1
|
Jia H, Zhang H, Liu Y, Guo J, Chen W, Zhang Y, Scarlat MM, Liu L, Hou Z. Identifying potential biomarkers for early evaluating mechanical compression injuries to skeletal muscle through proteomic analysis: A rat model. PLoS One 2025; 20:e0324706. [PMID: 40424254 PMCID: PMC12111613 DOI: 10.1371/journal.pone.0324706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
The skeletal muscle is highly susceptible to injury in daily life. Severe skeletal muscle injuries often result in incomplete regeneration, leading to functional impairment. In clinical practice, understanding the extent of skeletal muscle injury in limb trauma patients is crucial for selecting treatment modalities and assessing prognosis. Currently, there is a lack of specific indicators for evaluating the severity of mechanical skeletal muscle injury. Therefore, the aim of this study is to develop biomarkers for the early evaluation of different degrees of skeletal muscle injury. A rat model of skeletal muscle mechanical compression injury was established with varying degrees of injury severity, one control group, and two compression groups (Mild Injury and Severe Injury Group). LC-MS/MS-4D-DIA quantitative proteomics technology was used to detect the plasma proteome profile of rats in different injury groups at 3 hours post-injury, followed by bioinformatics analysis for data decoding. Rats in the mild and severe injury groups exhibited completely different degrees of injury and prognosis. The proteomic results of the plasma revealed that the relative quantification of 37 proteins increased along with the increase in injury, while 2 proteins decreased. These differentially expressed proteins (DEPs) included not only muscle-specific structural proteins but also metabolic-related proteins that might play crucial roles in tissue injury control, repair, and regeneration. Overall, the study has identified several potential protein biomarkers that can distinguish different degrees of skeletal muscle injury at an early stage. These protein biomarkers may be further developed to help clinicians identify patients with varying degrees of skeletal muscle injury, paving the way for personalized treatments.
Collapse
Affiliation(s)
- Huiyang Jia
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei Province, China
- Key Laboratory of Precise Assessment, Diagnosis, and Treament of Soft Tissue Injury of Hebei Province, China
| | - Heng Zhang
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei Province, China
- Key Laboratory of Precise Assessment, Diagnosis, and Treament of Soft Tissue Injury of Hebei Province, China
| | - Yan Liu
- Department of Endocrinology, Hebei Medical University Third Hospital, Shijiazhuang, Hebei Province, China
| | - Jialiang Guo
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei Province, China
- Key Laboratory of Precise Assessment, Diagnosis, and Treament of Soft Tissue Injury of Hebei Province, China
| | - Wei Chen
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei Province, China
- Key Laboratory of Precise Assessment, Diagnosis, and Treament of Soft Tissue Injury of Hebei Province, China
| | - Yingze Zhang
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei Province, China
- Key Laboratory of Precise Assessment, Diagnosis, and Treament of Soft Tissue Injury of Hebei Province, China
| | | | - Lin Liu
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei Province, China
- Key Laboratory of Precise Assessment, Diagnosis, and Treament of Soft Tissue Injury of Hebei Province, China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei Province, China
- Key Laboratory of Precise Assessment, Diagnosis, and Treament of Soft Tissue Injury of Hebei Province, China
| |
Collapse
|
2
|
Lin Y, Sato N, Hong S, Nakamura K, Ferrante EA, Yu ZX, Chen MY, Nakamura DS, Yang X, Clevenger RR, Hunt TJ, Taylor JL, Jeffries KR, Keeran KJ, Neidig LE, Mehta A, Schwartzbeck R, Yu SJ, Kelly C, Navarengom K, Takeda K, Adler SS, Choyke PL, Zou J, Murry CE, Boehm M, Dunbar CE. Long-term engraftment and maturation of autologous iPSC-derived cardiomyocytes in two rhesus macaques. Cell Stem Cell 2024; 31:974-988.e5. [PMID: 38843830 PMCID: PMC11227404 DOI: 10.1016/j.stem.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/14/2024] [Accepted: 05/10/2024] [Indexed: 07/08/2024]
Abstract
Cellular therapies with cardiomyocytes produced from induced pluripotent stem cells (iPSC-CMs) offer a potential route to cardiac regeneration as a treatment for chronic ischemic heart disease. Here, we report successful long-term engraftment and in vivo maturation of autologous iPSC-CMs in two rhesus macaques with small, subclinical chronic myocardial infarctions, all without immunosuppression. Longitudinal positron emission tomography imaging using the sodium/iodide symporter (NIS) reporter gene revealed stable grafts for over 6 and 12 months, with no teratoma formation. Histological analyses suggested capability of the transplanted iPSC-CMs to mature and integrate with endogenous myocardium, with no sign of immune cell infiltration or rejection. By contrast, allogeneic iPSC-CMs were rejected within 8 weeks of transplantation. This study provides the longest-term safety and maturation data to date in any large animal model, addresses concerns regarding neoantigen immunoreactivity of autologous iPSC therapies, and suggests that autologous iPSC-CMs would similarly engraft and mature in human hearts.
Collapse
Affiliation(s)
- Yongshun Lin
- iPSC Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Noriko Sato
- Laboratory of Cellular Therapeutics, Molecular Imaging Branch, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Sogun Hong
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| | - Elisa A Ferrante
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Zu Xi Yu
- Pathology Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Marcus Y Chen
- Cardiovascular Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Daisy S Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Timothy J Hunt
- Animal Surgery and Resources Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Joni L Taylor
- Animal Surgery and Resources Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | | | - Karen J Keeran
- Animal Surgery and Resources Core, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Lauren E Neidig
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Atul Mehta
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Robin Schwartzbeck
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Shiqin Judy Yu
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Conor Kelly
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Keron Navarengom
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA
| | - Kazuyo Takeda
- Microscopy and Imaging Core, CBER, FDA, Silver Spring, MD, USA
| | - Stephen S Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Peter L Choyke
- Laboratory of Cellular Therapeutics, Molecular Imaging Branch, National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Jizhong Zou
- iPSC Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA; Department of Medicine/Cardiology, University of Washington, Seattle, WA 98195, USA; Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington School of Medicine, Seattle, WA 98195, USA.
| | - Manfred Boehm
- Translational Vascular Medicine Branch, NHLBI, NIH, Bethesda, MD 20892, USA.
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, NHLBI, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
3
|
Bedada FB, Thompson BR, Mikkila JL, Chan SSK, Choi SH, Toso EA, Kyba M, Metzger JM. Inducing positive inotropy in human iPSC-derived cardiac muscle by gene editing-based activation of the cardiac α-myosin heavy chain. Sci Rep 2024; 14:3915. [PMID: 38365813 PMCID: PMC10873390 DOI: 10.1038/s41598-024-53395-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/31/2024] [Indexed: 02/18/2024] Open
Abstract
Human induced pluripotent stem cells and their differentiation into cardiac myocytes (hiPSC-CMs) provides a unique and valuable platform for studies of cardiac muscle structure-function. This includes studies centered on disease etiology, drug development, and for potential clinical applications in heart regeneration/repair. Ultimately, for these applications to achieve success, a thorough assessment and physiological advancement of the structure and function of hiPSC-CMs is required. HiPSC-CMs are well noted for their immature and sub-physiological cardiac muscle state, and this represents a major hurdle for the field. To address this roadblock, we have developed a hiPSC-CMs (β-MHC dominant) experimental platform focused on directed physiological enhancement of the sarcomere, the functional unit of cardiac muscle. We focus here on the myosin heavy chain (MyHC) protein isoform profile, the molecular motor of the heart, which is essential to cardiac physiological performance. We hypothesized that inducing increased expression of α-MyHC in β-MyHC dominant hiPSC-CMs would enhance contractile performance of hiPSC-CMs. To test this hypothesis, we used gene editing with an inducible α-MyHC expression cassette into isogeneic hiPSC-CMs, and separately by gene transfer, and then investigated the direct effects of increased α-MyHC expression on hiPSC-CMs contractility and relaxation function. Data show improved cardiac functional parameters in hiPSC-CMs induced with α-MyHC. Positive inotropy and relaxation was evident in comparison to β-MyHC dominant isogenic controls both at baseline and during pacing induced stress. This approach should facilitate studies of hiPSC-CMs disease modeling and drug screening, as well as advancing fundamental aspects of cardiac function parameters for the optimization of future cardiac regeneration, repair and re-muscularization applications.
Collapse
Affiliation(s)
- Fikru B Bedada
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
- Present Address: Department of Clinical Laboratory Sciences, College of Nursing and Allied Health Sciences, Howard University, Washington, DC, USA
| | - Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Jennifer L Mikkila
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Sunny S-K Chan
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Si Ho Choi
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Erik A Toso
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
- Lillehei Heart Institute, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN, 55455, USA.
| |
Collapse
|
4
|
Zhang C, Sun Y, Chen Z. An Efficient and Reproducible Method for the Isolation and Culture of Primary Cardiomyocytes from Adult Zebrafish. Zebrafish 2023. [PMID: 37262192 DOI: 10.1089/zeb.2023.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Abstract
Zebrafish is a popular animal model in regeneration studies due to their ability to regenerate the heart. Primary cardiomyocytes could be an alternative tool for studying the intrinsic mechanisms of cardiovascular disease in vitro. Thus, our objective is to develop an efficient protocol to isolate primary cardiomyocytes from zebrafish hearts. Low concentration of digestive enzyme (0.5 mg/mL collagenase type II) was utilized in our protocol to obtain single-cell suspension. The ventricles were fragmented, mechanically pipetted, and constantly shaken to ensure adequate contact between the tissues and the enzyme. Preplating the cell suspension onto culture plates for 2 h helped remove cardiac fibroblasts. The purity of isolated cells was validated by flow cytometry analysis of transgenic zebrafish with cardiomyocyte-specific expression of enhanced green fluorescent protein (EGFP) or endothelial cell-specific expression of mCherry. Quantitative real-time PCR analysis revealed a high level of the purity, with cardiac fibroblasts, endothelial cells, and epicardial cell markers scarcely detected in the purified cells. Altogether, this study established a reproducible protocol for isolating primary cardiomyocytes with high purity and activity from adult zebrafish hearts that can be cultured in vitro for up to 4 weeks. This protocol provides a valuable tool for studying the intrinsic mechanisms of cardiovascular disease in vitro using primary cardiomyocytes.
Collapse
Affiliation(s)
- Chunyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyi Sun
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyue Chen
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Cao Y, Redd MA, Fang C, Mizikovsky D, Li X, Macdonald PS, King GF, Palpant NJ. New Drug Targets and Preclinical Modelling Recommendations for Treating Acute Myocardial Infarction. Heart Lung Circ 2023:S1443-9506(23)00139-7. [PMID: 37230806 DOI: 10.1016/j.hlc.2022.12.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/28/2022] [Accepted: 12/15/2022] [Indexed: 05/27/2023]
Abstract
Acute myocardial infarction (AMI) is the leading cause of morbidity and mortality worldwide and the primary underlying risk factor for heart failure. Despite decades of research and clinical trials, there are no drugs currently available to prevent organ damage from acute ischaemic injuries of the heart. In order to address the increasing global burden of heart failure, drug, gene, and cell-based regeneration technologies are advancing into clinical testing. In this review we highlight the burden of disease associated with AMI and the therapeutic landscape based on market analyses. New studies revealing the role of acid-sensitive cardiac ion channels and other proton-gated ion channels in cardiac ischaemia are providing renewed interest in pre- and post-conditioning agents with novel mechanisms of action that may also have implications for gene- and cell-based therapeutics. Furthermore, we present guidelines that couple new cell technologies and data resources with traditional animal modelling pipelines to help de-risk drug candidates aimed at treating AMI. We propose that improved preclinical pipelines and increased investment in drug target identification for AMI is critical to stem the increasing global health burden of heart failure.
