1
|
Yang D, Sun W, Gao L, Zhao K, Zhuang Q, Cai Y. Cell competition as an emerging mechanism and therapeutic target in cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167769. [PMID: 40054587 DOI: 10.1016/j.bbadis.2025.167769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/18/2025] [Accepted: 02/27/2025] [Indexed: 03/17/2025]
Abstract
Cell competition, as an internal quality control mechanism that constantly monitor cell fitness and eliminate unfit cells, maintains proper embryogenesis and tissue integrity during early development and adult homeostasis. Recent studies have revealed that cell competition functions as a tumor-suppressive mechanism to defend against cancer by removing neoplastic cell, which however, is hijacked by tumor cells and drive cell competition in favor of mutant cells, thereby promoting cancer initiation and progression. In this review, with a special focus on mammalian systems, we discuss the latest insights into the mechanisms regulating cell competition and its dual role in tumor development. We also provide current strategies to modulate the direction of cell competition for the prevention and treatment of cancers.
Collapse
Affiliation(s)
- Dakai Yang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jintan, People's Republic of China; Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China.
| | - Wenyue Sun
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Lu Gao
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, People's Republic of China
| | - Kai Zhao
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jintan, People's Republic of China
| | - Qin Zhuang
- Department of General Practice, Affiliated Hospital of Jiangsu University, Zhenjiang, People's Republic of China.
| | - Yun Cai
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jintan, People's Republic of China.
| |
Collapse
|
2
|
Xie L, Jakutis G, Dooley CM, Guenther S, Kontarakis Z, Howard SP, Juan T, Stainier DYR. Induction of a transcriptional adaptation response by RNA destabilization events. EMBO Rep 2025; 26:2262-2279. [PMID: 40128410 PMCID: PMC12069562 DOI: 10.1038/s44319-025-00427-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025] Open
Abstract
Transcriptional adaptation (TA) is a cellular process whereby mRNA-destabilizing mutations are associated with the transcriptional upregulation of so-called adapting genes. The nature of the TA-triggering factor(s) remains unclear, namely whether an mRNA-borne premature termination codon or the subsequent mRNA decay process, and/or its products, elicits TA. Here, working with mouse Actg1, we first establish two types of perturbations that lead to mRNA destabilization: Cas9-induced mutations predicted to lead to mutant mRNA decay, and Cas13d-mediated mRNA cleavage. We find that both types of perturbations are effective in degrading Actg1 mRNA, and that they both upregulate Actg2. Notably, increased chromatin accessibility at the Actg2 locus was observed only in the Cas9-induced mutant cells but not in the Cas13d-targeted cells, suggesting that chromatin remodeling is not required for Actg2 upregulation. We further show that ribozyme-mediated Actg1 pre-mRNA cleavage also leads to a robust upregulation of Actg2, and that this upregulation is again independent of chromatin remodeling. Together, these data highlight the critical role of RNA destabilization events as a trigger for TA, or at least a TA-like response.
Collapse
Affiliation(s)
- Lihan Xie
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Gabrielius Jakutis
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Christopher M Dooley
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Stefan Guenther
- ECCPS Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Zacharias Kontarakis
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
- Genome Engineering and Measurement Laboratory (GEML), Eidgenössische Technische Hochschule (ETH) Zürich, Zürich, Switzerland
- Functional Genomics Center Zürich, ETH Zürich/University of Zürich, Zürich, 8057, Switzerland
| | - Sarah P Howard
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany
| | - Thomas Juan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Hessen, 61231, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Bad Nauheim, Giessen, Frankfurt, Germany.
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, 75 185, Sweden.
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Hessen, 61231, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Hessen, 61231, Germany.
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Bad Nauheim, Giessen, Frankfurt, Germany.
| |
Collapse
|
3
|
Limyati Y, Sanjaya A, Sebastian R, Gunadi JW, Jasaputra DK, Biben V, Lesmana R. The effects of YAP/TAZ in cardiomyocytes: a scoping review. Mol Biol Rep 2025; 52:392. [PMID: 40232357 DOI: 10.1007/s11033-025-10492-5] [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: 01/16/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
The Hippo signaling pathway, through its effectors' yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), plays a pivotal role in heart development, regeneration, and repair. Despite the well-recognized role of YAP in promoting cardiomyocyte proliferation and differentiation, the underlying mechanisms require further explanation. Therefore, this scoping review was conducted to explore the underlying mechanisms of YAP and TAZ in cardiomyocyte biology. In this scoping review, 138 studies were screened using PRISMA extension for scoping reviews guidelines to examine the upstream regulators, mechanisms, and therapeutic potential of YAP/TAZ in cardiomyocytes. Articles were selected based on relevance to YAP/TAZ signaling in cardiac regeneration and focused on upstream regulators, signaling pathways, and therapeutic applications. Data were extracted using standardized forms, and thematic analysis was performed iteratively. YAP activation regulated several processes, including cardiomyocyte proliferation, differentiation, and protection against oxidative stress. Mechanotransduction factors influence YAP activity, linking the biomechanical environment to cardiac regeneration. Novel upstream regulators, such as prorenin receptors, melatonin, and ERBB2, were identified as YAP/TAZ modulators. Moreover, downstream pathways such as Wnt/β-catenin, PI3K/Akt, and TLR-mediated inflammation confer their effects on cellular proliferation, mitochondrial dynamics, and inflammation. Several therapeutic targets involving YAP that could enhance cardiac regeneration while reducing fibrosis and inflammation were identified. However, significant research gaps remain, including the underexplored role of TAZ, necessity for in vivo studies and transcriptomics to elucidate cell-specific effects, and intricate regulatory networks of YAP/TAZ.
