound actin, α-actinin and vinculin proteins in blot overlays, cosedimentation assays, and EM negative staining techniques. Over-expression of ArgBP2 and ArgBP2-SH3 domains, but not YFP alone, led to loss of myofibrils in cardiomyocytes. Fluorescence recovery after photobleaching was used to measure the rapid dynamics of both the full length and some truncated versions of ArgBP2. Our results indicate that ArgBP2 may play an important role in the assembly and maintenance of myofibrils in cardiomyocytes.
Collapse
Affiliation(s)
- Jean M Sanger
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York 13210, USA.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Balasubramanian S, Mani SK, Kasiganesan H, Baicu CC, Kuppuswamy D. Hypertrophic stimulation increases beta-actin dynamics in adult feline cardiomyocytes. PLoS One 2010; 5:e11470. [PMID: 20635003 PMCID: PMC2902504 DOI: 10.1371/journal.pone.0011470] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 06/15/2010] [Indexed: 02/04/2023] Open
Abstract
The myocardium responds to hemodynamic stress through cellular growth and organ hypertrophy. The impact of cytoskeletal elements on this process, however, is not fully understood. While α-actin in cardiomyocytes governs muscle contraction in combination with the myosin motor, the exact role of β-actin has not been established. We hypothesized that in adult cardiomyocytes, as in non-myocytes, β-actin can facilitate cytoskeletal rearrangement within cytoskeletal structures such as Z-discs. Using a feline right ventricular pressure overload (RVPO) model, we measured the level and distribution of β-actin in normal and pressure overloaded myocardium. Resulting data demonstrated enriched levels of β-actin and enhanced translocation to the Triton-insoluble cytoskeletal and membrane skeletal complexes. In addition, RVPO in vivo and in vitro hypertrophic stimulation with endothelin (ET) or insulin in isolated adult cardiomyocytes enhanced the content of polymerized fraction (F-actin) of β-actin. To determine the localization and dynamics of β-actin, we adenovirally expressed GFP-tagged β-actin in isolated adult cardiomyocytes. The ectopically expressed β-actin-GFP localized to the Z-discs, costameres, and cell termini. Fluorescence recovery after photobleaching (FRAP) measurements of β-actin dynamics revealed that β-actin at the Z-discs is constantly being exchanged with β-actin from cytoplasmic pools and that this exchange is faster upon hypertrophic stimulation with ET or insulin. In addition, in electrically stimulated isolated adult cardiomyocytes, while β-actin overexpression improved cardiomyocyte contractility, immunoneutralization of β-actin resulted in a reduced contractility suggesting that β-actin could be important for the contractile function of adult cardiomyocytes. These studies demonstrate the presence and dynamics of β-actin in the adult cardiomyocyte and reinforce its usefulness in measuring cardiac cytoskeletal rearrangement during hypertrophic stimulation.
Collapse
Affiliation(s)
- Sundaravadivel Balasubramanian
- Cardiology Division, Department of Medicine, Gazes Cardiac Research Institute, Medical University of South Carolina, Charleston, South Carolina, United States of America.
| | | | | | | | | |
Collapse
|
21
|
Zhang ZQ, Bish LT, Holtzer H, Sweeney HL. Sarcomeric-alpha-actinin defective in vinculin-binding causes Z-line expansion and nemaline-like body formation in cultured chick myotubes. Exp Cell Res 2009; 315:748-59. [DOI: 10.1016/j.yexcr.2008.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2008] [Revised: 11/20/2008] [Accepted: 12/15/2008] [Indexed: 01/29/2023]
|
22
|
Sparrow JC, Schöck F. The initial steps of myofibril assembly: integrins pave the way. Nat Rev Mol Cell Biol 2009; 10:293-8. [PMID: 19190670 DOI: 10.1038/nrm2634] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Myofibril assembly results in a regular array of identical sarcomeres in striated muscle. Sarcomere structure is conserved across the animal kingdom, which implies that the mechanisms of myofibril assembly are also likely to be conserved. Recent advances from model genetic systems and insights from stress fibre cell biology have shed light on the mechanisms that set sarcomere spacing and the initial assembly of sarcomere arrays. We propose a model of integrin-dependent cell-matrix adhesion as the starting point for myofibrillogenesis.
Collapse
Affiliation(s)
- John C Sparrow
- Department of Biology, University of York, York, YO10 5YW, UK.
| | | |
Collapse
|
23
|
Toward physiological conditions for cell analyses: forces of heart muscle cells suspended between elastic micropillars. Biophys J 2007; 94:1854-66. [PMID: 17981895 DOI: 10.1529/biophysj.107.115766] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Almost each mammalian cell permanently applies forces to its environment. These forces are essential for many vital processes such as tissue formation or cell movement. In turn, the environmental conditions of cells strongly affect force production. Here we report on the development of an array of elastomeric micropillars as cellular environment. Within these micropillar arrays, we cultivated rat heart muscle cells (cardiac myocytes). For lattice constants between 20 and 30 mum, cells strongly preferred spanning between the elastic micropillars over adhering to the underlying flat substrate. In addition, the architectures of the cytoskeleton and of protein complexes formed for adhesion were strongly dependent on the environment of the cell. On flat parts of the substrates, we observed prominent stress fibers and focal adhesion sites. In contrast, cells suspended between micropillars exhibited well organized myofibers and costameric adhesions at the locations of Z-bands. These observations argue for close-to-nature environmental conditions within micropillar arrays. Resting as well as contraction forces of myocytes resulted in measurable pillar bending. Using an approximate theoretical treatment of elastically founded micropillars, we calculated average cell forces of 140 nN in the relaxed and 400 nN in the contracted state.
Collapse
|
24
|
Abstract
The assembly of sarcomeric proteins into the highly organized structure of the sarcomere is an ordered and complex process involving an array of structural and associated proteins. The sarcomere has shown itself to be considerably more complex than ever envisaged and may be considered one of the most complex macromolecular assemblies in biology. Studies over the last decade have helped to put a new face on the sarcomere, and, as such, the sarcomere is being redefined as a dynamic network of proteins capable of generating force and signalling with other cellular compartments and metabolic enzymes capable of controlling many facets of striated myocyte biology.