Collapse
Affiliation(s)
- Yuanzhao Cao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Meredith A Redd
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Chen Fang
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Dalia Mizikovsky
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Xichun Li
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia
| | - Peter S Macdonald
- Cardiopulmonary Transplant Unit, St Vincent's Hospital, Sydney, NSW, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Qld, Australia
| | - Nathan J Palpant
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Qld, Australia.
| |
Collapse
|
6
|
Chen FL, Shang LD, Lin YC, Chang BY, Hsiao YC. Label-Free, Portable, and Color-Indicating Cholesteric Liquid Crystal Test Kit for Acute Myocardial Infarction by Spectral Analysis and Naked-Eye Observation. BIOSENSORS 2022; 13:60. [PMID: 36671895 PMCID: PMC9856049 DOI: 10.3390/bios13010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/24/2022] [Accepted: 12/29/2022] [Indexed: 06/17/2023]
Abstract
The early diagnosis of acute myocardial infarction is difficult in patients with nondiagnostic characteristics. Acute myocardial infarction with chest pain is associated with increased mortality. This study developed a portable test kit based on cholesteric liquid crystals (CLCs) for the rapid detection of AMI through eye observation at home. The test kit was established on dimethyloctadecyl[3-(trimethoxysilyl)propyl]ammonium chloride-coated substrates covered by a CLC-binding antibody. Cardiac troponin I (cTnI) is a major biomarker of myocardial cellular injury in human blood. The data showed that the concentration of cTnI was related to light transmittance in a positive way. The proposed CLC test kit can be operated with a smartphone; therefore, it has high potential for use as a point-of-care device for home testing. Moreover, the CLC test kit is an effective and innovative device for the rapid testing of acute myocardial infarction-related diseases through eye observation, spectrometer, or even smartphone applications.
Collapse
Affiliation(s)
- Fu-Lun Chen
- Department of Internal Medicine, Division of Infectious Diseases, Wan Fang Hospital, Taipei Medical University, No.111, Sec. 3, Xinglong Rd., Wenshan Dist., Taipei 11600, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan
| | - Li-Dan Shang
- Department of Geography and Planning, University of Liverpool, Liverpool L69 3BX, UK
| | - Yen-Chung Lin
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St., Taipei 11031, Taiwan
- Department of Internal Medicine, Division of Nephrology, Taipei Medical University Hospital, 252 Wuxing St., Taipei 110, Taiwan
- TMU Research Center of Urology and Kidney (TMU-RCUK), Taipei Medical University, Taipei 110, Taiwan
| | - Bo-Yen Chang
- Department of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Cheng Hsiao
- Graduate Institute of Biomedical Optomechatronics, College of Biomedical Engineering, Taipei 11031, Taiwan
- International PhD Program for Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
7
|
Martin AA, Thompson BR, Hahn D, Angulski ABB, Hosny N, Cohen H, Metzger JM. Cardiac Sarcomere Signaling in Health and Disease. Int J Mol Sci 2022; 23:16223. [PMID: 36555864 PMCID: PMC9782806 DOI: 10.3390/ijms232416223] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
The cardiac sarcomere is a triumph of biological evolution wherein myriad contractile and regulatory proteins assemble into a quasi-crystalline lattice to serve as the central point upon which cardiac muscle contraction occurs. This review focuses on the many signaling components and mechanisms of regulation that impact cardiac sarcomere function. We highlight the roles of the thick and thin filament, both as necessary structural and regulatory building blocks of the sarcomere as well as targets of functionally impactful modifications. Currently, a new focus emerging in the field is inter-myofilament signaling, and we discuss here the important mediators of this mechanism, including myosin-binding protein C and titin. As the understanding of sarcomere signaling advances, so do the methods with which it is studied. This is reviewed here through discussion of recent live muscle systems in which the sarcomere can be studied under intact, physiologically relevant conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
Chuang EY, Ho TL, Wang YC, Hsiao YC. Smartphone and home-based liquid crystal sensor for rapid screening of acute myocardial infarction by naked-eye observation and image analysis. Talanta 2022; 250:123698. [PMID: 35763951 DOI: 10.1016/j.talanta.2022.123698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/27/2022]
Abstract
An early diagnosis of acute myocardial infarction (AMI) or thrombosis is complicated in patients with non-diagnostic features. AMI or thrombosis patients with chest pain are unintentionally discharged and have increased mortality. The study aimed to develop a smartphone biomedical sensor as a rapid test for AMI or thrombosis by naked-eye observation. The system was built on dimethyloctadecyl [3-(trimethoxysilyl)propyl]ammonium chloride (DMOAP)-coated glass substrates, which refers to a nematic liquid crystal (LC)-binding antibody. One of the main biomolecules, cardiac troponin I (cTnI), is a substance in blood in people whose bodies are vulnerable to suffering a myocardial infarction or thrombosis. The other medium, LC, is a sensing biomaterial as an earlier detection method of ameliorating the disadvantages of older methods. Results revealed that the density of cTnI was positively correlated with the coefficient of light transmittance, and it has a high chance of being developed as a point-of-care device for a home inspection as it can be operated with a smartphone. As discussed above, the nematic LC is an effective and innovative healthcare method as a rapid test for diagnosis of AMI or thrombosis related diseases by naked-eye observation.
Collapse
Affiliation(s)
- Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan; International PhD Program for Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Thi-Luu Ho
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yen-Chieh Wang
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Yu-Cheng Hsiao
- International PhD Program for Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan; Graduate Institute of Biomedical Optomechatronics, College of Biomedical Engineering, Taipei, 11031, Taiwan; Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11031, Taiwan; Stanford Byers Center for Biodesign, Stanford, USA.
| |
Collapse
|
9
|
Rasmussen M, Jin JP. Troponin Variants as Markers of Skeletal Muscle Health and Diseases. Front Physiol 2021; 12:747214. [PMID: 34733179 PMCID: PMC8559874 DOI: 10.3389/fphys.2021.747214] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Ca2 +-regulated contractility is a key determinant of the quality of muscles. The sarcomeric myofilament proteins are essential players in the contraction of striated muscles. The troponin complex in the actin thin filaments plays a central role in the Ca2+-regulation of muscle contraction and relaxation. Among the three subunits of troponin, the Ca2+-binding subunit troponin C (TnC) is a member of the calmodulin super family whereas troponin I (TnI, the inhibitory subunit) and troponin T (TnT, the tropomyosin-binding and thin filament anchoring subunit) are striated muscle-specific regulatory proteins. Muscle type-specific isoforms of troponin subunits are expressed in fast and slow twitch fibers and are regulated during development and aging, and in adaptation to exercise or disuse. TnT also evolved with various alternative splice forms as an added capacity of muscle functional diversity. Mutations of troponin subunits cause myopathies. Owing to their physiological and pathological importance, troponin variants can be used as specific markers to define muscle quality. In this focused review, we will explore the use of troponin variants as markers for the fiber contents, developmental and differentiation states, contractile functions, and physiological or pathophysiological adaptations of skeletal muscle. As protein structure defines function, profile of troponin variants illustrates how changes at the myofilament level confer functional qualities at the fiber level. Moreover, understanding of the role of troponin modifications and mutants in determining muscle contractility in age-related decline of muscle function and in myopathies informs an approach to improve human health.
Collapse
Affiliation(s)
- Monica Rasmussen
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
10
|
de Lange WJ, Farrell ET, Kreitzer CR, Jacobs DR, Lang D, Glukhov AV, Ralphe JC. Human iPSC-engineered cardiac tissue platform faithfully models important cardiac physiology. Am J Physiol Heart Circ Physiol 2021; 320:H1670-H1686. [PMID: 33606581 DOI: 10.1152/ajpheart.00941.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CM) may provide an important bridge between animal models and the intact human myocardium. Fulfilling this potential is hampered by their relative immaturity, leading to poor physiological responsiveness. hiPSC-CMs grown in traditional two-dimensional (2D) culture lack a t-tubular system, have only rudimentary intracellular calcium-handling systems, express predominantly embryonic sarcomeric protein isoforms, and preferentially use glucose as an energy substrate. Culturing hiPSC-CM in a variety of three-dimensional (3D) environments and the addition of nutritional, pharmacological, and electromechanical stimuli have proven, to various degrees, to be beneficial for maturation. We present a detailed assessment of a novel model in which hiPSC-CMs and hiPSC-derived cardiac fibroblasts are cocultured in a 3D fibrin matrix to form engineered cardiac tissue constructs (hiPSC-ECTs). The hiPSC-ECTs are responsive to physiological stimuli, including stretch, frequency, and β-adrenergic stimulation, develop a t-tubular system, and demonstrate calcium-handling and contractile kinetics that compare favorably with ventricular human myocardium. Furthermore, transcript levels of various genes involved in calcium-handling and contraction are increased. These markers of maturation become more robust over a relatively short period of time in culture (6 wk vs. 2 wk in hiPSC-ECTs). A comparison of the hiPSC-ECT molecular and performance variables with those of human cardiac tissue and other available engineered tissue platforms is provided to aid selection of the most appropriate platform for the research question at hand. Important and noteworthy aspects of this human cardiac model system are its reliance on "off-the-shelf" equipment, ability to provide detailed physiological performance data, and the ability to achieve a relatively mature cardiac physiology without additional nutritional, pharmacological, and electromechanical stimuli that may elicit unintended effects on function.NEW & NOTEWORTHY This study seeks to provide an in-depth assessment of contractile performance of human iPSC-derived cardiomyocytes cultured together with fibroblasts in a 3-dimensional-engineered tissue and compares performance both over time as cells mature, and with corresponding measures found in the literature using alternative 3D culture configurations. The suitability of 3D-engineered human cardiac tissues to model cardiac function is emphasized, and data provided to assist in the selection of the most appropriate configuration based on the target application.
Collapse
Affiliation(s)
- Willem J de Lange
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Emily T Farrell
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Caroline R Kreitzer
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Derek R Jacobs
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Di Lang
- Department of Medicine Cardiovascular Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - Alexey V Glukhov
- Department of Medicine Cardiovascular Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Wisconsin
| | - J Carter Ralphe
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
11
|
Santini L, Palandri C, Nediani C, Cerbai E, Coppini R. Modelling genetic diseases for drug development: Hypertrophic cardiomyopathy. Pharmacol Res 2020; 160:105176. [DOI: 10.1016/j.phrs.2020.105176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/16/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
|
12
|
Suh TC, Amanah AY, Gluck JM. Electrospun Scaffolds and Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Cardiac Tissue Engineering Applications. Bioengineering (Basel) 2020; 7:E105. [PMID: 32899986 PMCID: PMC7552723 DOI: 10.3390/bioengineering7030105] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 01/14/2023] Open
Abstract
Tissue engineering (TE) combines cells, scaffolds, and growth factors to assemble functional tissues for repair or replacement of tissues and organs. Cardiac TE is focused on developing cardiac cells, tissues, and structures-most notably the heart. This review presents the requirements, challenges, and research surrounding electrospun scaffolds and induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CMs) towards applications to TE hearts. Electrospinning is an attractive fabrication method for cardiac TE scaffolds because it produces fibers that demonstrate the optimal potential for mimicking the complex structure of the cardiac extracellular matrix (ECM). iPSCs theoretically offer the capacity to generate limitless numbers of CMs for use in TE hearts, however these iPSC-CMs are electrophysiologically, morphologically, mechanically, and metabolically immature compared to adult CMs. This presents a functional limitation to their use in cardiac TE, and research aiming to address this limitation is presented in this review.