Collapse
Affiliation(s)
- Yenni Limyati
- Doctoral Program, Faculty of Medicine, Universitas Padjadjaran, Bandung, West Java, 40164, Indonesia
- Department of Clinical Skills, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia
| | - Ardo Sanjaya
- Department of Anatomy, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia.
- Maranatha Biomedical Research Laboratory, Maranatha Christian University, Jl. Surya Sumantri No. 65, Bandung, West Java, 40146, Indonesia.
| | - Ray Sebastian
- Undergraduate Program in Medicine, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia
| | - Julia Windi Gunadi
- Maranatha Biomedical Research Laboratory, Maranatha Christian University, Jl. Surya Sumantri No. 65, Bandung, West Java, 40146, Indonesia
- Department of Physiology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia
| | - Diana Krisanti Jasaputra
- Department of Pharmacology, Faculty of Medicine, Maranatha Christian University, Bandung, West Java, 40164, Indonesia
| | - Vitriana Biben
- Department of Physical Medicine and Rehabilitation, Faculty of Medicine, Universitas Padjadjaran/Dr. Hasan Sadikin General Hospital Bandung, West Java, 40161, Indonesia
| | - Ronny Lesmana
- Physiology Molecular, Biological Activity Division, Central Laboratory, Sumedang, West Java, 45363, Indonesia
- Department of Biomedical Science, Faculty of Medicine, Universitas Padjadjaran Bandung, West Java, 40164, Indonesia
| |
Collapse
|
4
|
Koopmans T, van Rooij E. Molecular gatekeepers of endogenous adult mammalian cardiomyocyte proliferation. Nat Rev Cardiol 2025:10.1038/s41569-025-01145-y. [PMID: 40195566 DOI: 10.1038/s41569-025-01145-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
Irreversible cardiac fibrosis, cardiomyocyte death and chronic cardiac dysfunction after myocardial infarction pose a substantial global health-care challenge, with no curative treatments available. To regenerate the injured heart, cardiomyocytes must proliferate to replace lost myocardial tissue - a capability that adult mammals have largely forfeited to adapt to the demanding conditions of life. Using various preclinical models, our understanding of cardiomyocyte proliferation has progressed remarkably, leading to the successful reactivation of cell cycle induction in adult animals, with functional recovery after cardiac injury. Central to this success is the targeting of key pathways and structures that drive cardiomyocyte maturation after birth - nucleation and ploidy, sarcomere structure, developmental signalling, chromatin and epigenetic regulation, the microenvironment and metabolic maturation - forming a complex regulatory framework that allows efficient cellular contraction but restricts cardiomyocyte proliferation. In this Review, we explore the molecular pathways underlying these core mechanisms and how their manipulation can reactivate the cell cycle in cardiomyocytes, potentially contributing to cardiac repair.
Collapse
Affiliation(s)
- Tim Koopmans
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, Netherlands.
- Department of Cardiology, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
5
|
Liu S, Deshmukh V, Meng F, Wang Y, Morikawa Y, Steimle JD, Li RG, Wang J, Martin JF. Microtubules Sequester Acetylated YAP in the Cytoplasm and Inhibit Heart Regeneration. Circulation 2025; 151:59-75. [PMID: 39185559 PMCID: PMC11671299 DOI: 10.1161/circulationaha.123.067646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND The Hippo pathway effector YAP (Yes-associated protein) plays an essential role in cardiomyocyte proliferation and heart regeneration. In response to physiological changes, YAP moves in and out of the nucleus. The pathophysiological mechanisms regulating YAP subcellular localization after myocardial infarction remain poorly defined. METHODS We identified YAP acetylation at site K265 by in vitro acetylation followed by mass spectrometry analysis. We used adeno-associated virus to express YAP-containing mutations that either abolished acetylation (YAP-K265R) or mimicked acetylation (YAP-K265Q) and studied how acetylation regulates YAP subcellular localization in mouse hearts. We generated a cell line with YAP-K265R mutation and investigated the protein-protein interactors by YAP immunoprecipitation followed by mass spectrometry, then validated the YAP interaction in neonatal rat ventricular myocytes. We examined colocalization of YAP and TUBA4A (tubulin α 4A) by superresolution imaging. Furthermore, we developed YAP-K265R and αMHC-MerCreMer (MCM); Yap-loxP/K265R mutant mice to examine the pathophysiological role of YAP acetylation in cardiomyocytes during cardiac regeneration. RESULTS We found that YAP is acetylated at K265 by CBP (CREB-binding protein)/P300 (E1A-binding protein P300) and is deacetylated by nicotinamide phosphoribosyltransferase/nicotinamide adenine dinucleotide/sirtuins axis in cardiomyocytes. After myocardial infarction, YAP acetylation is increased, which promotes YAP cytoplasmic localization. Compared with controls, mice that were genetically engineered to express a K265R mutation that prevents YAP K265 acetylation showed improved cardiac regenerative ability and increased YAP nuclear localization. Mechanistically, YAP acetylation facilitates its interaction with TUBA4A, a component of the microtubule network that sequesters acetylated YAP in the cytoplasm. After myocardial infarction, the microtubule network increased in cardiomyocytes, resulting in the accumulation of YAP in the cytoplasm. CONCLUSIONS After myocardial infarction, decreased sirtuin activity enriches YAP acetylation at K265. The growing TUBA4A network sequesters acetylated YAP within the cytoplasm, which is detrimental to cardiac regeneration.