Collapse
Affiliation(s)
- Samuel Y Boateng
- The Center for Cardiovascular Research, Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
25
|
Dedkov EI, Stadnikov AA, Russell MW, Borisov AB. Formation of leptofibrils is associated with remodelling of muscle cells and myofibrillogenesis in the border zone of myocardial infarction. Micron 2006; 38:659-67. [PMID: 17015018 DOI: 10.1016/j.micron.2006.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2006] [Accepted: 08/31/2006] [Indexed: 10/24/2022]
Abstract
Leptofibrils, or leptomeres, remain the least studied cytoskeletal structures in muscle cells, and their function and mechanism of assembly are still poorly understood. Our ultrastructural study of the surviving cardiac myocytes located in the perinecrotic border zone of the infarcted left ventricle in rats revealed intense formation of leptofibrils and leptofibrillar clusters during 4-15 days following experimental myocardial infarction. In the perinecrotic myocytes, leptofibrils developed predominantly in the subsarcolemmal areas, near disassembled intercalated discs and at the sites of intense myofibrillogenesis in the peripheral zones of the sarcoplasm. We found that the development of these structures occurred before or at the time of assembly of myofibrils. In our material, leptofibrils consisted of longitudinally oriented filamentous bundles inserted in electron dense Z-band-like material and periodically crossed by 3-8 bands of this material with the period of cross-striation of 120-210 nm. The presence of leptofibrils in growing cytoplasmic processes and ruffles developing in the border zone in the areas of lost intercellular contacts indicates their formation de novo during post-infarction period. We observed four major morphological types of localization of these structures: (1) direct contact of one end of leptofibrils with Z bands of nascent, mature or disassembling myofibrils; (2) direct contact with the sarcolemma: (a) multifocal attachment of leptofibrils to the sarcolemma through the lateral surfaces of their minute Z band-like structures; (b) attachment of one or both ends of leptofibrils to the sarcolemma without contacts or in contact with myofibrils; (3) attachment of leptofibrils to subsarcolemmal accumulations of electron dense Z-band material in newly formed fasciae adherentes of the remodeled intercalated disks; (4) clustering and contacts of leptofibrils with one another predominantly at the level of their Z bands. Interestingly, most leptofibrils of all four types were topographically associated with the system of T-tubules, the sarcoplasmic reticulum and subsarcolemmal vesicles. Serial sections through the areas containing leptofibrils indicate their spindle-like or nearly cylindrical shape. Thus, we found that leptofibrils assemble in terminally differentiated cardiac myocytes following destabilization of their differentiated state and partial dedifferentiation induced by myocardial infarction. The results of this study demonstrate that formation of leptofibrils, earlier described mainly in the developing and malignant muscle, is temporally associated with adaptive structural remodelling and the activation of myofibrillogenesis in functionally overloaded cardiac myocytes of adult animals. Our findings suggest that re-expression of some structural characteristics of the embryonic muscle appear to represent one of the mechanisms that underlie adaptive plasticity of the myocardium following injury and under conditions of hyperfunction.
Collapse
Affiliation(s)
- Eduard I Dedkov
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA
| | | | | | | |
Collapse
|
26
|
Ohtsuka Y, Okamura Y. Voltage-dependent calcium influx mediates maturation of myofibril arrangement in ascidian larval muscle. Dev Biol 2006; 301:361-73. [PMID: 16962575 DOI: 10.1016/j.ydbio.2006.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Revised: 07/28/2006] [Accepted: 08/04/2006] [Indexed: 11/18/2022]
Abstract
Calcium signaling is important for multiple events during embryonic development. However, roles of calcium influx during embryogenesis have not been fully understood since routes of calcium influx are often redundant. To define roles of voltage-gated calcium channel (Cav) during embryogenesis, we have isolated an ascidian Cav beta subunit gene (TuCavbeta) and performed gene knockdown using the morpholino antisense oligonucleotide (MO). The suppression of Cav activity by TuCavbetaMO remarkably perturbed gastrulation and tail elongation. Further, larvae with normal morphology also failed to exhibit motility. Phalloidin-staining showed that arrangement of myofibrils was uncoordinated in muscle cells of TuCavbetaMO-injected larvae with normal tail. To further understand the roles of Cav activity in myofibrillogenesis, we tested pharmacological inhibitions with ryanodine, curare, and N-benzyl-p-toluensulphonamide (BTS). The treatment with ryanodine, an intracellular calcium release blocker, did not significantly affect the motility and establishment of the myofibril orientation. However, treatment with curare, an acetylcholine receptor blocker, and BTS, an actomyosin ATPase specific inhibitor, led to abnormal motility and irregular orientation of myofibrils that was similar to those of TuCavbetaMO-injected larvae. Our results suggest that contractile activation regulated by voltage-dependent calcium influx but not by intracellular calcium release is required for proper arrangement of myofibrils.
Collapse
Affiliation(s)
- Yukio Ohtsuka
- Neuroscience Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central 6, Higashi 1-1-1, Tsukuba, Ibaraki 305-8566, Japan.
| | | |
Collapse
|
27
|
Quach NL, Rando TA. Focal adhesion kinase is essential for costamerogenesis in cultured skeletal muscle cells. Dev Biol 2006; 293:38-52. [PMID: 16533505 DOI: 10.1016/j.ydbio.2005.12.040] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2005] [Revised: 12/12/2005] [Accepted: 12/14/2005] [Indexed: 02/03/2023]
Abstract
A central question in muscle biology is how costameres are formed and become aligned with underlying myofibrils in mature tissues. Costameres are composed of focal adhesion proteins, including vinculin and paxillin, and anchor myofibril Z-bands to the sarcolemma. In the present study, we investigated the process of costamere formation ("costamerogenesis") in differentiating primary mouse myoblasts. Using vinculin and paxillin as costameric markers, we found that two additional focal adhesion components, alpha5beta1 integrin and focal adhesion kinase (FAK), are associated with costameres. We have characterized costamerogenesis as occurring in three distinct stages based on the organizational pattern of these costameric proteins. We show that both costamerogenesis and myofibrillogenesis are initiated at sites of membrane contacts with the extracellular matrix and that their maturation is tightly coupled. To test the importance of FAK signaling in these processes, we analyzed cells expressing a dominant negative form of FAK (dnFAK). When cells expressing dnFAK were induced to differentiate, both costamerogenesis and myofibrillogenesis were disrupted although the expression of constituent proteins was not inhibited. Likewise, inhibiting FAK activity by reducing FAK levels using an siRNA approach also resulted in an inhibition of costamerogenesis and myofibrillogenesis. The relationship between costamere and myofibril formation was tested further by treating myotube cultures with potassium or tetrodotoxin to block contraction and disrupt myofibril organization. This also resulted in inhibition of costamere maturation. We present a model of costamerogenesis whereby signaling through FAK is essential for both normal costamerogenesis and normal myofibrillogenesis which are tightly coupled during skeletal myogenesis.
Collapse
Affiliation(s)
- Navaline L Quach
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
28
|
Abstract
Mechanotransduction refers to the cellular mechanisms by which load-bearing cells sense physical forces, transduce the forces into biochemical signals, and generate appropriate responses leading to alterations in cellular structure and function. This process affects the beat-to-beat regulation of cardiac performance but also affects the proliferation, differentiation, growth, and survival of the cellular components that comprise the human myocardium. This review focuses on the experimental evidence indicating that the costamere and its structurally related structure the focal adhesion complex are critical cytoskeletal elements involved in cardiomyocyte mechanotransduction. Biochemical signals originating from the extracellular matrix-integrin-costameric protein complex share many common features with those signals generated by growth factor receptors. The roles of key regulatory kinases and other muscle-specific proteins involved in mechanotransduction and growth factor signaling are discussed, and issues requiring further study in this field are outlined.