Collapse
Affiliation(s)
- Taylor Cook Suh
- Textile Engineering, Chemistry and Science Department, Wilson College of Textiles, NC State University, Raleigh, NC 27695, USA
| | - Alaowei Y Amanah
- Textile Engineering, Chemistry and Science Department, Wilson College of Textiles, NC State University, Raleigh, NC 27695, USA
| | - Jessica M Gluck
- Textile Engineering, Chemistry and Science Department, Wilson College of Textiles, NC State University, Raleigh, NC 27695, USA
| |
Collapse
|
13
|
Sarcomere integrated biosensor detects myofilament-activating ligands in real time during twitch contractions in live cardiac muscle. J Mol Cell Cardiol 2020; 147:49-61. [PMID: 32791214 DOI: 10.1016/j.yjmcc.2020.07.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/14/2020] [Accepted: 07/30/2020] [Indexed: 11/24/2022]
Abstract
The sarcomere is the functional unit of cardiac muscle, essential for normal heart function. To date, it has not been possible to study, in real time, thin filament-based activation dynamics in live cardiac muscle. We report here results from a cardiac troponin C (TnC) FRET-based biosensor integrated into the cardiac sarcomere via stoichiometric replacement of endogenous TnC. The TnC biosensor provides, for the first time, evidence of multiple thin filament activating ligands, including troponin I interfacing with TnC and cycling myosin, during a cardiac twitch. Results show that the TnC FRET biosensor transient significantly precedes that of peak twitch force. Using small molecules and genetic modifiers known to alter sarcomere activation, independently of the intracellular Ca2+ transient, the data show that the TnC biosensor detects significant effects of the troponin I switch domain as a sarcomere-activating ligand. Interestingly, the TnC biosensor also detected the effects of load-dependent altered myosin cycling, as shown by a significant delay in TnC biosensor transient inactivation during the isometric twitch. In addition, the TnC biosensor detected the effects of myosin as an activating ligand during the twitch by using a small molecule that directly alters cross-bridge cycling, independently of the intracellular Ca2+ transient. Collectively, these results aid in illuminating the basis of cardiac muscle contractile activation with implications for gene, protein, and small molecule-based strategies designed to target the sarcomere in regulating beat-to-beat heart performance in health and disease.
Collapse
|
14
|
Karbassi E, Fenix A, Marchiano S, Muraoka N, Nakamura K, Yang X, Murry CE. Cardiomyocyte maturation: advances in knowledge and implications for regenerative medicine. Nat Rev Cardiol 2020; 17:341-359. [PMID: 32015528 DOI: 10.1038/s41569-019-0331-x] [Citation(s) in RCA: 460] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2019] [Indexed: 12/20/2022]
Abstract
Our knowledge of pluripotent stem cell (PSC) biology has advanced to the point where we now can generate most cells of the human body in the laboratory. PSC-derived cardiomyocytes can be generated routinely with high yield and purity for disease research and drug development, and these cells are now gradually entering the clinical research phase for the testing of heart regeneration therapies. However, a major hurdle for their applications is the immature state of these cardiomyocytes. In this Review, we describe the structural and functional properties of cardiomyocytes and present the current approaches to mature PSC-derived cardiomyocytes. To date, the greatest success in maturation of PSC-derived cardiomyocytes has been with transplantation into the heart in animal models and the engineering of 3D heart tissues with electromechanical conditioning. In conventional 2D cell culture, biophysical stimuli such as mechanical loading, electrical stimulation and nanotopology cues all induce substantial maturation, particularly of the contractile cytoskeleton. Metabolism has emerged as a potent means to control maturation with unexpected effects on electrical and mechanical function. Different interventions induce distinct facets of maturation, suggesting that activating multiple signalling networks might lead to increased maturation. Despite considerable progress, we are still far from being able to generate PSC-derived cardiomyocytes with adult-like phenotypes in vitro. Future progress will come from identifying the developmental drivers of maturation and leveraging them to create more mature cardiomyocytes for research and regenerative medicine.
Collapse
Affiliation(s)
- Elaheh Karbassi
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Aidan Fenix
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Silvia Marchiano
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Naoto Muraoka
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kenta Nakamura
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Xiulan Yang
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.,Department of Pathology, University of Washington, Seattle, WA, USA
| | - Charles E Murry
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA. .,Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA. .,Department of Pathology, University of Washington, Seattle, WA, USA. .,Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA. .,Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
15
|
Xia C, Zhou D, Su Y, Zhou G, Yao L, Sun W, Liu Y. A liquid-crystal-based immunosensor for the detection of cardiac troponin I. Analyst 2020; 145:4569-4575. [DOI: 10.1039/d0an00425a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cardiac troponin I (cTnI) is one of the most sensitive and specific markers of myocardial cell injury. In this study, a label-free biosensor that utilizes the birefringence property of liquid crystal (LC) for the detection of cTnI is demonstrated.
Collapse
Affiliation(s)
- Chunli Xia
- Key Lab of In-fiber Integrated Optics
- Ministry Education of China
- Harbin Engineering University
- Harbin 150001
- China
| | - Dong Zhou
- Key Lab of In-fiber Integrated Optics
- Ministry Education of China
- Harbin Engineering University
- Harbin 150001
- China
| | - Yueming Su
- Key Lab of In-fiber Integrated Optics
- Ministry Education of China
- Harbin Engineering University
- Harbin 150001
- China
| | - Guangkai Zhou
- Department of head and neck surgery
- Affiliated Tumor Hospital of Harbin Medical University
- Harbin 150001
- China
| | - Lishuang Yao
- State Key Laboratory of Applied Optics
- Changchun Institute of Optics
- Fine Mechanics and Physics
- Chinese Academy of Sciences
- Changchun 130033
| | - Weimin Sun
- Key Lab of In-fiber Integrated Optics
- Ministry Education of China
- Harbin Engineering University
- Harbin 150001
- China
| | - Yongjun Liu
- Key Lab of In-fiber Integrated Optics
- Ministry Education of China
- Harbin Engineering University
- Harbin 150001
- China
| |
Collapse
|
16
|
Nakano SJ, Walker JS, Walker LA, Li X, Du Y, Miyamoto SD, Sucharov CC, Garcia AM, Mitchell MB, Ambardekar AV, Stauffer BL. Increased myocyte calcium sensitivity in end-stage pediatric dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2019; 317:H1221-H1230. [PMID: 31625780 DOI: 10.1152/ajpheart.00409.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Dilated cardiomyopathy (DCM) is the most common cause of heart failure (HF) in children, resulting in high mortality and need for heart transplantation. The pathophysiology underlying pediatric DCM is largely unclear; however, there is emerging evidence that molecular adaptations and response to conventional HF medications differ between children and adults. To gain insight into alterations leading to systolic dysfunction in pediatric DCM, we measured cardiomyocyte contractile properties and sarcomeric protein phosphorylation in explanted pediatric DCM myocardium (N = 8 subjects) compared with nonfailing (NF) pediatric hearts (N = 8 subjects). Force-pCa curves were generated from skinned cardiomyocytes in the presence and absence of protein kinase A. Sarcomeric protein phosphorylation was quantified with Pro-Q Diamond staining after gel electrophoresis. Pediatric DCM cardiomyocytes demonstrate increased calcium sensitivity (pCa50 =5.70 ± 0.0291), with an associated decrease in troponin (Tn)I phosphorylation compared with NF pediatric cardiomyocytes (pCa50 =5.59 ± 0.0271, P = 0.0073). Myosin binding protein C and TnT phosphorylation are also lower in pediatric DCM, whereas desmin phosphorylation is increased. Pediatric DCM cardiomyocytes generate peak tension comparable to that of NF pediatric cardiomyocytes [DCM 29.7 mN/mm2, interquartile range (IQR) 21.5-49.2 vs. NF 32.8 mN/mm2, IQR 21.5-49.2 mN/mm2; P = 0.6125]. In addition, cooperativity is decreased in pediatric DCM compared with pediatric NF (Hill coefficient: DCM 1.56, IQR 1.31-1.94 vs. NF 1.94, IQR 1.36-2.86; P = 0.0425). Alterations in sarcomeric phosphorylation and cardiomyocyte contractile properties may represent an impaired compensatory response, contributing to the detrimental DCM phenotype in children.NEW & NOTEWORTHY Our study is the first to demonstrate that cardiomyocytes from infants and young children with dilated cardiomyopathy (DCM) exhibit increased calcium sensitivity (likely mediated by decreased troponin I phosphorylation) compared with nonfailing pediatric cardiomyocytes. Compared with published values in adult cardiomyocytes, pediatric cardiomyocytes have notably decreased cooperativity, with a further reduction in the setting of DCM. Distinct adaptations in cardiomyocyte contractile properties may contribute to a differential response to pharmacological therapies in the pediatric DCM population.
Collapse
Affiliation(s)
- Stephanie J Nakano
- Division of Cardiology, Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | - John S Walker
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Xiaotao Li
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Yanmei Du
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Shelley D Miyamoto
- Division of Cardiology, Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Anastacia M Garcia
- Division of Cardiology, Department of Pediatrics, University of Colorado Denver, Aurora, Colorado
| | - Max B Mitchell
- Division of Cardiothoracic Surgery, Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Amrut V Ambardekar
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, Colorado
| | - Brian L Stauffer
- Division of Cardiology, Department of Medicine, University of Colorado Denver, Aurora, Colorado.,Division of Cardiology, Department of Medicine, Denver Health and Hospital Authority, Denver, Colorado
| |
Collapse
|
17
|
Learn from Your Elders: Developmental Biology Lessons to Guide Maturation of Stem Cell-Derived Cardiomyocytes. Pediatr Cardiol 2019; 40:1367-1387. [PMID: 31388700 PMCID: PMC6786957 DOI: 10.1007/s00246-019-02165-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023]
Abstract
Human pluripotent stem cells (hPSCs) offer a multifaceted platform to study cardiac developmental biology, understand disease mechanisms, and develop novel therapies. Remarkable progress over the last two decades has led to methods to obtain highly pure hPSC-derived cardiomyocytes (hPSC-CMs) with reasonable ease and scalability. Nevertheless, a major bottleneck for the translational application of hPSC-CMs is their immature phenotype, resembling that of early fetal cardiomyocytes. Overall, bona fide maturation of hPSC-CMs represents one of the most significant goals facing the field today. Developmental biology studies have been pivotal in understanding the mechanisms to differentiate hPSC-CMs. Similarly, evaluation of developmental cues such as electrical and mechanical activities or neurohormonal and metabolic stimulations revealed the importance of these pathways in cardiomyocyte physiological maturation. Those signals cooperate and dictate the size and the performance of the developing heart. Likewise, this orchestra of stimuli is important in promoting hPSC-CM maturation, as demonstrated by current in vitro maturation approaches. Different shades of adult-like phenotype are achieved by prolonging the time in culture, electromechanical stimulation, patterned substrates, microRNA manipulation, neurohormonal or metabolic stimulation, and generation of human-engineered heart tissue (hEHT). However, mirroring this extremely dynamic environment is challenging, and reproducibility and scalability of these approaches represent the major obstacles for an efficient production of mature hPSC-CMs. For this reason, understanding the pattern behind the mechanisms elicited during the late gestational and early postnatal stages not only will provide new insights into postnatal development but also potentially offer new scalable and efficient approaches to mature hPSC-CMs.