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH (S.L.)
| | - Vaibhav Deshmukh
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
- Department of Cell Biology & Physiology, Washington University School of Medicine, St Louis, MO (V.D.)
| | - Fansen Meng
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| | | | - Yuka Morikawa
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - Jeffrey D Steimle
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| | - Rich Gang Li
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - Jun Wang
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, (S.L., Y.M., R.G.L., J.W., J.F.M.)
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX (V.D., F.M., J.D.S., J.F.M.)
| |
Collapse
|
6
|
Wu H, Che YN, Lan Q, He YX, Liu P, Chen MT, Dong L, Liu MN. The Multifaceted Roles of Hippo-YAP in Cardiovascular Diseases. Cardiovasc Toxicol 2024; 24:1410-1427. [PMID: 39365552 DOI: 10.1007/s12012-024-09926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 09/23/2024] [Indexed: 10/05/2024]
Abstract
The Hippo-yes-associated protein (YAP) signaling pathway plays a crucial role in cell proliferation, differentiation, and death. It is known to have impact on the progression and development of cardiovascular diseases (CVDs) as well as in the regeneration of cardiomyocytes (CMs). However, further research is needed to understand the molecular mechanisms by which the Hippo-YAP pathway affects the pathological processes of CVDs in order to evaluate its potential clinical applications. In this review, we have summarized the recent findings on the role of the Hippo-YAP pathway in CVDs such as myocardial infarction, heart failure, and cardiomyopathy, as well as its in CM development. This review calls attention to the potential roles of the Hippo-YAP pathway as a relevant target for the future treatment of CVDs.
Collapse
Affiliation(s)
- Hao Wu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yan-Nan Che
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Lan
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Yi-Xiang He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| | - Ming-Tai Chen
- Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, PR China.
| | - Li Dong
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
| | - Meng-Nan Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China.
| |
Collapse
|
7
|
Wang Y, Chatterjee E, Li G, Xu J, Xiao J. Force-sensing protein expression in response to cardiovascular mechanotransduction. EBioMedicine 2024; 110:105412. [PMID: 39481337 PMCID: PMC11554632 DOI: 10.1016/j.ebiom.2024.105412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Force-sensing biophysical cues in microenvironment, including extracellular matrix performances, stretch-mediated mechanics, shear stress and flow-induced hemodynamics, have a significant influence in regulating vascular morphogenesis and cardiac remodeling by mechanotransduction. Once cells perceive these extracellular mechanical stimuli, Piezo activation promotes calcium influx by forming integrin-adhesion-coupling receptors. This induces robust contractility of cytoskeleton structures to further transmit biomechanical alternations into nuclei by regulating Hippo-Yes associated protein (YAP) signaling pathway between cytoplasmic and nuclear translocation. Although biomechanical stimuli are widely studied in cardiovascular diseases, the expression of force-sensing proteins in response to cardiovascular mechanotransduction has not been systematically concluded. Therefore, this review will summarize the force-sensing Piezo, cytoskeleton and YAP proteins to mediate extracellular mechanics, and also give the prominent emphasis on intrinsic connection of these mechanical proteins and cardiovascular mechanotransduction. Extensive insights into cardiovascular mechanics may provide some new strategies for cardiovascular clinical therapy.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai 200135, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
8
|
Ray S, DeSilva C, Dasgupta I, Mana-Capelli S, Cruz-Calderon N, McCollum D. The ability of the LIMD1 and TRIP6 LIM domains to bind strained f-actin is critical for their tension dependent localization to adherens junctions and association with the Hippo pathway kinase LATS1. Cytoskeleton (Hoboken) 2024; 81:436-447. [PMID: 38426816 PMCID: PMC11366040 DOI: 10.1002/cm.21847] [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: 11/17/2023] [Revised: 02/02/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
A key step in regulation of Hippo pathway signaling in response to mechanical tension is recruitment of the LIM domain proteins TRIP6 and LIMD1 to adherens junctions. Mechanical tension also triggers TRIP6 and LIMD1 to bind and inhibit the Hippo pathway kinase LATS1. How TRIP6 and LIMD1 are recruited to adherens junctions in response to tension is not clear, but previous studies suggested that they could be regulated by the known mechanosensory proteins α-catenin and vinculin at adherens junctions. We found that the three LIM domains of TRIP6 and LIMD1 are necessary and sufficient for tension-dependent localization to adherens junctions. The LIM domains of TRIP6, LIMD1, and certain other LIM domain proteins have been shown to bind to actin networks under strain/tension. Consistent with this, we show that TRIP6 and LIMD1 colocalize with the ends of actin fibers at adherens junctions. Point mutations in a key conserved residue in each LIM domain that are predicted to impair binding to f-actin under strain inhibits TRIP6 and LIMD1 localization to adherens junctions and their ability to bind to and recruit LATS1 to adherens junctions. Together these results show that the ability of TRIP6 and LIMD1 to bind to strained actin underlies their ability to localize to adherens junctions and regulate LATS1 in response to mechanical tension.