Collapse
Affiliation(s)
- Allen M Samarel
- Cardiovascular Institute, Loyola Univ. Medical Center, Bldg. 110, Rm. 5222, 2160 South First Ave., Maywood, IL 60153, USA.
| |
Collapse
|
29
|
Watzka SBC, Steiner M, Samorapoompichit P, Gross K, Coles JG, Wolner E, Weigel G. Establishment of Vessel-Like Structures in Long-Term Three-Dimensional Tissue Culture of Myocardium: An Electron Microscopy Study. ACTA ACUST UNITED AC 2004; 10:1684-94. [PMID: 15684677 DOI: 10.1089/ten.2004.10.1684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
To assess whether long-term three-dimensional (3D) tissue culture of myocardium enables the in vitro establishment of vessel-like structures, myocardial tissue from newborn mice was incubated under conditions of 3D culture for at least 3 weeks, and studied by phase-contrast microscopy, conventional histology, immunohistochemistry, and electron microscopy. During 3 weeks of culture, a mean 24.35 +/- 3.74% of all aggregates contracted spontaneously. The contracting aggregates displayed a tissue-like architecture with small basal and apical zones, and a large central zone. The basal and apical zone consisted of immature mesenchymal cells. The underlying shell of the aggregate contained many cardiomyocytes. Vessel-like structures were found concentrated within the aggregates. Immunohistochemistry showed that up to 15% of the cells in the central zone of the aggregate were positive for the endothelial-specific BS-I lectin. Vessel-like structures were formed by cells, which often showed intracytoplasmatic lumena. Surrounding the neocapillaries, structures of a rudimentary basal membrane could be detected. A 3D culture of myocardial tissue permits the establishment of a rudimentary capillary network within the tissue aggregates, which presumably guarantees a sufficient tissue perfusion up to a maximum aggregate diameter of approximately 900 microm.
Collapse
Affiliation(s)
- Stefan B C Watzka
- Division of Cardiothoracic Surgery, Vienna General Hospital, Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
30
|
Dudnakova TV, Lakomkin VL, Tsyplenkova VG, Shekhonin BV, Shirinsky VP, Kapelko VI. Alterations in myocardial ultrastructure and protein expression after a single injection of isoproterenol. Mol Cell Biochem 2004; 252:173-81. [PMID: 14577591 DOI: 10.1023/a:1025579624695] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Immunochemical and electron microscopic characterization of rat myocardium was conducted 2 h and 3 weeks after a single injection of isoproterenol in rats. The relative content of several myospecific proteins (KRP--kinase-related protein, desmin), cytoskeletal proteins (tubulin, vinculin, myosin light chain kinase--MLCK) and extracellular matrix protein fibronectin was determined by immunoblotting. Two hours after injection of 50 mg/kg isoproterenol a destruction of some cardiomyocytes, contracture of myofibrils and mild edema of intercellular space was observed. The content of all the studied proteins except KRP decreased below control levels. This situation sustained 3 weeks after injection and paralleled alterations in cardiomyocyte ultrastructure. Areas of myofibrillar contracture and lysis were noted, glycogen granules were sparse; mitochondria contained arrow-like inclusions that are characteristic for calcium overload, also huge mitochondria contacting each other by specialized intermitochondrial contacts were detected. Clumps of unripe elastic fibers in enlarged intercellular space were combined with increased deposition of collagens type I and III forming areas of fibrosis. The smaller dosage of isoproterenol (10 mg/kg) rendered no significant damage in the acute postinjection period but 3 weeks later it induced the thickening of extracellular matrix around cardiac cells and the increase in KRP and tubulin content by 26 and 32%, correspondingly. MLCK levels remained depressed throughout the experiment. The rise in KRP expression was also observed after the addition of isoproterenol to cultured chicken embryo cardiomyocytes. Obtained results indicate that even a single injection of isoproterenol creates long lasting structural alterations in cardiac muscle accompanied by the increased expression of extracellular matrix proteins and several sarcoplasmic proteins apparently involved in hypertrophic response of cardiomyocytes.
Collapse
Affiliation(s)
- Tatyana V Dudnakova
- Institute of Experimental, Russian Cardiological Scientific and Productive Complex, Moscow, Russia
| | | | | | | | | | | |
Collapse
|
31
|
Wakimoto K, Fujimura H, Iwamoto T, Oka T, Kobayashi K, Kita S, Kudoh S, Kuro-o M, Nabeshima YI, Shigekawa M, Imai Y, Komuro I. Na+/Ca2+ exchanger-deficient mice have disorganized myofibrils and swollen mitochondria in cardiomyocytes. Comp Biochem Physiol B Biochem Mol Biol 2003; 135:9-15. [PMID: 12781968 DOI: 10.1016/s1096-4959(03)00057-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Na(+)/Ca(2+) exchanger (NCX1) plays a key role in maintaining Ca(2+) homeostasis in cardiomyocytes. Disruption of Ncx1 gene in mice results in embryonic lethality between embryonic day 9 and 10, with the mice lacking spontaneous heartbeats. We examined the mechanism of lack of heartbeats in Ncx1-deficient mice. Ultrastructual analysis demonstrated that Ncx1-deficient mice showed severe disorganization of myofibrils, a lack of Z-lines and swelling of mitochondria in cardiomyocytes. However, the expressions of cardiac-specific genes including transcription factor genes and contractile protein genes were not changed in Ncx1-deficient mice. Abnormal Ca(2+) handling itself or the lack of heartbeats due to the inactivation of Ncx1 gene may cause the disorganization of myofibrillogenesis. Although NCX1 protein levels were decreased in heterozygous mice, there were no changes in NCX2 and NCX3 protein levels between wild type and heterozygous mice.
Collapse
Affiliation(s)
- Koji Wakimoto
- Discovery Research Laboratory, Tanabe Seiyaku Co., Ltd, 3-16-89 Kashima, Yodogawa-ku, Osaka 532-8505, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Juhasz O, Zhu Y, Garg R, Anisimov SV, Boheler KR. Analysis of altered genomic expression profiles in the senescent and diseased myocardium using cDNA microarrays. Eur J Heart Fail 2002; 4:687-97. [PMID: 12453538 DOI: 10.1016/s1388-9842(02)00169-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cardiac function deteriorates with aging or disease. Short term, any changes in heart function may be beneficial, but long term the alterations are often detrimental. At a molecular level, functional adaptations involve quantitative and qualitative changes in gene expression. Analysis of all the RNA transcripts present in a cell's population (transcriptome) offers unprecedented opportunities to map these transitions. Microarrays (chips), capable of evaluating thousands of transcripts in one assay, are ideal for transcriptome analyses. Gene expression profiling provides information about the dynamics of total genome expression in response to environmental changes and may point to candidate genes responsible for the cascade of events that result in disease or are a consequence of aging. The aim of this review is to describe how comparisons of cellular transcriptomes by cDNA array based techniques provide information about the dynamics of total gene expression, and how the results can be applied to the study of cardiovascular disease and aging.