Collapse
|
18
|
TnI Structural Interface with the N-Terminal Lobe of TnC as a Determinant of Cardiac Contractility. Biophys J 2019; 114:1646-1656. [PMID: 29642034 DOI: 10.1016/j.bpj.2018.02.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 12/24/2022] Open
Abstract
The heterotrimeric cardiac troponin complex is a key regulator of contraction and plays an essential role in conferring Ca2+ sensitivity to the sarcomere. During ischemic injury, rapidly accumulating protons acidify the myoplasm, resulting in markedly reduced Ca2+ sensitivity of the sarcomere. Unlike the adult heart, sarcomeric Ca2+ sensitivity in fetal cardiac tissue is comparatively pH insensitive. Replacement of the adult cardiac troponin I (cTnI) isoform with the fetal troponin I (ssTnI) isoform renders adult cardiac contractile machinery relatively insensitive to acidification. Alignment and functional studies have determined histidine 132 of ssTnI to be the predominant source of this pH insensitivity. Substitution of histidine at the cognate position 164 in cTnI confers the same pH insensitivity to adult cardiac myocytes. An alanine at position 164 of cTnI is conserved in all mammals, with the exception of the platypus, which expresses a proline. Prolines are biophysically unique because of their innate conformational rigidity and helix-disrupting function. To provide deeper structure-function insight into the role of the TnC-TnI interface in determining contractility, we employed a live-cell approach alongside molecular dynamics simulations to ascertain the chemo-mechanical implications of the disrupted helix 4 of cTnI where position 164 exists. This important motif belongs to the critical switch region of cTnI. Substitution of a proline at position 164 of cTnI in adult rat cardiac myocytes causes increased contractility independent of alterations in the Ca2+ transient. Free-energy perturbation calculations of cTnC-Ca2+ binding indicate no difference in cTnC-Ca2+ affinity. Rather, we propose the enhanced contractility is derived from new salt bridge interactions between cTnI helix 4 and cTnC helix A, which are critical in determining pH sensitivity and contractility. Molecular dynamics simulations demonstrate that cTnI A164P structurally phenocopies ssTnI under baseline but not acidotic conditions. These findings highlight the evolutionarily directed role of the TnI-cTnC interface in determining cardiac contractility.
Collapse
|
19
|
Veltri T, Landim-Vieira M, Parvatiyar MS, Gonzalez-Martinez D, Dieseldorff Jones KM, Michell CA, Dweck D, Landstrom AP, Chase PB, Pinto JR. Hypertrophic Cardiomyopathy Cardiac Troponin C Mutations Differentially Affect Slow Skeletal and Cardiac Muscle Regulation. Front Physiol 2017; 8:221. [PMID: 28473771 PMCID: PMC5397416 DOI: 10.3389/fphys.2017.00221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/27/2017] [Indexed: 12/22/2022] Open
Abstract
Mutations in TNNC1-the gene encoding cardiac troponin C (cTnC)-that have been associated with hypertrophic cardiomyopathy (HCM) and cardiac dysfunction may also affect Ca2+-regulation and function of slow skeletal muscle since the same gene is expressed in both cardiac and slow skeletal muscle. Therefore, we reconstituted rabbit soleus fibers and bovine masseter myofibrils with mutant cTnCs (A8V, C84Y, E134D, and D145E) associated with HCM to investigate their effects on contractile force and ATPase rates, respectively. Previously, we showed that these HCM cTnC mutants, except for E134D, increased the Ca2+ sensitivity of force development in cardiac preparations. In the current study, an increase in Ca2+ sensitivity of isometric force was only observed for the C84Y mutant when reconstituted in soleus fibers. Incorporation of cTnC C84Y in bovine masseter myofibrils reduced the ATPase activity at saturating [Ca2+], whereas, incorporation of cTnC D145E increased the ATPase activity at inhibiting and saturating [Ca2+]. We also tested whether reconstitution of cardiac fibers with troponin complexes containing the cTnC mutants and slow skeletal troponin I (ssTnI) could emulate the slow skeletal functional phenotype. Reconstitution of cardiac fibers with troponin complexes containing ssTnI attenuated the Ca2+ sensitization of isometric force when cTnC A8V and D145E were present; however, it was enhanced for C84Y. In summary, although the A8V and D145E mutants are present in both muscle types, their functional phenotype is more prominent in cardiac muscle than in slow skeletal muscle, which has implications for the protein-protein interactions within the troponin complex. The C84Y mutant warrants further investigation since it drastically alters the properties of both muscle types and may account for the earlier clinical onset in the proband.
Collapse
Affiliation(s)
- Tiago Veltri
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - Michelle S. Parvatiyar
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of MedicineMiami, FL, USA
| | - David Gonzalez-Martinez
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | | | - Clara A. Michell
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - David Dweck
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| | - Andrew P. Landstrom
- Section of Pediatric Cardiology, Department of Pediatrics, Baylor College of MedicineHouston, TX, USA
| | - P. Bryant Chase
- Department of Biological Science, Florida State UniversityTallahassee, FL, USA
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State University College of MedicineTallahassee, FL, USA
| |
Collapse
|
20
|
Bedada FB, Martindale JJ, Arden E, Metzger JM. Molecular inotropy mediated by cardiac miR-based PDE4D/PRKAR1α/phosphoprotein signaling. Sci Rep 2016; 6:36803. [PMID: 27833092 PMCID: PMC5105063 DOI: 10.1038/srep36803] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 10/21/2016] [Indexed: 01/05/2023] Open
Abstract
Molecular inotropy refers to cardiac contractility that can be modified to affect overall heart pump performance. Here we show evidence of a new molecular pathway for positive inotropy by a cardiac-restricted microRNA (miR). We report enhanced cardiac myocyte performance by acute titration of cardiac myosin-embedded miR-208a. The observed positive effect was independent of host gene myosin effects with evidence of negative regulation of cAMP-specific 3',5'-cyclic phosphodiesterase 4D (PDE4D) and the regulatory subunit of PKA (PRKAR1α) content culminating in PKA-site dependent phosphorylation of cardiac troponin I (cTnI) and phospholamban (PLN). Further, acute inhibition of miR-208a in adult myocytes in vitro increased PDE4D expression causing reduced isoproterenol-mediated phosphorylation of cTnI and PLN. Next, rAAV-mediated miR-208a gene delivery enhanced heart contractility and relaxation parameters in vivo. Finally, acute inducible increases in cardiac miR-208a in vivo reduced PDE4D and PRKAR1α, with evidence of increased content of several complementary miRs harboring the PDE4D recognition sequence. Physiologically, this resulted in significant cardiac cTnI and PLN phosphorylation and improved heart performance in vivo. As phosphorylation of cTnI and PLN is critical to myocyte function, titration of miR-208a represents a potential new mechanism to enhance myocardial performance via the PDE4D/PRKAR1α/PKA phosphoprotein signaling pathway.
Collapse
Affiliation(s)
- Fikru B. Bedada
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| | - Joshua J. Martindale
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| | - Erik Arden
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455 USA
| |
Collapse
|
21
|
Gillis TE, Klaiman JM, Foster A, Platt MJ, Huber JS, Corso MY, Simpson JA. Dissecting the role of the myofilament in diaphragm dysfunction during the development of heart failure in mice. Am J Physiol Heart Circ Physiol 2015; 310:H572-86. [PMID: 26702144 DOI: 10.1152/ajpheart.00773.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/21/2015] [Indexed: 11/22/2022]
Abstract
Dyspnea and reduced exercise capacity, caused, in part, by respiratory muscle dysfunction, are common symptoms in patients with heart failure (HF). However, the etiology of diaphragmatic dysfunction has not been identified. To investigate the effects of HF on diaphragmatic function, models of HF were surgically induced in CD-1 mice by transverse aortic constriction (TAC) and acute myocardial infarction (AMI), respectively. Assessment of myocardial function, isolated diaphragmatic strip function, myofilament force-pCa relationship, and phosphorylation status of myofilament proteins was performed at either 2 or 18 wk postsurgery. Echocardiography and invasive hemodynamics revealed development of HF by 18 wk postsurgery in both models. In vitro diaphragmatic force production was preserved in all groups while morphometric analysis revealed diaphragmatic atrophy and fibrosis in 18 wk TAC and AMI groups. Isometric force-pCa measurements of myofilament preparations revealed reduced Ca(2+) sensitivity of force generation and force generation at half-maximum and maximum Ca(2+) activation in 18 wk TAC. The rate of force redevelopment (ktr) was reduced in all HF groups at high levels of Ca(2+) activation. Finally, there were significant changes in the myofilament phosphorylation status of the 18 wk TAC group. This includes a decrease in the phosphorylation of troponin T, desmin, myosin light chain (MLC) 1, and MLC 2 as well as a shift in myosin isoforms. These results indicate that there are multiple changes in diaphragmatic myofilament function, which are specific to the type and stage of HF and occur before overt impairment of in vitro force production.
Collapse
Affiliation(s)
- Todd E Gillis
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada; Cardiovascular Research Center, University of Guelph, Guelph, Ontario, Canada; and
| | - Jordan M Klaiman
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada; Cardiovascular Research Center, University of Guelph, Guelph, Ontario, Canada; and
| | - Andrew Foster
- Cardiovascular Research Center, University of Guelph, Guelph, Ontario, Canada; and Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Mathew J Platt
- Cardiovascular Research Center, University of Guelph, Guelph, Ontario, Canada; and Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jason S Huber
- Cardiovascular Research Center, University of Guelph, Guelph, Ontario, Canada; and Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Melissa Y Corso
- Cardiovascular Research Center, University of Guelph, Guelph, Ontario, Canada; and Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jeremy A Simpson
- Cardiovascular Research Center, University of Guelph, Guelph, Ontario, Canada; and Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
22
|
Sheng JJ, Jin JP. TNNI1, TNNI2 and TNNI3: Evolution, regulation, and protein structure-function relationships. Gene 2015; 576:385-94. [PMID: 26526134 DOI: 10.1016/j.gene.2015.10.052] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/21/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022]
Abstract
Troponin I (TnI) is the inhibitory subunit of the troponin complex in the sarcomeric thin filament of striated muscle and plays a central role in the calcium regulation of contraction and relaxation. Vertebrate TnI has evolved into three isoforms encoded by three homologous genes: TNNI1 for slow skeletal muscle TnI, TNNI2 for fast skeletal muscle TnI and TNNI3 for cardiac TnI, which are expressed under muscle type-specific and developmental regulations. To summarize the current knowledge on the TnI isoform genes and products, this review focuses on the evolution, gene regulation, posttranslational modifications, and structure-function relationship of TnI isoform proteins. Their physiological and medical significances are also discussed.
Collapse
Affiliation(s)
- Juan-Juan Sheng
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jian-Ping Jin
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
23
|
Xu Y, Liu L, Pan B, Zhu J, Nan C, Huang X, Tian J. DNA methylation regulates mouse cardiac myofibril gene expression during heart development. J Biomed Sci 2015; 22:88. [PMID: 26475623 PMCID: PMC4609054 DOI: 10.1186/s12929-015-0203-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 10/09/2015] [Indexed: 12/31/2022] Open
Abstract
Background It is well known that epigenetic modifications play an important role in controlling the regulation of gene expression during the development. Our previous studies have demonstrated that the expression of fetal troponin I gene (also called slow skeletal troponin I, ssTnI) is predominated in the fetal stage, reduced after birth and disappeared in the adulthood. The mechanism underlying the developmentally related ssTnI gene regulation is not clear. In this study, we have explored the epigenetic role of DNA methylation in the regulation of ssTnI expression in the heart during the development. Results The DNA methylation levels of CpG island and CpG dinucleotides region were detected using methylation specific PCR (MSP) and bisulfite sequence PCR (BSP) in 2000 bp upstream and 100 bp upstream of ssTnI gene promoter. Real time RT-PCR and Western blot were used to detect ssTnI mRNA and protein expression levels. We found that DNA methylation levels of the CpG dinucleotides region in ssTnI gene promoter were increased with the development, corresponding to a decreased expression of ssTnI gene in mouse heart. However the DNA methylation levels of CpG islands in this gene were not changed during the development. Application of a methylation inhibitor, 5-Azacytidine, in cultured myocardial cells partially prevented the decline of ssTnI expression. Conclusion Our results indicate that DNA methylation, as an epigenetic intervention, plays a role in the regulation of the fetal TnI gene expression in the heat during the development.