Collapse
Affiliation(s)
- Samriddha Ray
- Department of Biochemistry & Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, 01605
| | - Chamika DeSilva
- Department of Biochemistry & Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, 01605
| | - Ishani Dasgupta
- Department of Biochemistry & Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, 01605
| | - Sebastian Mana-Capelli
- Department of Biochemistry & Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, 01605
| | - Natasha Cruz-Calderon
- Department of Biochemistry & Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, 01605
| | - Dannel McCollum
- Department of Biochemistry & Molecular Biotechnology, UMass Chan Medical School, Worcester, MA, 01605
| |
Collapse
|
9
|
Chirikian O, Faynus MA, Merk M, Singh Z, Muray C, Pham J, Chialastri A, Vander Roest A, Goldstein A, Pyle T, Lane KV, Roberts B, Smith JE, Gunawardane RN, Sniadecki NJ, Mack DL, Davis J, Bernstein D, Streichan SJ, Clegg DO, Dey SS, Wilson MZ, Pruitt BL. YAP dysregulation triggers hypertrophy by CCN2 secretion and TGFβ uptake in human pluripotent stem cell-derived cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597045. [PMID: 38895282 PMCID: PMC11185505 DOI: 10.1101/2024.06.03.597045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Hypertrophy Cardiomyopathy (HCM) is the most prevalent hereditary cardiovascular disease - affecting >1:500 individuals. Advanced forms of HCM clinically present with hypercontractility, hypertrophy and fibrosis. Several single-point mutations in b-myosin heavy chain (MYH7) have been associated with HCM and increased contractility at the organ level. Different MYH7 mutations have resulted in increased, decreased, or unchanged force production at the molecular level. Yet, how these molecular kinetics link to cell and tissue pathogenesis remains unclear. The Hippo Pathway, specifically its effector molecule YAP, has been demonstrated to be reactivated in pathological hypertrophic growth. We hypothesized that changes in force production (intrinsically or extrinsically) directly alter the homeostatic mechano-signaling of the Hippo pathway through changes in stresses on the nucleus. Using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), we asked whether homeostatic mechanical signaling through the canonical growth regulator, YAP, is altered 1) by changes in the biomechanics of HCM mutant cardiomyocytes and 2) by alterations in the mechanical environment. We use genetically edited hiPSC-CM with point mutations in MYH7 associated with HCM, and their matched controls, combined with micropatterned traction force microscopy substrates to confirm the hypercontractile phenotype in MYH7 mutants. We next modulate contractility in healthy and disease hiPSC-CMs by treatment with positive and negative inotropic drugs and demonstrate a correlative relationship between contractility and YAP activity. We further demonstrate the activation of YAP in both HCM mutants and healthy hiPSC-CMs treated with contractility modulators is through enhanced nuclear deformation. We conclude that the overactivation of YAP, possibly initiated and driven by hypercontractility, correlates with excessive CCN2 secretion (connective tissue growth factor), enhancing cardiac fibroblast/myofibroblast transition and production of known hypertrophic signaling molecule TGFβ. Our study suggests YAP being an indirect player in the initiation of hypertrophic growth and fibrosis in HCM. Our results provide new insights into HCM progression and bring forth a testbed for therapeutic options in treating HCM.
Collapse
|
10
|
Song M, Wang H, Liu C, Jin S, Liu B, Sun W. Non-coding RNAs as regulators of the Hippo pathway in cardiac development and cardiovascular disease. Front Pharmacol 2024; 15:1348280. [PMID: 38698813 PMCID: PMC11063341 DOI: 10.3389/fphar.2024.1348280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Cardiovascular diseases pose a serious threat to human health. The onset of cardiovascular diseases involves the comprehensive effects of multiple genes and environmental factors, and multiple signaling pathways are involved in regulating the occurrence and development of cardiovascular diseases. The Hippo pathway is a highly conserved signaling pathway involved in the regulation of cell proliferation, apoptosis, and differentiation. Recently, it has been widely studied in the fields of cardiovascular disease, cancer, and cell regeneration. Non-coding RNA (ncRNAs), which are important small molecules for the regulation of gene expression in cells, can directly target genes and have diverse regulatory functions. Recent studies have found that ncRNAs interact with Hippo pathway components to regulate myocardial fibrosis, cardiomyocyte proliferation, apoptosis, and hypertrophy and play an important role in cardiovascular disease. In this review, we describe the mode of action of ncRNAs in regulating the Hippo pathway, provide new ideas for further research, and identify molecules involved in the mechanism of action of ncRNAs and the Hippo pathway as potential therapeutic targets, with the aim of finding new modes of action for the treatment and prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Mengyang Song
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Caixia Liu
- Department of Neurology, The Liaoning Province People’s Hospital, Shenyang, China
| | - Sijie Jin
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Wang N, Xu X, Guan F, Zheng Y, Shou Y, Xu T, Shen G, Chen H, Lin Y, Cong W, Jin L, Zhu Z. α-Catenin promotes dermal fibroblasts proliferation and migration during wound healing via FAK/YAP activation. FASEB J 2024; 38:e23410. [PMID: 38193545 DOI: 10.1096/fj.202302251r] [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: 11/01/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024]
Abstract
Skin wound healing is a complex and organized biological process, and the dermal fibroblasts play a crucial role. α-Catenin is known to be involved in regulating various cellular signals, and its role in wound healing remains unclear. Here, we have identified the pivotal role of the α-catenin/FAK/YAP signaling axis in the proliferation and migration of dermal fibroblasts, which contributes to the process of skin wound healing. Briefly, when α-catenin was knocked down specifically in dermal fibroblasts, the wound healing rate is significantly delayed. Moreover, interfering with α-catenin can impede the proliferation and migration of dermal fibroblasts both in vitro and in vivo. Mechanistically, the overexpression of α-catenin upregulates the nuclear accumulation of YAP and transcription of downstream target genes, resulting in enhanced the proliferation and migration of dermal fibroblasts. Furthermore, the FAK Tyr397 phosphorylation inhibitor blocked the promoting effects of α-catenin on YAP activation. Importantly, the continuous phosphorylation mutation of FAK Tyr397 reversed the retardatory effects of α-catenin knockdown on wound healing, by increasing the vitality of fibroblasts. Likewise, α-catenin/FAK was validated as a therapeutic target for wound healing in the db/db chronic trauma model. In summary, our findings have revealed a novel mechanism by which α-catenin facilitates the function of fibroblasts through the activity of the FAK/YAP signaling axis. These findings define a promising therapeutic strategy for accelerating the wound healing process.