Collapse
Affiliation(s)
- Ondrej Juhasz
- Laboratory of Cardiovascular Science, Gerontology Research Center, National Institute on Aging, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
33
|
Sinn HW, Balsamo J, Lilien J, Lin JJC. Localization of the novel Xin protein to the adherens junction complex in cardiac and skeletal muscle during development. Dev Dyn 2002; 225:1-13. [PMID: 12203715 DOI: 10.1002/dvdy.10131] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Previously, we demonstrated that chick embryos treated with antisense oligonucleotides against a striated muscle-specific Xin exhibit abnormal cardiac morphogenesis (Wang et al. [1999] Development 126:1281-1294); therefore, we surmised a role for Xin in cardiac development. Herein, we examine the developmental expression of Xin through immunofluorescent staining of whole-mount mouse embryos and frozen heart sections. Xin expression is first observed within the heart tube of embryonic day 8.0 (E8.0) mice, exhibiting a peripheral localization within the cardiomyocytes. Colocalization of Xin with both beta-catenin and N-cadherin is observed throughout embryogenesis and into adulthood. Additionally, Xin is found associated with beta-catenin within the N-cadherin complex in embryonic chick hearts by coimmunoprecipitation. Xin is detected earlier than vinculin in the developing heart and colocalizes with vinculin at the intercalated disc but not at the sarcolemma within embryonic and postnatal hearts. At E10.0, Xin is also detected in the developing somites and later in the myotendon junction of skeletal muscle but not within the costameric regions of muscle. In cultured C2C12 myotubes, the Xin protein is found in many speckled and filamentous structures, coincident with tropomyosin in the stress fibers. Additionally, Xin is enriched in the regions of cell-cell contacts. These data demonstrate that Xin is one of the components at the adherens junction of cardiac muscle, and its counterpart in skeletal muscle, the myotendon junction. Furthermore, temporal and spatial expressions of Xin in relation to intercalated disc proteins and thin filament proteins suggest roles for Xin in the formation of cell-cell contacts and possibly in myofibrillogenesis.
Collapse
Affiliation(s)
- Haley W Sinn
- Department of Biological Sciences, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
34
|
Heidkamp MC, Bayer AL, Kalina JA, Eble DM, Samarel AM. GFP-FRNK disrupts focal adhesions and induces anoikis in neonatal rat ventricular myocytes. Circ Res 2002; 90:1282-9. [PMID: 12089066 DOI: 10.1161/01.res.0000023201.41774.ea] [Citation(s) in RCA: 104] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Focal adhesion kinase (FAK) is a nonreceptor protein tyrosine kinase involved in adhesion-dependent signal transduction. FAK is highly expressed in cultured neonatal rat ventricular myocytes (NRVMs) and undergoes tyrosine autophosphorylation in response to cell adhesion, stretch, and growth factor stimulation. We previously showed that inhibition of FAK phosphorylation by adenovirally mediated overexpression of FRNK (the autonomously expressed C-terminal domain of FAK) prevented endothelin-1 (ET)-induced NRVM hypertrophy. One question raised by these studies was whether FRNK localized to focal adhesions and displaced FAK from sites required for downstream signaling. Therefore, we constructed a replication-defective adenovirus encoding a GFP-FRNK fusion protein (Adv-GFP-FRNK) and examined its effects on NRVM cytoarchitecture and signaling. Uninfected NRVMs contained small amounts of endogenous FRNK. NRVMs infected with Adv-GFP-FRNK expressed much larger amounts of a 66-/68-kDa protein that localized to costameres and focal adhesions. GFP-FRNK overexpression suppressed basal and ET-induced FAK phosphorylation and also inhibited ET-induced phosphorylation of PYK2, the other member of the FAK family of nonreceptor protein tyrosine kinases. In contrast, GFP-FRNK overexpression did not prevent ET-induced ERK, JNK, or p70S6K phosphorylation. Furthermore, GFP-FRNK resulted in the loss of detectable FAK and paxillin in focal adhesions, which was accompanied by reduced levels of total paxillin and, ultimately, cell detachment and apoptosis. We conclude that FRNK functions as a dominant-negative inhibitor of adhesion-dependent signaling by displacing FAK from focal adhesions and interfering with the anchorage of NRVMs that is necessary for cell survival, a process known as anoikis.
Collapse
Affiliation(s)
- Maria C Heidkamp
- Cardiovascular Institute, Loyola University Chicago, Stritch School of Medicine, Maywood, Ill, USA.
| | | | | | | | | |
Collapse
|
35
|
Chlopčíková Š, Psotová J, Miketová P. NEONATAL RAT CARDIOMYOCYTES - A MODEL FOR THE STUDY OF MORPHOLOGICAL, BIOCHEMICAL AND ELECTROPHYSIOLOGICAL CHARACTERISTICS OF THE HEART. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2001. [DOI: 10.5507/bp.2001.011] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
36
|
Kovacic-Milivojević B, Roediger F, Almeida EA, Damsky CH, Gardner DG, Ilić D. Focal adhesion kinase and p130Cas mediate both sarcomeric organization and activation of genes associated with cardiac myocyte hypertrophy. Mol Biol Cell 2001; 12:2290-307. [PMID: 11514617 PMCID: PMC58595 DOI: 10.1091/mbc.12.8.2290] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Hypertrophic terminally differentiated cardiac myocytes show increased sarcomeric organization and altered gene expression. Previously, we established a role for the nonreceptor tyrosine kinase Src in signaling cardiac myocyte hypertrophy. Here we report evidence that p130Cas (Cas) and focal adhesion kinase (FAK) regulate this process. In neonatal cardiac myocytes, tyrosine phosphorylation of Cas and FAK increased upon endothelin (ET) stimulation. FAK, Cas, and paxillin were localized in sarcomeric Z-lines, suggesting that the Z-line is an important signaling locus in these cells. Cas, alone or in cooperation with Src, modulated basal and ET-stimulated atrial natriuretic peptide (ANP) gene promoter activity, a marker of cardiac hypertrophy. Expression of the C-terminal focal adhesion-targeting domain of FAK interfered with localization of endogenous FAK to Z-lines. Expression of the Cas-binding proline-rich region 1 of FAK hindered association of Cas with FAK and impaired the structural stability of sarcomeres. Collectively, these results suggest that interaction of Cas with FAK, together with their localization to Z-lines, is critical to assembly of sarcomeric units in cardiac myocytes in culture. Moreover, expression of the focal adhesion-targeting and/or the Cas-binding proline-rich regions of FAK inhibited ANP promoter activity and suppressed ET-induced ANP and brain natriuretic peptide gene expression. In summary, assembly of signaling complexes that include the focal adhesion proteins Cas, FAK, and paxillin at Z-lines in the cardiac myocyte may regulate, either directly or indirectly, both cytoskeletal organization and gene expression associated with cardiac myocyte hypertrophy.