Collapse
Affiliation(s)
- Yang Xu
- Department of Cardiology, Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, P.R. of China
| | - Lingjuan Liu
- Department of Cardiology, Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, P.R. of China.,Ministry of Education Key Laboratory of Child Development and Disorders; Key Laboratory of Pediatrics in Chongqing, CSTC2009CA5002; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, P.R. of China
| | - Bo Pan
- Department of Cardiology, Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, P.R. of China
| | - Jing Zhu
- Ministry of Education Key Laboratory of Child Development and Disorders; Key Laboratory of Pediatrics in Chongqing, CSTC2009CA5002; Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, P.R. of China
| | - Changlong Nan
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL, 33431, USA
| | - Xupei Huang
- Department of Biomedical Science, Charlie E. Schmidt College of Medicine, Florida Atlantic University, 777 Glades Road, Boca Raton, FL, 33431, USA.
| | - Jie Tian
- Department of Cardiology, Heart Centre, Children's Hospital of Chongqing Medical University, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, P.R. of China.
| |
Collapse
|
24
|
Thompson BR, Metzger JM. Cell biology of sarcomeric protein engineering: disease modeling and therapeutic potential. Anat Rec (Hoboken) 2015; 297:1663-9. [PMID: 25125179 DOI: 10.1002/ar.22966] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 12/12/2013] [Accepted: 12/12/2013] [Indexed: 11/09/2022]
Abstract
The cardiac sarcomere is the functional unit for myocyte contraction. Ordered arrays of sarcomeric proteins, held in stoichiometric balance with each other, respond to calcium to coordinate contraction and relaxation of the heart. Altered sarcomeric structure-function underlies the primary basis of disease in multiple acquired and inherited heart disease states. Hypertrophic and restrictive cardiomyopathies are caused by inherited mutations in sarcomeric genes and result in altered contractility. Ischemia-mediated acidosis directly alters sarcomere function resulting in decreased contractility. In this review, we highlight the use of acute genetic engineering of adult cardiac myocytes through stoichiometric replacement of sarcomeric proteins in these disease states with particular focus on cardiac troponin I. Stoichiometric replacement of disease causing mutations has been instrumental in defining the molecular mechanisms of hypertrophic and restrictive cardiomyopathy in a cellular context. In addition, taking advantage of stoichiometric replacement through gene therapy is discussed, highlighting the ischemia-resistant histidine-button, A164H cTnI. Stoichiometric replacement of sarcomeric proteins offers a potential gene therapy avenue to replace mutant proteins, alter sarcomeric responses to pathophysiologic insults, or neutralize altered sarcomeric function in disease.
Collapse
Affiliation(s)
- Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | | |
Collapse
|
25
|
Abstract
Traditional methods for DNA transfection are often inefficient and toxic for terminally differentiated cells, such as cardiac myocytes. Vector-based gene transfer is an efficient approach for introducing exogenous cDNA into these types of primary cell cultures. In this chapter, separate protocols for adult rat cardiac myocyte isolation and gene transfer with recombinant adenovirus are provided and are routinely utilized for studying the effects of sarcomeric proteins on myofilament function.
Collapse
|
26
|
Thompson BR, Houang EM, Sham YY, Metzger JM. Molecular determinants of cardiac myocyte performance as conferred by isoform-specific TnI residues. Biophys J 2014; 106:2105-14. [PMID: 24853739 DOI: 10.1016/j.bpj.2014.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/14/2014] [Accepted: 04/04/2014] [Indexed: 11/27/2022] Open
Abstract
Troponin I (TnI) is the molecular switch of the sarcomere. Cardiac myocytes express two isoforms of TnI during development. The fetal heart expresses the slow skeletal TnI (ssTnI) isoform and shortly after birth ssTnI is completely and irreversibly replaced by the adult cardiac TnI (cTnI) isoform. These two isoforms have important functional differences; broadly, ssTnI is a positive inotrope, especially under acidic/hypoxic conditions, whereas cTnI facilitates faster relaxation performance. Evolutionary directed changes in cTnI sequence suggest cTnI evolved to favor relaxation performance in the mammalian heart. To investigate the mechanism, we focused on several notable TnI isoform and trans-species-specific residues located in TnI's helix 4 using structure/function and molecular dynamics analyses. Gene transduction of adult cardiac myocytes by cTnIs with specific helix 4 ssTnI substitutions, Q157R/A164H/E166V/H173N (QAEH), and A164H/H173N (AH), were investigated. cTnI QAEH is similar in these four residues to ssTnI and nonmammalian chordate cTnIs, whereas cTnI AH is similar to fish cTnI in these four residues. In comparison to mammalian cTnI, cTnI QAEH and cTnI AH showed increased contractility and slowed relaxation, which functionally mimicked ssTnI expressing myocytes. cTnI QAEH molecular dynamics simulations demonstrated altered intermolecular interactions between TnI helix 4 and cTnC helix A, specifically revealing a new, to our knowledge, electrostatic interaction between R171of cTnI and E15 of cTnC, which structurally phenocopied the ssTnI conformation. Free energy perturbation calculation of cTnC Ca(2+) binding for these conformations showed relative increased calcium binding for cTnI QAEH compared to cTnI. Taken together, to our knowledge, these new findings provide evidence that the evolutionary-directed coordinated acquisition of residues Q157, A164, E166, H173 facilitate enhanced relaxation performance in mammalian adult cardiac myocytes.
Collapse
Affiliation(s)
- Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Evelyne M Houang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota; Center for Drug Design, University of Minnesota Academic Health Center, Minneapolis, Minnesota
| | - Yuk Y Sham
- Center for Drug Design, University of Minnesota Academic Health Center, Minneapolis, Minnesota
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota.
| |
Collapse
|
27
|
Acquisition of a quantitative, stoichiometrically conserved ratiometric marker of maturation status in stem cell-derived cardiac myocytes. Stem Cell Reports 2014; 3:594-605. [PMID: 25358788 PMCID: PMC4223713 DOI: 10.1016/j.stemcr.2014.07.012] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/22/2014] [Accepted: 07/24/2014] [Indexed: 12/11/2022] Open
Abstract
There is no consensus in the stem cell field as to what constitutes the mature cardiac myocyte. Thus, helping formalize a molecular signature for cardiac myocyte maturation would advance the field. In the mammalian heart, inactivation of the “fetal” TNNI gene, TNNI1 (ssTnI), together in temporal concert with its stoichiometric replacement by the adult TNNI gene product, TNNI3 (cTnI), represents a quantifiable ratiometric maturation signature. We examined the TNNI isoform transition in human induced pluripotent stem cell (iPSC) cardiac myocytes (hiPSC-CMs) and found the fetal TNNI signature, even during long-term culture. Rodent stem cell-derived and primary myocytes, however, transitioned to the adult TnI profile. Acute genetic engineering of hiPSC-CMs enabled a rapid conversion toward the mature TnI profile. While there is no single marker to denote the mature cardiac myocyte, we propose that tracking the cTnI:ssTnI protein isoform ratio provides a valuable maturation signature to quantify myocyte maturation status across laboratories. The TNNI gene switch is a quantitative maturation signal for hiPSC-CMs TnI isoform ratio is necessary, but not sufficient, to establish the mature state TNNI protein isoform switching is stalled in hiPSC-CMs Gene transfer enables acquisition of the mature TNNI signature in hiPSC-CMs
Collapse
|
28
|
Robertson IM, Pineda-Sanabria SE, Holmes PC, Sykes BD. Conformation of the critical pH sensitive region of troponin depends upon a single residue in troponin I. Arch Biochem Biophys 2014; 552-553:40-9. [DOI: 10.1016/j.abb.2013.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 11/18/2013] [Accepted: 12/05/2013] [Indexed: 12/20/2022]
|
29
|
The art of the deal in myofilament modulation of function. J Mol Cell Cardiol 2014; 72:238-40. [PMID: 24732213 DOI: 10.1016/j.yjmcc.2014.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/02/2014] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
|
30
|
Katrukha IA. Human cardiac troponin complex. Structure and functions. BIOCHEMISTRY (MOSCOW) 2014; 78:1447-65. [DOI: 10.1134/s0006297913130063] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Ford SJ, Chandra M. The effects of slow skeletal troponin I expression in the murine myocardium are influenced by development-related shifts in myosin heavy chain isoform. J Physiol 2012; 590:6047-63. [PMID: 22966157 DOI: 10.1113/jphysiol.2012.240085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Troponin I (TnI) and myosin heavy chain (MHC) are two contractile regulatory proteins that undergo major shifts in isoform expression as cardiac myocytes mature from embryonic to adult stages. To date, many studies have investigated individual effects of embryonic vs. cardiac isoforms of either TnI or MHC on cardiac muscle function and contractile dynamics. Thus, we sought to determine whether concomitant expression of the embryonic isoforms of both TnI and MHC had functional effects that were not previously observed. Adult transgenic (TG) mice that express the embryonic isoform of TnI, slow skeletal TnI (ssTnI), were treated with propylthiouracil (PTU) to revert MHC expression from adult (α-MHC) to embryonic (β-MHC) isoforms. Cardiac muscle fibres from these mice contained ∼80% β-MHC and ∼34% ssTnI of total MHC or TnI, respectively, allowing us to test the functional effects of ssTnI in the presence of β-MHC. Detergent-skinned cardiac muscle fibre bundles were used to study how the interplay between MHC and TnI modulate muscle length-mediated effect on crossbridge (XB) recruitment dynamics, Ca(2+)-activated tension, and ATPase activity. One major finding was that the model-predicted XB recruitment rate (b) was enhanced significantly by ssTnI, and this speeding effect of ssTnI on XB recruitment rate was much greater (3.8-fold) when β-MHC was present. Another major finding was that the previously documented ssTnI-mediated increase in myofilament Ca(2+) sensitivity (pCa(50)) was blunted when β-MHC was present. ssTnI expression increased pCa(50) by 0.33 in α-MHC fibres, whereas ssTnI increased pCa(50) by only 0.05 in β-MHC fibres. Our study provides new evidence for significant interplay between MHC and TnI isoforms that is essential for tuning cardiac contractile function. Thus, MHC-TnI interplay may provide a developmentally dependent mechanism to enhance XB recruitment dynamics at a time when Ca(2+)-handling mechanisms are underdeveloped, and to prevent excessive ssTnI-dependent inotropy (increased Ca(2+) sensitivity) in the embryonic myocardium.