Collapse
Affiliation(s)
- Nan Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Xiejun Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Fangqian Guan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yeyi Zheng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yanni Shou
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Tianpeng Xu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Guoxiu Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Hui Chen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yifan Lin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| | - Zhongxin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P.R. China
| |
Collapse
|
12
|
van Ham WB, Meijboom EEM, Ligtermoet ML, Nikkels PGJ, van Veen TAB. Maturation and Function of the Intercalated Disc: Report of Two Pediatric Cases Focusing on Cardiac Development and Myocardial Hyperplasia. J Cardiovasc Dev Dis 2023; 10:354. [PMID: 37623366 PMCID: PMC10455643 DOI: 10.3390/jcdd10080354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023] Open
Abstract
The development of the normal human heart, ranging from gestational age to the mature adult heart, relies on a very delicate and timely orchestrated order of processes. One of the most striking alterations in time is the gradual extinction of the ability for cardiomyocytes to proliferate. Once passing this event, cardiomyocytes grow and increase in contractile strength by means of physiological hypertrophy. This process, importantly, seems to depend on an adequate development of electromechanical coupling that is achieved by the appropriate formation of the intercellular junction named the intercalated disc (ICD). In this report, we describe two sudden death cases of young and apparently healthy-born individuals without external abnormalities compared to an age-matched control. Histological examination, including the comparison with the age-matched and histology-matched controls, showed a disturbed formation of the protein machinery composing the electromechanical junctions at the ICD and an increased nuclei count for both patients. As a cause or consequence, cardiomyocytes in both sudden death cases showed signs of a delayed developmental stage, presumably resulting in an exaggerated degree of hyperplasia.
Collapse
Affiliation(s)
- Willem B. van Ham
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Esmeralda E. M. Meijboom
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Merel L. Ligtermoet
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Peter G. J. Nikkels
- Department of Pathology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Toon A. B. van Veen
- Department of Medical Physiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
13
|
Lee YL, Mathur J, Walter C, Zmuda H, Pathak A. Matrix obstructions cause multiscale disruption in collective epithelial migration by suppressing leader cell function. Mol Biol Cell 2023; 34:ar94. [PMID: 37379202 PMCID: PMC10398892 DOI: 10.1091/mbc.e22-06-0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/06/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
During disease and development, physical changes in extracellular matrix cause jamming, unjamming, and scattering in epithelial migration. However, whether disruptions in matrix topology alter collective cell migration speed and cell-cell coordination remains unclear. We microfabricated substrates with stumps of defined geometry, density, and orientation, which create obstructions for migrating epithelial cells. Here, we show that cells lose their speed and directionality when moving through densely spaced obstructions. Although leader cells are stiffer than follower cells on flat substrates, dense obstructions cause overall cell softening. Through a lattice-based model, we identify cellular protrusions, cell-cell adhesions, and leader-follower communication as key mechanisms for obstruction-sensitive collective cell migration. Our modeling predictions and experimental validations show that cells' obstruction sensitivity requires an optimal balance of cell-cell adhesions and protrusions. Both MDCK (more cohesive) and α-catenin-depleted MCF10A cells were less obstruction sensitive than wild-type MCF10A cells. Together, microscale softening, mesoscale disorder, and macroscale multicellular communication enable epithelial cell populations to sense topological obstructions encountered in challenging environments. Thus, obstruction-sensitivity could define "mechanotype" of cells that collectively migrate yet maintain intercellular communication.
Collapse
Affiliation(s)
- Ye Lim Lee
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Christopher Walter
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Hannah Zmuda
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
14
|
Reitz C, Tavassoli M, Kim D, Shah S, Lakin R, Teng A, Zhou YQ, Li W, Hadipour-Lakmehsari S, Backx P, Emili A, Oudit G, Kuzmanov U, Gramolini A. Proteomics and phosphoproteomics of failing human left ventricle identifies dilated cardiomyopathy-associated phosphorylation of CTNNA3. Proc Natl Acad Sci U S A 2023; 120:e2212118120. [PMID: 37126683 PMCID: PMC10175742 DOI: 10.1073/pnas.2212118120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 03/24/2023] [Indexed: 05/03/2023] Open
Abstract
The prognosis and treatment outcomes of heart failure (HF) patients rely heavily on disease etiology, yet the majority of underlying signaling mechanisms are complex and not fully elucidated. Phosphorylation is a major point of protein regulation with rapid and profound effects on the function and activity of protein networks. Currently, there is a lack of comprehensive proteomic and phosphoproteomic studies examining cardiac tissue from HF patients with either dilated dilated cardiomyopathy (DCM) or ischemic cardiomyopathy (ICM). Here, we used a combined proteomic and phosphoproteomic approach to identify and quantify more than 5,000 total proteins with greater than 13,000 corresponding phosphorylation sites across explanted left ventricle (LV) tissue samples, including HF patients with DCM vs. nonfailing controls (NFC), and left ventricular infarct vs. noninfarct, and periinfarct vs. noninfarct regions of HF patients with ICM. Each pair-wise comparison revealed unique global proteomic and phosphoproteomic profiles with both shared and etiology-specific perturbations. With this approach, we identified a DCM-associated hyperphosphorylation cluster in the cardiomyocyte intercalated disc (ICD) protein, αT-catenin (CTNNA3). We demonstrate using both ex vivo isolated cardiomyocytes and in vivo using an AAV9-mediated overexpression mouse model, that CTNNA3 phosphorylation at these residues plays a key role in maintaining protein localization at the cardiomyocyte ICD to regulate conductance and cell-cell adhesion. Collectively, this integrative proteomic/phosphoproteomic approach identifies region- and etiology-associated signaling pathways in human HF and describes a role for CTNNA3 phosphorylation in the pathophysiology of DCM.