Collapse
Affiliation(s)
- B Kovacic-Milivojević
- Metabolic Research Unit, University of California San Francisco, San Francisco, California 94143-0540, USA
| | | | | | | | | | | |
Collapse
|
37
|
McElhinny AS, Labeit S, Gregorio CC. Probing the functional roles of titin ligands in cardiac myofibril assembly and maintenance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2001; 481:67-86; discussion 86-8. [PMID: 10987067 DOI: 10.1007/978-1-4615-4267-4_5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sarcomeres of cardiac muscle are comprised of numerous proteins organized in an elegantly precise order. The exact mechanism of how these proteins are assembled into myofibrils during heart development is not yet understood, although existing in vitro and in vivo model systems have provided great insight into this complex process. It has been proposed by several groups that the giant elastic protein titin acts as a "molecular template" to orchestrate sarcomeric organization during myofibrillogenesis. Titin's highly modular structure, composed of both repeating and unique domains that interact with a wide spectrum of contractile and regulatory ligands, supports this hypothesis. Recent functional studies have provided clues to the physiological significance of the interaction of titin with several titin-binding proteins in the context of live cardiac cells. Improved models of cardiac myofibril assembly, along with the application of powerful functional studies in live cells, as well as the characterization of additional titin ligands, is likely to reveal surprising new functions for the titin third filament system.
Collapse
Affiliation(s)
- A S McElhinny
- Department of Cell Biology and Anatomy, University of Arizona, Tucson, USA
| | | | | |
Collapse
|
38
|
LoRusso SM, Rhee D, Sanger JM, Sanger JW. Premyofibrils in spreading adult cardiomyocytes in tissue culture: evidence for reexpression of the embryonic program for myofibrillogenesis in adult cells. CELL MOTILITY AND THE CYTOSKELETON 2000; 37:183-98. [PMID: 9227849 DOI: 10.1002/(sici)1097-0169(1997)37:3<183::aid-cm1>3.0.co;2-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Do adult cardiomyocytes use the same pathways hypothesized for the formation of myofibrils in embryonic cardiomyocytes in tissue culture. [Rhee, et al., Cell Motil. Cytoskeleton 28:1-24, 1994]? Premyofibrils in embryonic cardiomyocytes are composed of short sarcomeric units of alpha-actinin (Z-bodies) and actin filaments held together by short nonmuscle myosin IIB filaments. Premyofibrils are believed to be transformed into nascent myofibrils by their capture of muscle-specific myosin II filaments aligned in aperiodic arrays. Nascent myofibrils are thought to transform into mature myofibrils by the loss of nonmuscle myosin IIB, the fusion of the Z-bodies into Z-bands, and the periodic alignment of muscle myosin II filaments into A-bands. Freshly isolated cat and rat adult cardiomyocytes placed in tissue culture lack premyofibrils and nascent myofibrils. Adult cardiomyocytes spreading in culture reinitiate the synthesis of nonmuscle myosin IIB. Moreover, patterns similar to the proposed embryonic myofibrillar program first detected in spreading chick embryonic hearts were also detected in these spreading adult mammalian cardiomyocytes. The isolated adult cardiomyocytes begin to spread after 1 day in culture by sending out lamellipodia. When these cells are injected with fluorescently labeled alpha-actinin, linear arrays of short spacings of beaded alpha-actinin bodies are detected in the spreading edges of the adult cardiomyocytes. These dense bodies (Z-bodies) stain positively for the same sarcomeric-specific isoform of alpha-actinin that is in the Z-bands of mature sarcomeres. These linear arrays of alpha-actinin-containing Z-bodies have other characteristics of premyofibrils and are detected only in the spreading regions of the cells. Thus, these premyofibrils at the edges of the spreading adult cardiomyocytes stain positively for nonmuscle myosin IIB but negatively for muscle-specific myosin II. Initially, no vinculin is associated with any parts of the premyofibrils in the spreading regions of the early spreading cardiomyocytes. However, later, vinculin is found to be associated with the ends of the premyofibrils. Fibers that stain solidly for muscle-specific myosin II (i.e., nascent myofibrils) are localized between the peripheral premyofibrils and the centrally positioned, mature myofibrils. It is suggested that the puzzling ability of cardiomyocytes in hypertrophic hearts to reinitiate the synthesis of fetal sarcomeric proteins may be related to the reinitiation of the embryonic premyofibril program for myofibrillogenesis.
Collapse
Affiliation(s)
- S M LoRusso
- Department of Cell and Developmental Biology, Pennsylvania Muscle Institute, University of Pennsylvania School of Medicine, Philadelphia 19104-6058, USA
| | | | | | | |
Collapse
|
39
|
Eble DM, Strait JB, Govindarajan G, Lou J, Byron KL, Samarel AM. Endothelin-induced cardiac myocyte hypertrophy: role for focal adhesion kinase. Am J Physiol Heart Circ Physiol 2000; 278:H1695-707. [PMID: 10775151 DOI: 10.1152/ajpheart.2000.278.5.h1695] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelin-1 (ET) produces neonatal rat ventricular myocyte (NRVM) hypertrophy and activates focal adhesion kinase (FAK) in other cell types. In the present study, we examined whether ET activated FAK in NRVM and whether FAK was necessary and/or sufficient for ET-induced NRVM hypertrophy. Chronic ET-1 stimulation (100 nM, 48 h) increased protein-to-DNA and myosin heavy chain (MHC)-to-DNA ratios and stimulated the assembly of newly synthesized MHC into sarcomeres. ET-1 also induced the assembly of focal adhesions and costameres, as evidenced by increased phosphotyrosine, FAK, and paxillin immunostaining. Acutely, ET treatment rapidly increased tyrosine phosphorylation of FAK and paxillin. FAK was also activated by phorbol 12-myristate 13-acetate (2 microM, 5 min). Pretreatment with chelerythrine (5 microM) or rottlerin (10 microM) completely blocked ET-induced FAK phosphorylation, indicating that protein kinase C activation was upstream of ET-induced FAK activation. In contrast, ET-induced FAK activation was not affected by blocking calcium influx via L-type voltage-gated calcium channels. Adenoviruses (Adv) containing FAK and FAK-related nonkinase (FRNK) were used to specifically define the role of FAK in ET-induced hypertrophy. ET stimulation failed to increase total protein-to-DNA or MHC-to-DNA ratios or to stimulate sarcomeric assembly in myocytes infected with Adv-FRNK. However, Adv-FAK alone did not increase total protein-to-DNA or MHC-to-DNA ratios and failed to increase the number or size of myofibrils as evidenced by double immunofluorescence labeling for MHC and FAK. Thus, although FAK is necessary for ET-induced NRVM hypertrophy, other ET-generated signals are also required to elicit the hypertrophic phenotype.