Collapse
Affiliation(s)
- Steven J Ford
- Department of Veterinary and Comparative Anatomy, Pharmacology, and Physiology (VCAPP), Washington State University, Pullman, WA 99164, USA.
| | | |
Collapse
|
32
|
Davis J, Yasuda S, Palpant NJ, Martindale J, Stevenson T, Converso K, Metzger JM. Diastolic dysfunction and thin filament dysregulation resulting from excitation-contraction uncoupling in a mouse model of restrictive cardiomyopathy. J Mol Cell Cardiol 2012; 53:446-57. [PMID: 22683325 DOI: 10.1016/j.yjmcc.2012.05.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 05/22/2012] [Accepted: 05/29/2012] [Indexed: 10/28/2022]
Abstract
Restrictive cardiomyopathy (RCM) has been linked to mutations in the thin filament regulatory protein cardiac troponin I (cTnI). As the pathogenesis of RCM from genotype to clinical phenotype is not fully understood, transgenic (Tg) mice were generated with cardiac specific expression of an RCM-linked missense mutation (R193H) in cTnI. R193H Tg mouse hearts with 15% stoichiometric replacement had smaller hearts and significantly elevated end diastolic pressures (EDP) in vivo. Using a unique carbon microfiber-based assay, membrane intact R193H adult cardiac myocytes generated higher passive tensions across a range of physiologic sarcomere lengths resulting in significant Ca(2+) independent cellular diastolic tone that was manifest in vivo as elevated organ-level EDP. Sarcomere relaxation and Ca(2+) decay was uncoupled in isolated R193H Tg adult myocytes due to the increase in myofilament Ca(2+) sensitivity of tension, decreased passive compliance of the sarcomere, and adaptive in vivo changes in which phospholamban (PLN) content was decreased. Further evidence of Ca(2+) and mechanical uncoupling in R193H Tg myocytes was demonstrated by the biphasic response of relaxation to increased pacing frequency versus the negative staircase seen with Ca(2+) decay. In comparison, non-transgenic myocyte relaxation closely paralleled the accelerated Ca(2+) decay. Ca(2+) transient amplitude was also significantly blunted in R193H Tg myocytes despite normal mechanical shortening resulting in myocyte hypercontractility when compared to non-transgenics. These results identify for the first time that a single point mutation in cTnI, R193H, directly causes elevated EDP due to a myocyte intrinsic loss of compliance independent of Ca(2+) cycling or altered cardiac morphology. The compound influence of impaired relaxation and elevated EDP represents a clinically severe form of diastolic dysfunction similar to the hemodynamic state documented in RCM patients.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Liu B, Lee RS, Biesiadecki BJ, Tikunova SB, Davis JP. Engineered troponin C constructs correct disease-related cardiac myofilament calcium sensitivity. J Biol Chem 2012; 287:20027-36. [PMID: 22511780 DOI: 10.1074/jbc.m111.334953] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aberrant myofilament Ca(2+) sensitivity is commonly observed with multiple cardiac diseases, especially familial cardiomyopathies. Although the etiology of the cardiomyopathies remains unclear, improving cardiac muscle Ca(2+) sensitivity through either pharmacological or genetic approaches shows promise of alleviating the disease-related symptoms. Due to its central role as the Ca(2+) sensor for cardiac muscle contraction, troponin C (TnC) stands out as an obvious and versatile target to reset disease-associated myofilament Ca(2+) sensitivity back to normal. To test the hypothesis that aberrant myofilament Ca(2+) sensitivity and its related function can be corrected through rationally engineered TnC constructs, three thin filament protein modifications representing different proteins (troponin I or troponin T), modifications (missense mutation, deletion, or truncation), and disease subtypes (familial or acquired) were studied. A fluorescent TnC was utilized to measure Ca(2+) binding to TnC in the physiologically relevant biochemical model system of reconstituted thin filaments. Consistent with the pathophysiology, the restrictive cardiomyopathy mutation, troponin I R192H, and ischemia-induced truncation of troponin I (residues 1-192) increased the Ca(2+) sensitivity of TnC on the thin filament, whereas the dilated cardiomyopathy mutation, troponin T ΔK210, decreased the Ca(2+) sensitivity of TnC on the thin filament. Rationally engineered TnC constructs corrected the abnormal Ca(2+) sensitivities of the thin filament, reconstituted actomyosin ATPase activity, and force generation in skinned trabeculae. Thus, the present study provides a novel and versatile therapeutic strategy to restore diseased cardiac muscle Ca(2+) sensitivity.
Collapse
Affiliation(s)
- Bin Liu
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | |
Collapse
|
34
|
Dynamic reciprocity of sodium and potassium channel expression in a macromolecular complex controls cardiac excitability and arrhythmia. Proc Natl Acad Sci U S A 2012; 109:E2134-43. [PMID: 22509027 DOI: 10.1073/pnas.1109370109] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The cardiac electrical impulse depends on an orchestrated interplay of transmembrane ionic currents in myocardial cells. Two critical ionic current mechanisms are the inwardly rectifying potassium current (I(K1)), which is important for maintenance of the cell resting membrane potential, and the sodium current (I(Na)), which provides a rapid depolarizing current during the upstroke of the action potential. By controlling the resting membrane potential, I(K1) modifies sodium channel availability and therefore, cell excitability, action potential duration, and velocity of impulse propagation. Additionally, I(K1)-I(Na) interactions are key determinants of electrical rotor frequency responsible for abnormal, often lethal, cardiac reentrant activity. Here, we have used a multidisciplinary approach based on molecular and biochemical techniques, acute gene transfer or silencing, and electrophysiology to show that I(K1)-I(Na) interactions involve a reciprocal modulation of expression of their respective channel proteins (Kir2.1 and Na(V)1.5) within a macromolecular complex. Thus, an increase in functional expression of one channel reciprocally modulates the other to enhance cardiac excitability. The modulation is model-independent; it is demonstrable in myocytes isolated from mouse and rat hearts and with transgenic and adenoviral-mediated overexpression/silencing. We also show that the post synaptic density, discs large, and zonula occludens-1 (PDZ) domain protein SAP97 is a component of this macromolecular complex. We show that the interplay between Na(v)1.5 and Kir2.1 has electrophysiological consequences on the myocardium and that SAP97 may affect the integrity of this complex or the nature of Na(v)1.5-Kir2.1 interactions. The reciprocal modulation between Na(v)1.5 and Kir2.1 and the respective ionic currents should be important in the ability of the heart to undergo self-sustaining cardiac rhythm disturbances.
Collapse
|
35
|
pH-responsive titratable inotropic performance of histidine-modified cardiac troponin I. Biophys J 2012; 102:1570-9. [PMID: 22500757 DOI: 10.1016/j.bpj.2012.01.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 01/11/2012] [Accepted: 01/17/2012] [Indexed: 11/22/2022] Open
Abstract
Cardiac troponin I (cTnI) functions as the molecular switch of the thin filament. Studies have shown that a histidine button engineered into cTnI (cTnI A164H) specifically enhances inotropic function in the context of numerous pathophysiological challenges. To gain mechanistic insight into the basis of this finding, we analyzed histidine ionization states in vitro by studying the myofilament biophysics of amino acid substitutions that act as constitutive chemical mimetics of altered histidine ionization. We also assessed the role of histidine-modified cTnI in silico by means of molecular dynamics simulations. A functional in vitro analysis of myocytes at baseline (pH 7.4) indicated similar cellular contractile function and myofilament calcium sensitivity between myocytes expressing wild-type (WT) cTnI and cTnI A164H, whereas the A164R variant showed increased myofilament calcium sensitivity. Under acidic conditions, compared with WT myocytes, the myocytes expressing cTnI A164H maintained a contractile performance similar to that observed for the constitutively protonated cTnI A164R variant. Molecular dynamics simulations showed similar intermolecular atomic contacts between the WT and the deprotonated cTnI A164H variant. In contrast, simulations of protonated cTnI A164H showed various potential structural configurations, one of which included a salt bridge between His-164 of cTnI and Glu-19 of cTnC. This salt bridge was recapitulated in simulations of the cTnI A164R variant. These data suggest that differential histidine ionization may be necessary for cTnI A164H to act as a molecular sensor capable of modulating sarcomere performance in response to changes in the cytosolic milieu.
Collapse
|
36
|
Robertson IM, Holmes PC, Li MX, Pineda-Sanabria SE, Baryshnikova OK, Sykes BD. Elucidation of isoform-dependent pH sensitivity of troponin i by NMR spectroscopy. J Biol Chem 2011; 287:4996-5007. [PMID: 22179777 DOI: 10.1074/jbc.m111.301499] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Myocardial ischemia is characterized by reduced blood flow to cardiomyocytes, which can lead to acidosis. Acidosis decreases the calcium sensitivity and contractile efficiency of cardiac muscle. By contrast, skeletal and neonatal muscles are much less sensitive to changes in pH. The pH sensitivity of cardiac muscle can be reduced by replacing cardiac troponin I with its skeletal or neonatal counterparts. The isoform-specific response of troponin I is dictated by a single histidine, which is replaced by an alanine in cardiac troponin I. The decreased pH sensitivity may stem from the protonation of this histidine at low pH, which would promote the formation of electrostatic interactions with negatively charged residues on troponin C. In this study, we measured acid dissociation constants of glutamate residues on troponin C and of histidine on skeletal troponin I (His-130). The results indicate that Glu-19 comes in close contact with an ionizable group that has a pK(a) of ∼6.7 when it is in complex with skeletal troponin I but not when it is bound to cardiac troponin I. The pK(a) of Glu-19 is decreased when troponin C is bound to skeletal troponin I and the pK(a) of His-130 is shifted upward. These results strongly suggest that these residues form an electrostatic interaction. Furthermore, we found that skeletal troponin I bound to troponin C tighter at pH 6.1 than at pH 7.5. The data presented here provide insights into the molecular mechanism for the pH sensitivity of different muscle types.
Collapse
Affiliation(s)
- Ian M Robertson
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | | | | | | | | | | |
Collapse
|
37
|
Interstitial fluid flow and cyclic strain differentially regulate cardiac fibroblast activation via AT1R and TGF-β1. Exp Cell Res 2011; 318:75-84. [PMID: 22020089 DOI: 10.1016/j.yexcr.2011.10.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2011] [Revised: 09/30/2011] [Accepted: 10/06/2011] [Indexed: 01/31/2023]
Abstract
Cardiac fibroblasts are exposed to both cyclic strain and interstitial fluid flow in the myocardium. The balance of these stimuli is affected by fibrotic scarring, during which the fibroblasts transition to a myofibroblast phenotype. The present study investigates the mechanisms by which cardiac fibroblasts seeded in three-dimensional (3D) collagen gels differentiate between strain and fluid flow. Neonatal cardiac fibroblast-seeded 3D collagen gels were exposed to interstitial flow and/or cyclic strain and message levels of collagens type I and III, transforming growth factor β1 (TGF-β1), and α-smooth muscle actin (α-SMA) were assessed. Flow was found to significantly increase and strain to decrease expression of myofibroblast markers. Corresponding immunofluorescence indicated that flow and strain differentially regulated α-SMA protein expression. The effect of flow was inhibited by exposure to losartan, an angiotensin II type 1 receptor (AT1R) blocker, and by introduction of shRNA constructs limiting AT1R expression. Blocking of TGF-β also inhibited the myofibroblast transition, suggesting that flow-mediated cell signaling involved both AT1R and TGF-β1. Reduced smad2 phosphorylation in response to cyclic strain suggested that TGF-β is part of the mechanism by which cardiac fibroblasts differentiate between strain-induced and flow-induced mechanical stress. Our experiments show that fluid flow and mechanical deformation have distinct effects on cardiac fibroblast phenotype. Our data suggest a mechanism in which fluid flow directly acts on AT1R and causes increased TGF-β1 expression, whereas cyclic strain reduces activation of smad proteins. These results have relevance to the pathogenesis and treatment of heart failure.