Collapse
Affiliation(s)
- Cristine J. Reitz
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Marjan Tavassoli
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Da Hye Kim
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Saumya Shah
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2R3
| | - Robert Lakin
- Department of Biology, York University, Toronto, ONM3J 1P3
| | - Allen C. T. Teng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Yu-Qing Zhou
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Wenping Li
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Sina Hadipour-Lakmehsari
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Peter H. Backx
- Department of Biology, York University, Toronto, ONM3J 1P3
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston, MA02118
- Department of Biology, Boston University School of Medicine, Boston, MA02118
- The Centre for Network Systems Biology, Boston University School of Medicine, Boston, MA02118
| | - Gavin Y. Oudit
- Department of Medicine, University of Alberta, Edmonton, ABT6G 2R3
- Mazankowski Alberta Heart Institute, Edmonton, ABT6G 2B7
| | - Uros Kuzmanov
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| | - Anthony O. Gramolini
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ONM5S 1M8
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ONM5G 1M1
| |
Collapse
|
15
|
Li X, McLain C, Samuel MS, Olson MF, Radice GL. Actomyosin-mediated cellular tension promotes Yap nuclear translocation and myocardial proliferation through α5 integrin signaling. Development 2023; 150:dev201013. [PMID: 36621002 PMCID: PMC10110499 DOI: 10.1242/dev.201013] [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: 06/09/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023]
Abstract
The cardiomyocyte phenotypic switch from a proliferative to terminally differentiated state results in the loss of regenerative potential of the mammalian heart shortly after birth. Nonmuscle myosin IIB (NM IIB)-mediated actomyosin contractility regulates cardiomyocyte cytokinesis in the embryonic heart, and NM IIB levels decline after birth, suggesting a role for cellular tension in the regulation of cardiomyocyte cell cycle activity in the postnatal heart. To investigate the role of actomyosin contractility in cardiomyocyte cell cycle arrest, we conditionally activated ROCK2 kinase domain (ROCK2:ER) in the murine postnatal heart. Here, we show that α5/β1 integrin and fibronectin matrix increase in response to actomyosin-mediated tension. Moreover, activation of ROCK2:ER promotes nuclear translocation of Yap, a mechanosensitive transcriptional co-activator, and enhances cardiomyocyte proliferation. Finally, we show that reduction of myocardial α5 integrin rescues the myocardial proliferation phenotype in ROCK2:ER hearts. These data demonstrate that cardiomyocytes respond to increased intracellular tension by altering their intercellular contacts in favor of cell-matrix interactions, leading to Yap nuclear translocation, thus uncovering a function for nonmuscle myosin contractility in promoting cardiomyocyte proliferation in the postnatal heart.
Collapse
Affiliation(s)
- Xiaofei Li
- Cardiovascular Research Center, Lifespan Cardiovascular Institute, Rhode Island Hospital, Department of Medicine, Division of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Callie McLain
- Cardiovascular Research Center, Lifespan Cardiovascular Institute, Rhode Island Hospital, Department of Medicine, Division of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Michael S. Samuel
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5000, Australia
| | - Michael F. Olson
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, M5B 2K3 Canada
| | - Glenn L. Radice
- Cardiovascular Research Center, Lifespan Cardiovascular Institute, Rhode Island Hospital, Department of Medicine, Division of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, USA
| |
Collapse
|
16
|
Li R, Huang W. Yes-Associated Protein and Transcriptional Coactivator with PDZ-Binding Motif in Cardiovascular Diseases. Int J Mol Sci 2023; 24:ijms24021666. [PMID: 36675179 PMCID: PMC9861006 DOI: 10.3390/ijms24021666] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Yes-associated protein (YAP, also known as YAP1) and its paralogue TAZ (with a PDZ-binding motif) are transcriptional coactivators that switch between the cytoplasm and nucleus and regulate the organ size and tissue homeostasis. This review focuses on the research progress on YAP/TAZ signaling proteins in myocardial infarction, cardiac remodeling, hypertension and coronary heart disease, cardiomyopathy, and aortic disease. Based on preclinical studies on YAP/TAZ signaling proteins in cellular/animal models and clinical patients, the potential roles of YAP/TAZ proteins in some cardiovascular diseases (CVDs) are summarized.