Collapse
Affiliation(s)
- D M Eble
- Cardiovascular Institute and Departments of Medicine and Physiology, Loyola University Chicago, Maywood, Illinois 60153, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Eizema K, van Heugten HA, Bezstarosti K, van Setten MC, Lamers JM. Endothelin-1 responsiveness of a 1.4 kb phospholamban promoter fragment in rat cardiomyocytes transfected by the gene gun. J Mol Cell Cardiol 2000; 32:311-21. [PMID: 10722806 DOI: 10.1006/jmcc.1999.1076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transcriptional regulation of an isolated rat phospholamban (PL) promoter fragment in rat cardiomyocytes was analyzed by applying a new method to reach substantially higher transfection efficiencies: gene gun biolistics. The gene gun transfection method was optimized for application to primary cultures of rat neonatal cardiomyocytes. Cells, cultured at different densities (0.75-1.50x10(5)cells/cm(2)) in serum-free medium, were transfected with DNA coated gold particles. A transfection efficiency of up to 10% could be achieved (compared to <1% with other methods) by the gene gun as checked using a RSV- beta-Gal construct. Cardiomyocytes were stimulated by endothelin-1 (ET-1) (10(-8)M) to induce hypertrophy, thereby yielding the characteristic changes in gene expression (upregulation of Atrial Natriuretic Factor (ANF) and downregulation of PL). The basal activity of an ANF promoter fragment (increasing from the lowest to highest density 2.6-fold) and its ET-1 inducibility (only significant upregulation of 2.6-fold, at lowest density) appeared to be dependent on the plating density of the cardiomyocytes. A PL promoter fragment was isolated, sequenced and 1.4 kb was subcloned in a luciferase reporter vector. The basal activity of the PL promoter fragment was not dependent on the plating density. ET-1 did not downregulate the PL promoter, rather a significant upregulation (1.4-fold) was found at the highest plating density. In conclusion, plating density of the cardiomyocytes can influence promoter activity as shown with an ANF promoter fragment. A newly isolated and sequenced rat PL promoter fragment did not direct gene expression as expected on basis of downregulation of the PL gene by ET-1 observed in this model.
Collapse
Affiliation(s)
- K Eizema
- Department of Biochemistry, Erasmus University Rotterdam, Rotterdam, 3000 DR, Netherlands
| | | | | | | | | |
Collapse
|
41
|
Eble DM, Qi M, Strait J, Samarel AM. Contraction-Dependent Hypertrophy of Neonatal Rat Ventricular Myocytes: Potential Role for Focal Adhesion Kinase. PROGRESS IN EXPERIMENTAL CARDIOLOGY 2000. [DOI: 10.1007/978-1-4615-4423-4_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
42
|
Horackova M, Morash B, Byczko Z. Altered transarcolemmal Ca transport modifies the myofibrillar ultrastructure and protein metabolism in cultured adult ventricular cardiomyocytes. Mol Cell Biochem 2000; 204:21-33. [PMID: 10718621 DOI: 10.1023/a:1007080828602] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The present study was designed to investigate how prolonged (24-72 h) exposure to modifiers of Ca transarcolemmal transport affects the myofibrillar structure, protein turnover and content of myofibrillar proteins in adult guinea pig cardiomyocytes maintained beating synchronously in long-term cultures. First we established the functional responses (the contractile activity and [Ca]i transients) of the cultured myocytes to acute exposures to several drugs used in this study. The ultrastructural characteristics of these cultures under the various treatments were determined using immunohistochemistry and confocal scanning laser microscopy, and their biochemical properties were evaluated using analysis of total cellular protein content, myofibrillar protein content and SDS-PAGE electrophoretic examination. We compared the effects of 24, 36 and 72 h-long exposures to the various specific Ca-flux modifiers. Increased Ca influx via CaL-channel agonist (Bay K 8644) or via the reversed-mode of the Na/Ca exchanger (veratrine) did not alter the myofibrillar structure or the specific protein profiles or proteosynthesis. However, when cytosolic Ca was increased by three different types of inhibitors of Ca extrusion from the cells via Na/Ca exchange, (Na-free solution, 5 mM NiCl2 and 10(-6) M ouabain), very significant changes in all investigated parameters occurred almost immediately. Twenty-four h-long exposure to Na-free did not affect significantly the total cellular protein (TCP), but the protein synthesis was decreased by 87% and the total myofibrillar protein (TMP) content was decreased by 38%. The myofibrils were heavily fragmented. Similarly, 24 h-long exposure to 5 mM NiCl2 did not affect the TCP, but it reduced protein synthesis by about 90% and decreased the total myofibrillar protein content by 30%. These effects were even more pronounced at 72 h of exposure and they were accompanied with a complete disassembly of myofilaments. Exposure to 10(-6) M ouabain over 72 h resulted in > 80% inhibition of protein synthesis, a 45% decrease in TCP content and a 53% in TMP content. In contrast, 10(-7) M ouabain did not produce any such changes. The changes produced by the Na/Ca-exchange inhibitors were accompanied by only minor changes in DNA content, indicating that the myocytes remained viable. Moreover, these effects were not due to the associated contractile arrest, since exposure to CaL-channel antagonists (5-20 microM nifedipine or 10 microM verapamil) produced only very minor changes in the myofibrillar structure and in protein profiles. Our data demonstrate that short-term (up to 72 h) increased Ca influx or contractile arrest of well-interconnected, spontaneously beating adult cardiomyocytes does not affect their ultrastructural characteristics or their myofibrillar protein turnover greatly, while any situations leading to Ca accumulation (via inhibition of Na/Ca exchange) affect cardiomyocyte function and ultrastructure almost immediately. These data are in sharp contrast to those previously reported from immature, neonatal myocytes.