Collapse
|
38
|
Auerbach DS, Grzda KR, Furspan PB, Sato PY, Mironov S, Jalife J. Structural heterogeneity promotes triggered activity, reflection and arrhythmogenesis in cardiomyocyte monolayers. J Physiol 2011; 589:2363-81. [PMID: 21486795 DOI: 10.1113/jphysiol.2010.200576] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Patients with structural heart disease are predisposed to arrhythmias by incompletely understood mechanisms. We hypothesized that tissue expansions promote source-to-sink mismatch leading to early after-depolarizations (EADs) and reflection of impulses in monolayers of well-polarized neonatal rat ventricular cardiomyocytes.We traced electrical propagation optically in patterned monolayers consisting of two wide regions connected by a thin isthmus.Structural heterogeneities provided a substrate for EADs, retrograde propagation along the same pathway (reflection) and reentry initiation. Reflection always originated during the action potential (AP) plateau at the distal expansion. To determine whether increased sodium current(INa) would promote EADs, we employed adenoviral transfer of Nav1.5 (Ad-Nav1.5). Compared with uninfected and adenoviral expression of green fluorescent protein (Ad-GFP; viral control),Ad-Nav1.5 significantly increased Nav1.5 protein expression, peak and persistent INa density, A Pupstroke velocity, AP duration, conduction velocity and EAD incidence, as well as reflection incidence (29.2%, n =48 vs. uninfected, 9.4%, n =64; and Ad-GFP, 4.8%, n =21). Likewise,the persistent INa agonist veratridine (0.05–3 μM) prolonged the AP, leading to EADs and reflection. Reflection led to functional reentry distally and bigeminal and trigeminal rhythms proximally. Reflection was rare in the absence of structural heterogeneities.Computer simulations demonstrated the importance of persistent INa in triggering reflection and predicted that the gradient between the depolarizing cells at the distal expansion and the repolarizing cells within the isthmus enabled retrograde flow of depolarizing electrotonic current to trigger EADs and reflection. A combination of a substrate (structural heterogeneity) and a trigger (increased persistent INa and EADs) promotes reflection and arrhythmogenesis.
Collapse
Affiliation(s)
- David S Auerbach
- Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48108, USA
| | | | | | | | | | | |
Collapse
|
39
|
Turner I, Belema-Bedada F, Martindale J, Townsend D, Wang W, Palpant N, Yasuda SC, Barnabei M, Fomicheva E, Metzger JM. Molecular cardiology in translation: gene, cell and chemical-based experimental therapeutics for the failing heart. J Cardiovasc Transl Res 2010; 1:317-27. [PMID: 19956787 DOI: 10.1007/s12265-008-9065-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Acquired and inherited diseases of the heart represent a major health care issue in this country and throughout the World. Clinical medicine has made important advancements in the past quarter century to enable several effective treatment regimes for cardiac patients. Nevertheless, it is apparent that even with the best care, current treatment strategies and therapeutics are inadequate for treating heart disease, leaving it arguably the most pressing health issue today. In this context it is important to seek new approaches to redress the functional deficits in failing myocardium. This review focuses on several recent gene, cell and chemical-based experimental therapeutics currently being developed in the laboratory for potential translation to patient care. For example, new advances in bio-sensing inducible gene expression systems offer the potential for designer cardio-protective proteins to be expressed only during hypoxia/ischemia in the heart. Stem cells continue to offer the promise of cardiac repair, and some recent advances are discussed here. In addition, discovery and applications of synthetic polymers are presented as a chemical-based strategy for acute and chronic treatment of diseased and failing cardiac tissue. Collectively, these approaches serve as the front lines in basic biomedical research, with an eye toward translation of these findings to clinically meaningful applications in cardiac disease.
Collapse
Affiliation(s)
- Immanuel Turner
- Department of Integrative Biology & Physiology, University of Minnesota, Medical School, 6-125 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shaffer JF, Gillis TE. Evolution of the regulatory control of vertebrate striated muscle: the roles of troponin I and myosin binding protein-C. Physiol Genomics 2010; 42:406-19. [PMID: 20484158 DOI: 10.1152/physiolgenomics.00055.2010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Troponin I (TnI) and myosin binding protein-C (MyBP-C) are key regulatory proteins of contractile function in vertebrate muscle. TnI modulates the Ca2+ activation signal, while MyBP-C regulates cross-bridge cycling kinetics. In vertebrates, each protein is distributed as tissue-specific paralogs in fast skeletal (fs), slow skeletal (ss), and cardiac (c) muscles. The purpose of this study is to characterize how TnI and MyBP-C have changed during the evolution of vertebrate striated muscle and how tissue-specific paralogs have adapted to different physiological conditions. To accomplish this we have completed phylogenetic analyses using the amino acid sequences of all known TnI and MyBP-C isoforms. This includes 99 TnI sequences (fs, ss, and c) from 51 different species and 62 MyBP-C sequences from 26 species, with representatives from each vertebrate group. Results indicate that the role of protein kinase A (PKA) and protein kinase C (PKC) in regulating contractile function has changed during the evolution of vertebrate striated muscle. This is reflected in an increased number of phosphorylatable sites in cTnI and cMyBP-C in endothermic vertebrates and the loss of two PKC sites in fsTnI in a common ancestor of mammals, birds, and reptiles. In addition, we find that His132, Val134, and Asn141 in human ssTnI, previously identified as enabling contractile function during cellular acidosis, are present in all vertebrate cTnI isoforms except those from monotremes, marsupials, and eutherian mammals. This suggests that the replacement of these residues with alternative residues coincides with the evolution of endothermy in the mammalian lineage.
Collapse
Affiliation(s)
- Justin F. Shaffer
- Department of Bioengineering, University of Washington, Seattle, Washington; and
| | - Todd E. Gillis
- Department of Integrative Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
41
|
Palpant NJ, Houang EM, Delport W, Hastings KEM, Onufriev AV, Sham YY, Metzger JM. Pathogenic peptide deviations support a model of adaptive evolution of chordate cardiac performance by troponin mutations. Physiol Genomics 2010; 42:287-99. [PMID: 20423961 PMCID: PMC3032286 DOI: 10.1152/physiolgenomics.00033.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 04/23/2010] [Indexed: 11/22/2022] Open
Abstract
In cardiac muscle, the troponin (cTn) complex is a key regulator of myofilament calcium sensitivity because it serves as a molecular switch required for translating myocyte calcium fluxes into sarcomeric contraction and relaxation. Studies of several species suggest that ectotherm chordates have myofilaments with heightened calcium responsiveness. However, genetic polymorphisms in cTn that cause increased myofilament sensitivity to activating calcium in mammals result in cardiac disease including arrhythmias, diastolic dysfunction, and increased susceptibility to sudden cardiac death. We hypothesized that specific residue modifications in the regulatory arm of troponin I (TnI) were critical in mediating the observed decrease in myofilament calcium sensitivity within the mammalian taxa. We performed large-scale phylogenetic analysis, atomic resolution molecular dynamics simulations and modeling, and computational alanine scanning. This study provides evidence that a His to Ala substitution within mammalian cardiac TnI (cTnI) reduced the thermodynamic potential at the interface between cTnI and cardiac TnC (cTnC) in the calcium-saturated state by disrupting a strong intermolecular electrostatic interaction. This key residue modification reduced myofilament calcium sensitivity by making cTnI molecularly untethered from cTnC. To meet the requirements for refined mammalian adult cardiac performance, we propose that compensatory evolutionary pressures favored mutations that enhanced the relaxation properties of cTn by decreasing its sensitivity to activating calcium.
Collapse
Affiliation(s)
- Nathan J Palpant
- Department of Integrative Biology and Physiology, University of Minnesota Academic Health Center, 321 Church Street SE, Minneapolis, MN 55455, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Ke Y, Sheehan KA, Egom EEA, Lei M, Solaro RJ. Novel bradykinin signaling in adult rat cardiac myocytes through activation of p21-activated kinase. Am J Physiol Heart Circ Physiol 2010; 298:H1283-9. [PMID: 20154261 PMCID: PMC2853422 DOI: 10.1152/ajpheart.01070.2009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Although bradykinin (BK) is known to exert effects on the myocardium, its intracellular signaling pathways remain poorly understood. Experiments in other cell types indicated that p21-activated kinase-1 (Pak1), a Ser/Thr kinase downstream of small monomeric G proteins, is activated by BK. We previously reported that the expression of active Pak1 in adult cardiac myocytes induced activation of protein phosphatase 2A and dephosphorylation of myofilament proteins (Ke et al. Circ Res 94: 194–200, 2004). In experiments reported here, we tested the hypothesis that BK signals altered protein phosphorylation in adult rat cardiac myocytes through the activation and translocation of Pak1. Treatment of myocytes with BK resulted in the activation of Pak1 as demonstrated by increased autophosphorylation at Thr423 and a diminished striated localization, which is present in the basal state. BK induced dephosphorylation of both cardiac troponin I and phospholamban. Treatment of isolated myocytes with BK also blunted the effect of isoproterenol to enhance peak Ca2+ and relaxation of Ca2+ transients. Protein phosphatase 2A was demonstrated to associate with both Pak 1 and phospholamban. Our studies indicate a novel signaling mechanism for BK in adult rat cardiac myocytes and support our hypothesis that Pak 1 is a significant regulator of phosphatase activity in the heart.
Collapse
Affiliation(s)
- Yunbo Ke
- Department of Physiology and Biophysics and Center for Cardiovascular Research, University of Illinois at Chicago, USA
| | | | | | | | | |
Collapse
|
43
|
Davis J, Metzger JM. Combinatorial effects of double cardiomyopathy mutant alleles in rodent myocytes: a predictive cellular model of myofilament dysregulation in disease. PLoS One 2010; 5:e9140. [PMID: 20161772 PMCID: PMC2818843 DOI: 10.1371/journal.pone.0009140] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 01/19/2010] [Indexed: 12/05/2022] Open
Abstract
Inherited cardiomyopathy (CM) represents a diverse group of cardiac muscle diseases that present with a broad spectrum of symptoms ranging from benign to highly malignant. Contributing to this genetic complexity and clinical heterogeneity is the emergence of a cohort of patients that are double or compound heterozygotes who have inherited two different CM mutant alleles in the same or different sarcomeric gene. These patients typically have early disease onset with worse clinical outcomes. Little experimental attention has been directed towards elucidating the physiologic basis of double CM mutations at the cellular-molecular level. Here, dual gene transfer to isolated adult rat cardiac myocytes was used to determine the primary effects of co-expressing two different CM-linked mutant proteins on intact cardiac myocyte contractile physiology. Dual expression of two CM mutants, that alone moderately increase myofilament activation, tropomyosin mutant A63V and cardiac troponin mutant R146G, were shown to additively slow myocyte relaxation beyond either mutant studied in isolation. These results were qualitatively similar to a combination of moderate and strong activating CM mutant alleles alphaTmA63V and cTnI R193H, which approached a functional threshold. Interestingly, a combination of a CM myofilament deactivating mutant, troponin C G159D, together with an activating mutant, cTnIR193H, produced a hybrid phenotype that blunted the strong activating phenotype of cTnIR193H alone. This is evidence of neutralizing effects of activating/deactivating mutant alleles in combination. Taken together, this combinatorial mutant allele functional analysis lends molecular insight into disease severity and forms the foundation for a predictive model to deconstruct the myriad of possible CM double mutations in presenting patients.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, United States of America
| |
Collapse
|
44
|
Hoffman RMB, Sykes BD. Isoform-specific variation in the intrinsic disorder of troponin I. Proteins 2009; 73:338-50. [PMID: 18433059 DOI: 10.1002/prot.22063] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Various intrinsic disorder (ID) prediction algorithms were applied to the three tissue isoforms of troponin I (TnI). The results were interpreted in terms of the known structure and dynamics of troponin. In line with previous results, all isoforms of TnI were predicted to have large stretches of ID. The predictions show that the C-termini of all isoforms are extensively disordered as is the N-terminal extension of the cardiac isoform. Cardiac TnI likely belongs to the group of intrinsically disordered signalling hub proteins. For a given portion of the protein sequence, most ID prediction approaches indicate isoform-dependent variations in the probability of disorder. Comparison of machine learning and physically based approaches suggests the ID variations are only partially attributable to local variations in the ratio of charged to hydrophobic residues. The VSL2B algorithm predicts the largest variations in ID across the isoforms, with the cardiac isoform having the highest probability of structured regions, and the fast-skeletal isoform having no intrinsic structure. The region corresponding to residues 57-95 of the fast-skeletal isoform, known to form a coiled coil substructure with troponin T, was highly variable between isoforms. The isoform-specific ID variations may have mechanistic significance, modulating the extent to which conformational fluctuations in tropomyosin are communicated to the troponin complex. We discuss structural mechanisms for this communication. Overall, the results motivate the development of predictors designed to address relative levels of disorder between highly similar proteins.