Collapse
|
17
|
Thanuthanakhun N, Kim MH, Kino-oka M. Cell Behavioral Dynamics as a Cue in Optimizing Culture Stabilization in the Bioprocessing of Pluripotent Stem Cells. Bioengineering (Basel) 2022; 9:669. [PMID: 36354580 PMCID: PMC9687444 DOI: 10.3390/bioengineering9110669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 04/23/2024] Open
Abstract
Pluripotent stem cells (PSCs) are important for future regenerative medicine therapies. However, in the production of PSCs and derivatives, the control of culture-induced fluctuations in the outcome of cell quality remains challenging. A detailed mechanistic understanding of how PSC behaviors are altered in response to biomechanical microenvironments within a culture is necessary for rational bioprocessing optimization. In this review, we discuss recent insights into the role of cell behavioral and mechanical homeostasis in modulating the states and functions of PSCs during culture processes. We delineate promising ways to manipulate the culture variability through regulating cell behaviors using currently developed tools. Furthermore, we anticipate their potential implementation for designing a culture strategy based on the concept of Waddington's epigenetic landscape that may provide a feasible solution for tuning the culture quality and stability in the bioprocessing space.
Collapse
Affiliation(s)
- Naruchit Thanuthanakhun
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Mee-Hae Kim
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Masahiro Kino-oka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
- Research Base for Cell Manufacturability, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
18
|
An R, Wang J, Chen X, Xu R, Hu J, Liu Z, Wei C, Zhang C, Yuan B. YAP signaling is involved in WDR1-regulated proliferation and migration of non-small-cell lung cancer cells. Exp Biol Med (Maywood) 2022; 247:1619-1629. [PMID: 35861209 PMCID: PMC9597210 DOI: 10.1177/15353702221110645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
As a major co-factor of F-actin depolymerization, WD-repeat domain 1 (WDR1) affects the cellular microenvironment by cytoskeleton remodeling, thereby influencing cell molecular behavior. Our previous study showed that WDR1 activates YAP (Yes-associated protein) signaling in non-small-cell lung cancer (NSCLC) cells, but the mechanism remains unclear. We discovered that knockdown WDR1 in NSCLC cells decreased the expression of YAP and the nucleus-to-cytoplasm ratio. Disruption of cortical stress by drugs significantly inhibited YAP nuclear trafficking and enhanced YAP phosphorylation. In WDR1-knockdown NSCLC cells, inhibition of Hippo pathway reduced the nuclear exclusion of YAP and phosphorylated YAP. Our data suggest that WDR1-mediated cortical stress might be involved in regulating YAP signaling, thereby affecting the proliferation and migration of NSCLC cells.
Collapse
Affiliation(s)
- Ran An
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China
| | - Junyan Wang
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China
| | - Xuan Chen
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China
| | - Ruifeng Xu
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China
| | - Jisheng Hu
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China
| | - Zhongying Liu
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China
| | - Chanjuan Wei
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China
| | - Chenxi Zhang
- Central Laboratory, Nanjing Chest
Hospital, The Affiliated Nanjing Brain Hospital of Nanjing Medical University,
Nanjing 210029, P. R. China
| | - Baiyin Yuan
- Biomedical Research Institute, College
of Life Science and Health, Wuhan University of Science and Technology, Wuhan
430081, P.R. China,Baiyin Yuan.
| |
Collapse
|
19
|
Rashid SA, Blanchard AT, Combs JD, Fernandez N, Dong Y, Cho HC, Salaita K. DNA Tension Probes Show that Cardiomyocyte Maturation Is Sensitive to the Piconewton Traction Forces Transmitted by Integrins. ACS NANO 2022; 16:5335-5348. [PMID: 35324164 PMCID: PMC11238821 DOI: 10.1021/acsnano.1c04303] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cardiac muscle cells (CMCs) are the unit cells that comprise the heart. CMCs go through different stages of differentiation and maturation pathways to fully mature into beating cells. These cells can sense and respond to mechanical cues through receptors such as integrins which influence maturation pathways. For example, cell traction forces are important for the differentiation and development of functional CMCs, as CMCs cultured on varying substrate stiffness function differently. Most work in this area has focused on understanding the role of bulk extracellular matrix stiffness in mediating the functional fate of CMCs. Given that stiffness sensing mechanisms are mediated by individual integrin receptors, an important question in this area pertains to the specific magnitude of integrin piconewton (pN) forces that can trigger CMC functional maturation. To address this knowledge gap, we used DNA adhesion tethers that rupture at specific thresholds of force (∼12, ∼56, and ∼160 pN) to test whether capping peak integrin tension to specific magnitudes affects CMC function. We show that adhesion tethers with greater force tolerance lead to functionally mature CMCs as determined by morphology, twitching frequency, transient calcium flux measurements, and protein expression (F-actin, vinculin, α-actinin, YAP, and SERCA2a). Additionally, sarcomeric actinin alignment and multinucleation were significantly enhanced as the mechanical tolerance of integrin tethers was increased. Taken together, the results show that CMCs harness defined pN integrin forces to influence early stage development. This study represents an important step toward biophysical characterization of the contribution of pN forces in early stage cardiac differentiation.