Collapse
Affiliation(s)
- M Horackova
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University Halifax, Nova Scotia, Canada
| | | | | |
Collapse
|
43
|
Simpson DG, Majeski M, Borg TK, Terracio L. Regulation of cardiac myocyte protein turnover and myofibrillar structure in vitro by specific directions of stretch. Circ Res 1999; 85:e59-69. [PMID: 10559148 DOI: 10.1161/01.res.85.10.e59] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have examined how different degrees (0.5%, 1.0%, 2.5%, 5.0%, and 10.0%) and directions of stretch regulate the turnover and accumulation of contractile proteins in cultured neonatal cardiac myocytes (NCMs). In pulse-chase experiments, stellate-shaped NCMs with random arrays of myofibrils (MFs) exhibited a threshold response to stretch. With respect to unstretched controls, the turnover of the contractile protein pool was suppressed 50% to 100% in stellate NCMs stretched 1.0% to 5.0% and was unaltered in stellate NCMs stretched 0.5% or 10.0%. The posttranslational metabolism of myosin heavy chain (MHC) and actin was regulated in parallel with the total contractile protein pool. The turnover of the cytoplasmic protein pool remained unchanged in response to stretch. NCMs plated onto an aligned matrix of type I collagen expressed an elongated, rod-like cell shape. The MFs of these cells were distributed in parallel with one another along a single unique axis. The tissue-like pattern of organization of these cultures made it possible to assay how specific directions of stretch affected cardiac protein turnover and MF organization. In pulse-chase experiments, stretch in parallel with the MFs did not alter the turnover of the total contractile protein pool, the cytoplasmic protein pool, MHC, or actin. The total cellular concentration of MHC and actin remained constant, and MF alignment was not overtly affected. In contrast, even modest degrees of stretch across the short axis of the MFs suppressed total contractile protein turnover, the turnover of MHC and actin, and promoted the accumulation of these MF subunits. The parallel alignment of MFs deteriorated in myocytes stretched greater than 5%. The characteristic response of aligned myocytes to stretch was not affected by the contractile state of the cells. Isoproterenol (ISO) treatment in concert with stretch in parallel with the MFs modestly accelerated contractile protein turnover. Conversely, contractile protein turnover was suppressed in cells treated with ISO and stretched across the short axis of the MFs. Contractile arrest with nifedipine (NIFED) accelerated total myofibrillar protein turnover. Stretch across the short axis, but not in parallel with the MFs, suppressed protein turnover in cells treated with NIFED. The turnover of the cytosolic proteins remained constant under all conditions assayed. These data suggest that specific directions of stretch may play a crucial role in regulating MF organization and the metabolism of contractile proteins in the cardiac myocyte.
Collapse
Affiliation(s)
- D G Simpson
- Department of Anatomy, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
44
|
Bick RJ, Snuggs MB, Poindexter BJ, Buja LM, Van Winkle WB. Physical, contractile and calcium handling properties of neonatal cardiac myocytes cultured on different matrices. CELL ADHESION AND COMMUNICATION 1998; 6:301-10. [PMID: 9865464 DOI: 10.3109/15419069809010789] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Extracellular matrix components play a vital role in the determination of heart cell growth, development of spontaneous contractile activity and morphologic differentiation. In this work we studied the physical and contractile changes in neonatal rat cardiac myocytes over the first four days of growth on three different extracellular matrices. We compared commercial laminin and fibronectin, plus a fibroblast-derived extracellular matrix, which we have termed cardiogel. Myocytes cultured on cardiogel were characterized by greater cellular area and volume when compared to cells cultured on the other single-component matrices. Spontaneous contractile activity appeared first in the cells grown on cardiogel, sometimes as early as the first day post-plating, in contrast to day three in the cells cultured on laminin. Measurements of cardiac myocyte contractility i.e. percent shortening and time to peak contraction, were made on each of the first four days in each culture. Myocytes cultured on cardiogel developed maximum shortening more rapidly than the other cultures, and an earlier response to electrical pacing. Histochemical staining for myocyte mitochondrial content, revealed that the cardiogel-supported cells exhibited the earliest development of this organelle and, after four days, the greatest abundance. This reflects both a greater cell size, as well as response to increasing energy demands. Due to the increase in volume and contractile activity exhibited by the cardiogel grown myocytes, we employed calcium binding and uptake experiments to determine the comparative cellular capacities for calcium and as an indicator of sarcoplasmic reticulum development. Also whole cell phosphorylation in the presence of low detergent was assayed, to correlate calcium uptake with phosphorylation, in an attempt to examine possible increases in calcium pump number and other phosphorylatable proteins. In agreement with our physical and contractile data, we found that the cells grown on cardiogel showed a greater calcium uptake over the first four days of culture, and increased phosphorylation. However, calcium binding was not dramatically different comparing the three culture matrices. Based on our data, the fibroblast-derived cardiogel is the matrix of choice supporting earliest maturation of neonatal cardiomyocytes, in terms of spontaneous contractions, calcium handling efficiency, cell size and development of a subcellular organelle, the mitochondrion.
Collapse
Affiliation(s)
- R J Bick
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston, UTHMS, 77030, USA.
| | | | | | | | | |
Collapse
|
45
|
Liu Y, Leri A, Li B, Wang X, Cheng W, Kajstura J, Anversa P. Angiotensin II stimulation in vitro induces hypertrophy of normal and postinfarcted ventricular myocytes. Circ Res 1998; 82:1145-59. [PMID: 9633915 DOI: 10.1161/01.res.82.11.1145] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
To determine whether angiotensin II (Ang II) stimulation of adult ventricular myocytes in vitro results in cellular hypertrophy, the changes in myocyte volume and protein content per cell were examined by confocal microscopy. Moreover, the possibility was considered that the upregulation of Ang II receptors on myocytes after infarction may potentiate and/or accelerate Ang II-mediated myocyte growth. Left ventricular myocytes isolated from control and failing hearts 3 days after infarction were cultured for 3 and 7 days in the presence of Ang II. Normal myocytes did not show an increase in volume and protein content at 3 days, but a 16% and 20% increase in these respective parameters was found at 7 days. Cell growth was faster and greater in myocytes from postinfarcted hearts. In these cells, myocyte volume increased 23% and protein content increased 28% at 3 days after Ang II administration. The higher hypertrophic reaction of myocytes from infarcted hearts occurred in spite of a 19% larger volume at isolation. In both groups of myocytes, the AT1 receptor blocker losartan completely inhibited the consequences of Ang II. Conversely, the AT2 receptor antagonist PD123319 had no effect on Ang II-induced hypertrophy. In conclusion, Ang II promotes myocyte growth through the activation of AT1 receptors, which modulate the time and magnitude of this cellular response.