Collapse
Affiliation(s)
- Ryan M B Hoffman
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
45
|
Palpant NJ, D'Alecy LG, Metzger JM. Single histidine button in cardiac troponin I sustains heart performance in response to severe hypercapnic respiratory acidosis in vivo. FASEB J 2009; 23:1529-40. [PMID: 19141534 DOI: 10.1096/fj.08-121996] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Intracellular acidosis is a profound negative regulator of myocardial performance. We hypothesized that titrating myofilament calcium sensitivity by a single histidine substituted cardiac troponin I (A164H) would protect the whole animal physiological response to acidosis in vivo. To experimentally induce severe hypercapnic acidosis, mice were exposed to a 40% CO(2) challenge. By echocardiography, it was found that systolic function and ventricular geometry were maintained in cTnI A164H transgenic (Tg) mice. By contrast, non-Tg (Ntg) littermates experienced rapid and marked cardiac decompensation during this same challenge. For detailed hemodymanic assessment, Millar pressure-conductance catheterization was performed while animals were treated with a beta-blocker, esmolol, during a severe hypercapnic acidosis challenge. Survival and load-independent measures of contractility were significantly greater in Tg vs. Ntg mice. This assay showed that Ntg mice had 100% mortality within 5 min of acidosis. By contrast, systolic and diastolic function were protected in Tg mice during acidosis, and they had 100% survival. This study shows that, independent of any beta-adrenergic compensation, myofilament-based molecular manipulation of inotropy by histidine-modified troponin I maintains cardiac inotropic and lusitropic performance and markedly improves survival during severe acidosis in vivo.
Collapse
Affiliation(s)
- Nathan J Palpant
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
46
|
Davis J, Westfall MV, Townsend D, Blankinship M, Herron TJ, Guerrero-Serna G, Wang W, Devaney E, Metzger JM. Designing heart performance by gene transfer. Physiol Rev 2008; 88:1567-651. [PMID: 18923190 DOI: 10.1152/physrev.00039.2007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The birth of molecular cardiology can be traced to the development and implementation of high-fidelity genetic approaches for manipulating the heart. Recombinant viral vector-based technology offers a highly effective approach to genetically engineer cardiac muscle in vitro and in vivo. This review highlights discoveries made in cardiac muscle physiology through the use of targeted viral-mediated genetic modification. Here the history of cardiac gene transfer technology and the strengths and limitations of viral and nonviral vectors for gene delivery are reviewed. A comprehensive account is given of the application of gene transfer technology for studying key cardiac muscle targets including Ca(2+) handling, the sarcomere, the cytoskeleton, and signaling molecules and their posttranslational modifications. The primary objective of this review is to provide a thorough analysis of gene transfer studies for understanding cardiac physiology in health and disease. By comparing results obtained from gene transfer with those obtained from transgenesis and biophysical and biochemical methodologies, this review provides a global view of cardiac structure-function with an eye towards future areas of research. The data presented here serve as a basis for discovery of new therapeutic targets for remediation of acquired and inherited cardiac diseases.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Palpant NJ, Day SM, Herron TJ, Converso KL, Metzger JM. Single histidine-substituted cardiac troponin I confers protection from age-related systolic and diastolic dysfunction. Cardiovasc Res 2008; 80:209-18. [PMID: 18635554 DOI: 10.1093/cvr/cvn198] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Contractile dysfunction associated with myocardial ischaemia is a significant cause of morbidity and mortality in the elderly. Strategies to protect the aged heart from ischaemia-mediated pump failure are needed. We hypothesized that troponin I-mediated augmentation of myofilament calcium sensitivity would protect cardiac function in aged mice. METHODS AND RESULTS To address this, we investigated transgenic (Tg) mice expressing a histidine-substituted form of adult cardiac troponin I (cTnI A164H), which increases myofilament calcium sensitivity in a pH-dependent manner. Serial echocardiography revealed that Tg hearts showed significantly improved systolic function at 4 months, which was sustained for 2 years based on ejection fraction and velocity of circumferential fibre shortening. Age-related diastolic dysfunction was also attenuated in Tg mice as assessed by Doppler measurements of the mitral valve inflow and lateral annulus Doppler tissue imaging. During acute hypoxia, cardiac contractility significantly improved in aged Tg mice made evident by increased stroke volume, end systolic pressure, and +dP/dt compared with non-transgenic mice. CONCLUSION This study shows that increasing myofilament function by means of a pH-responsive histidine button engineered into cTnI results in enhanced baseline heart function in Tg mice over their lifetime, and during acute hypoxia improves survival in aged mice by maintaining cardiac contractility.
Collapse
Affiliation(s)
- Nathan J Palpant
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, 1301 E. Catherine Street, 7727 Medical Science II, Ann Arbor, MI 48109-0622, USA
| | | | | | | | | |
Collapse
|
48
|
Day SM, Coutu P, Wang W, Herron T, Turner I, Shillingford M, Lacross NC, Converso KL, Piao L, Li J, Lopatin AN, Metzger JM. Cardiac-directed parvalbumin transgene expression in mice shows marked heart rate dependence of delayed Ca2+ buffering action. Physiol Genomics 2008; 33:312-22. [PMID: 18334547 DOI: 10.1152/physiolgenomics.00302.2007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Relaxation abnormalities are prevalent in heart failure and contribute to clinical outcomes. Disruption of Ca2+ homeostasis in heart failure delays relaxation by prolonging the intracellular Ca2+ transient. We sought to speed cardiac relaxation in vivo by cardiac-directed transgene expression of parvalbumin (Parv), a cytosolic Ca2+ buffer normally expressed in fast-twitch skeletal muscle. A key feature of Parv's function resides in its Ca2+/Mg2+ binding affinities that account for delayed Ca2+ buffering in response to the intracellular Ca2+ transient. Cardiac Parv expression decreased sarcoplasmic reticulum Ca2+ content without otherwise altering intracellular Ca2+ homeostasis. At high physiological mouse heart rates in vivo, Parv modestly accelerated relaxation without affecting cardiac morphology or systolic function. Ex vivo pacing of the isolated heart revealed a marked heart rate dependence of Parv's delayed Ca2+ buffering effects on myocardial performance. As the pacing frequency was lowered (7 to 2.5 Hz), the relaxation rates increased in Parv hearts. However, as pacing rates approached the dynamic range in humans, Parv hearts demonstrated decreased contractility, consistent with Parv buffering systolic Ca2+. Mathematical modeling and in vitro studies provide the underlying mechanism responsible for the frequency-dependent fractional Ca2+ buffering action of Parv. Future studies directed toward refining the dose and frequency-response relationships of Parv in the heart or engineering novel Parv-based Ca2+ buffers with modified Mg2+ and Ca2+ affinities to limit systolic Ca2+ buffering may hold promise for the development of new therapies to remediate relaxation abnormalities in heart failure.
Collapse
Affiliation(s)
- Sharlene M Day
- Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0644, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Westfall MV, Metzger JM. Single amino acid substitutions define isoform-specific effects of troponin I on myofilament Ca2+ and pH sensitivity. J Mol Cell Cardiol 2007; 43:107-18. [PMID: 17602701 PMCID: PMC2043486 DOI: 10.1016/j.yjmcc.2007.05.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 04/23/2007] [Accepted: 05/15/2007] [Indexed: 11/25/2022]
Abstract
Troponin I isoforms play a key role in determining myofilament Ca2+ sensitivity in cardiac muscle. The goal here was to identify domain clusters and residues that confer troponin I isoform-specific myofilament Ca2+ and pH sensitivities of contraction. Key domains/residues that contribute to troponin I isoform-specific Ca2+ and pH sensitivity were studied using gene transfer of a slow skeletal troponin I (ssTnI) template, with targeted cardiac troponin I (cTnI) residue substitutions. Substitutions in ssTnI with cognate cTnI residues R125Q, H132A, and V134E, studied both independently and together (ssTnIQAE), resulted in efficient stoichiometric replacement of endogenous myofilament cTnI in adult cardiac myocytes. In permeabilized myocytes, the pCa50 of tension ([Ca2+] required for half maximal force), and the acidosis-induced rightward shift of pCa50 were converted to the cTnI phenotype in myocytes expressing ssTnIQAE or ssTnIH132A, and there was no functionally additive effect of ssTnIQAE versus ssTnIH132A. Interestingly, only the acidosis-induced shift in Ca2+ sensitivity was comparable to cTnI in myocytes expressing ssTnIV134E, while ssTnIR125Q fully retained the ssTnI phenotype. An additional ssTnIN141H substitution, which lies within the same structural region of TnI as V134, produced a shift in myofilament Ca2+ sensitivity comparable to cTnI at physiological pH, while the acidic pH response was similar to the effect of wild-type ssTnI. Analysis of sarcomere shortening in intact adult cardiac myocytes was consistent with the force measurements. Targeted substitutions in the carboxyl portion of TnI produced residue-specific influences on myofilament Ca2+ and pH sensitivity of force and give new molecular insights into the TnI isoform dependence of myofilament function.
Collapse
Affiliation(s)
- Margaret V Westfall
- Department of Surgery, Cardiac Surgery Section, University of Michigan, 1150 W. Medical Center Drive, B560 MSRB II, Ann Arbor, MI 48109-0686, USA.
| | | |
Collapse
|
50
|
Yasuda SI, Coutu P, Sadayappan S, Robbins J, Metzger JM. Cardiac transgenic and gene transfer strategies converge to support an important role for troponin I in regulating relaxation in cardiac myocytes. Circ Res 2007; 101:377-86. [PMID: 17615373 DOI: 10.1161/circresaha.106.145557] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Elucidating the relative roles of cardiac troponin I (cTnI) and phospholamban (PLN) in beta-adrenergic-mediated hastening of cardiac relaxation has been challenging and controversial. To test the hypothesis that beta-adrenergic phosphorylation of cTnI has a prominent role in accelerating cardiac myocyte relaxation performance we used transgenic (Tg) mice bearing near complete replacement of native cTnI with a beta-adrenergic phospho-mimetic of cTnI whereby tandem serine codons 23/24 were converted to aspartic acids (cTnI S23/24D). Adult cardiac myocytes were isolated and contractility determined at physiological temperature under unloaded and loaded conditions using micro-carbon fibers. At baseline, cTnI S23/24D myocytes had significantly faster relaxation times relative to controls, and isoproterenol stimulation (Iso) had only a small effect to further speed relaxation in cTnI S23/24D myocytes (delta Iso: 7.2 ms) relative to the maximum Iso effect (31.2 ms) in control. The Ca(2+) transient decay rate was similarly accelerated by Iso in Tg and nontransgenic (Ntg) myocytes. Gene transfer of cTnI S23/24D to myocytes in primary culture showed comparable findings. Gene transfer of cTnI with both serines 23/24 converted to alanines (cTnI S23/24A), or gene transfer of slow skeletal TnI, both of which lack PKA phosphorylation sites, significantly blunted Iso-mediated enhanced relaxation compared with controls. Gene transfer of wild-type cTnI had no effect on relaxation. These findings support a key role of cTnI in myocyte relaxation and highlight a direct contribution of the myofilaments in modulating the dynamics of myocardial performance.
Collapse
Affiliation(s)
- So-ichiro Yasuda
- Department of Molecular and Integrative Physiology, 7730 Medical Science II, University of Michigan, Ann Arbor, MI 48109-0622, USA
| | | | | | | | | |
Collapse
|