Collapse
MESH Headings
- Angiotensin II/pharmacology
- Animals
- Cardiomegaly/metabolism
- Cardiomegaly/pathology
- Cell Size/drug effects
- Cells, Cultured
- Male
- Microscopy, Confocal
- Muscle Fibers, Skeletal/chemistry
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Myocardial Infarction/drug therapy
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardium/chemistry
- Myocardium/cytology
- Rats
- Rats, Sprague-Dawley
- Receptors, Angiotensin/analysis
- Receptors, Angiotensin/metabolism
- Up-Regulation/drug effects
- Up-Regulation/physiology
- Vasoconstrictor Agents/pharmacology
- Ventricular Function, Left
- Ventricular Function, Right
Collapse
Affiliation(s)
- Y Liu
- Department of Medicine, New York Medical College, Valhalla 10595, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Eble DM, Qi M, Waldschmidt S, Lucchesi PA, Byron KL, Samarel AM. Contractile activity is required for sarcomeric assembly in phenylephrine-induced cardiac myocyte hypertrophy. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C1226-37. [PMID: 9612209 DOI: 10.1152/ajpcell.1998.274.5.c1226] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Agonist-induced hypertrophy of cultured neonatal rat ventricular myocytes (NRVM) has been attributed to biochemical signals generated during receptor activation. However, NRVM hypertrophy can also be induced by spontaneous or electrically stimulated contractile activity in the absence of exogenous neurohormonal stimuli. Using single-cell imaging of fura 2-loaded myocytes, we found that low-density, noncontracting NRVM begin to generate intracellular Ca2+ concentration ([Ca2+]i) transients and contractile activity within minutes of exposure to the alpha 1-adrenergic agonist phenylephrine (PE; 50 microM). However, NRVM pretreated with verapamil and then stimulated with PE failed to elicit [Ca2+]i transients and beating. We therefore examined whether PE-induced [Ca2+]i transients and contractile activity were required to elicit specific aspects of the hypertrophic phenotype. PE treatment (48-72 h) increased cell size, total protein content, total protein-to-DNA ratio, and myosin heavy chain (MHC) isoenzyme content. PE also stimulated sarcomeric protein assembly and prolonged MHC half-life. However, blockade of voltage-gated L-type Ca2+ channels with verapamil, diltiazem, or nifedipine (10 microM) blocked PE-induced total protein and MHC accumulation and prevented the time-dependent assembly of myofibrillar proteins into sarcomeres. Inhibition of actin-myosin cross-bridge cycling with 2,3-butanedione monoxime (7.5 mM) also prevented PE-induced total protein and MHC accumulation, indicating that mechanical activity, rather than [Ca2+]i transients per se, was required. In contrast, blockade of [Ca2+]i transients and contractile activity did not prevent the PE-induced increase in cell surface area, activation of the mitogen-activated protein kinases ERK1 and ERK2, or upregulation of atrial natriuretic factor gene expression. Thus contractile activity is required to elicit some but not all aspects of the the hypertrophic phenotype induced by alpha 1-adrenergic receptor activation.
Collapse
Affiliation(s)
- D M Eble
- Cardiovascular Institute, Loyola University Chicago Stritch School of Medicine, Maywood, Illinois 60153, USA
| | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Gao T, Puri TS, Gerhardstein BL, Chien AJ, Green RD, Hosey MM. Identification and subcellular localization of the subunits of L-type calcium channels and adenylyl cyclase in cardiac myocytes. J Biol Chem 1997; 272:19401-7. [PMID: 9235939 DOI: 10.1074/jbc.272.31.19401] [Citation(s) in RCA: 148] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The properties of cardiac L-type channels have been well characterized electrophysiologically, and many such studies have demonstrated that the channels are regulated by a cAMP-dependent pathway. However, the subunit composition of native cardiac L-type calcium channels has not been completely defined. Furthermore, a very important question exists regarding the status of the C-terminal domain of the pore-forming alpha1 subunit, as this domain has the potential to be the target of protein kinases but may be truncated as a result of post-translational processing. In the present studies, the alpha1C and beta2 subunits were identified by subunit-specific antibodies after partial purification from heart membranes, or immunoprecipitation from cardiac myocytes. Both the beta2 and the full-length alpha1C subunits were found to be expressed and co-localized in intact cardiac myocytes along T-tubule membranes. Using a quantitative antibody binding analysis, we demonstrated that the majority of the alpha1C subunits in intact cardiac myocytes appear to be full-length. In addition, we observed that adenylyl cyclase is localized in a pattern similar to the channel subunits in cardiac myocytes. Taken together, our results provide new insights into the structural basis for understanding the regulation of L-type calcium channels by a cAMP-mediated signaling pathway.
Collapse
Affiliation(s)
- T Gao
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University Medical School, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
49
|
Arber S, Hunter JJ, Ross J, Hongo M, Sansig G, Borg J, Perriard JC, Chien KR, Caroni P. MLP-deficient mice exhibit a disruption of cardiac cytoarchitectural organization, dilated cardiomyopathy, and heart failure. Cell 1997; 88:393-403. [PMID: 9039266 DOI: 10.1016/s0092-8674(00)81878-4] [Citation(s) in RCA: 607] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
MLP is a LIM-only protein of terminally differentiated striated muscle cells, where it accumulates at actin-based structures involved in cytoarchitecture organization. To assess its role in muscle differentiation, we disrupted the MLP gene in mice. MLP (-/-) mice developed dilated cardiomyopathy with hypertrophy and heart failure after birth. Ultrastructural analysis revealed dramatic disruption of cardiomyocyte cytoarchitecture. At birth, these hearts were not hypertrophic, but already abnormally soft, with cell-autonomous and MLP-sensitive alterations in cytoarchitecture. Thus, MLP promotes proper cardiomyocyte cytoarchitecture, whose perturbation can lead to dilated cardiomyopathy. In vivo analysis revealed that MLP-deficient mice reproduce the morphological and clinical picture of dilated cardiomyopathy and heart failure in humans, providing the first model for this condition in a genetically manipulatable organism.
Collapse
Affiliation(s)
- S Arber
- Friedrich Miescher Institute, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Li RK, Jia ZQ, Weisel RD, Mickle DA, Zhang J, Mohabeer MK, Rao V, Ivanov J. Cardiomyocyte transplantation improves heart function. Ann Thorac Surg 1996; 62:654-60; discussion 660-1. [PMID: 8783989 DOI: 10.1016/s0003-4975(96)00389-x] [Citation(s) in RCA: 279] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Transplantation of cultured cardiomyocytes into myocardial scar tissue may prevent heart failure. METHODS Scar tissue was produced in the left ventricular free wall of 15 rats (weight, 450 g) by cryoinjury. Seven animals had operation only and survived for 8 weeks (sham group). Four weeks after cryoinjury, cultured fetal rat cardiomyocytes or culture medium was injected into the scar tissue of transplantation (n = 5) and control (n = 5) animals, respectively. Five other rats were sacrificed for scar assessment. Eight weeks after cryoinjury heart function in the transplantation, control, and sham groups was measured using a Langendorff preparation. Histologic studies were performed to quantify the extent of the scar and the transplanted cells. RESULTS Four weeks after cryoinjury, 36% +/- 4% (mean +/- 1 standard error) of the left ventricular free wall surface area was scar tissue. At 8 weeks, the scar size had increased (p < 0.01) to 55% +/- 3% in the control group. Although the scar size (43% +/- 2%) in the transplantation group at 8 weeks was not significantly different from that at 4 weeks, it was less (p < 0.05) than that in the control group. Hearts in the sham group had no scar tissue. The transplanted cardiomyocytes had formed cardiac tissue within the myocardial scar. Systolic and developed pressures in the transplantation group hearts were greater (p = 0.0001) than in the control group hearts but less (p < 0.01) than those in the sham group hearts. CONCLUSIONS The transplanted cardiomyocytes formed cardiac tissue in the myocardial scar, limited scar expansion, and improved heart function compared with findings in the control hearts.
Collapse
Affiliation(s)
- R K Li
- Department of Clinical Biochemistry, Toronto Hospital-General Division, University of Toronto, Ontario, Canada
| | | | | | | | | | | | | | | |
Collapse
|