Collapse
Affiliation(s)
- Sk Aysha Rashid
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Aaron T Blanchard
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - J Dale Combs
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Natasha Fernandez
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 1405 Clifton Road NE, Atlanta, Georgia 30322, United States
| | - Yixiao Dong
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Hee Cheol Cho
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 1405 Clifton Road NE, Atlanta, Georgia 30322, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| | - Khalid Salaita
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, Georgia 30332, United States
| |
Collapse
|
20
|
Yeruva S, Waschke J. Structure and regulation of desmosomes in intercalated discs: Lessons from epithelia. J Anat 2022; 242:81-90. [PMID: 35128661 PMCID: PMC9773171 DOI: 10.1111/joa.13634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022] Open
Abstract
For electromechanical coupling of cardiomyocytes, intercalated discs (ICDs) are pivotal as highly specialized intercellular contact areas. ICD consists of adhesive contacts, such as desmosomes and adherens junctions (AJs) that are partially intermingled and thereby form an area composita to provide mechanical strength, as well as gap junctions (GJ) and sodium channels for excitation propagation. In contrast, in epithelia, mixed junctions with features of desmosomes and AJs are regarded as transitory primarily during the formation of desmosomes. The anatomy of desmosomes is defined by a typical ultrastructure with dense intracellular plaques anchoring the cadherin-type adhesion molecules to the intermediate filament cytoskeleton. Desmosomal diseases characterized by impaired adhesive and signalling functions of desmosomal contacts lead to arrhythmogenic cardiomyopathy when affecting cardiomyocytes and cause pemphigus when manifesting in keratinocytes or present as cardiocutaneous syndromes when both cell types are targeted by the disease, which underscores the high biomedical relevance of these cell contacts. Therefore, comparative analyses regarding the structure and regulation of desmosomal contacts in cardiomyocytes and epithelial cells are helpful to better understand disease pathogenesis. In this brief review, we describe the structural properties of ICD compared to epithelial desmosomes and suggest that mechanisms regulating adhesion may at least in part be comparable. Also, we discuss whether phenomena such as hyperadhesion or the bidirectional regulation of desmosomes to serve as signalling hubs in epithelial cells may also be relevant for ICD.
Collapse
Affiliation(s)
- Sunil Yeruva
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| | - Jens Waschke
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| |
Collapse
|
21
|
Sakabe M, Thompson M, Chen N, Verba M, Hassan A, Lu R, Xin M. Inhibition of β1-AR/Gαs signaling promotes cardiomyocyte proliferation in juvenile mice through activation of RhoA-YAP axis. eLife 2022; 11:74576. [PMID: 36479975 PMCID: PMC9767473 DOI: 10.7554/elife.74576] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The regeneration potential of the mammalian heart is incredibly limited, as cardiomyocyte proliferation ceases shortly after birth. β-adrenergic receptor (β-AR) blockade has been shown to improve heart functions in response to injury; however, the underlying mechanisms remain poorly understood. Here, we inhibited β-AR signaling in the heart using metoprolol, a cardio-selective β blocker for β1-adrenergic receptor (β1-AR) to examine its role in heart maturation and regeneration in postnatal mice. We found that metoprolol enhanced cardiomyocyte proliferation and promoted cardiac regeneration post myocardial infarction, resulting in reduced scar formation and improved cardiac function. Moreover, the increased cardiomyocyte proliferation was also induced by the genetic deletion of Gnas, the gene encoding G protein alpha subunit (Gαs), a downstream effector of β-AR. Genome wide transcriptome analysis revealed that the Hippo-effector YAP, which is associated with immature cardiomyocyte proliferation, was upregulated in the cardiomyocytes of β-blocker treated and Gnas cKO hearts. Moreover, the increased YAP activity is modulated by RhoA signaling. Our pharmacological and genetic studies reveal that β1-AR-Gαs-YAP signaling axis is involved in regulating postnatal cardiomyocyte proliferation. These results suggest that inhibiting β-AR-Gαs signaling promotes the regenerative capacity and extends the cardiac regenerative window in juvenile mice by activating YAP-mediated transcriptional programs.
Collapse
Affiliation(s)
- Masahide Sakabe
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Michael Thompson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Nong Chen
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Mark Verba
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Aishlin Hassan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| |
Collapse
|
22
|
Changes in chromatin accessibility landscape and histone H3 core acetylation during valproic acid-induced differentiation of embryonic stem cells. Epigenetics Chromatin 2021; 14:58. [PMID: 34955095 PMCID: PMC8711205 DOI: 10.1186/s13072-021-00432-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/17/2021] [Indexed: 11/27/2022] Open
Abstract
Directed differentiation of mouse embryonic stem cells (mESCs) or induced pluripotent stem cells (iPSCs) provides powerful models to dissect the molecular mechanisms leading to the formation of specific cell lineages. Treatment with histone deacetylase inhibitors can significantly enhance the efficiency of directed differentiation. However, the mechanisms are not well understood. Here, we use CUT&RUN in combination with ATAC-seq to determine changes in both histone modifications and genome-wide chromatin accessibility following valproic acid (VPA) exposure. VPA induced a significant increase in global histone H3 acetylation (H3K56ac), a core histone modification affecting nucleosome stability, as well as enrichment at loci associated with cytoskeletal organization and cellular morphogenesis. In addition, VPA altered the levels of linker histone H1 subtypes and the total histone H1/nucleosome ratio indicative of initial differentiation events. Notably, ATAC-seq analysis revealed changes in chromatin accessibility of genes involved in regulation of CDK serine/threonine kinase activity and DNA duplex unwinding. Importantly, changes in chromatin accessibility were evident at several key genomic loci, such as the pluripotency factor Lefty, cardiac muscle troponin Tnnt2, and the homeodomain factor Hopx, which play critical roles in cardiomyocyte differentiation. Massive parallel transcription factor (TF) footprinting also indicates an increased occupancy of TFs involved in differentiation toward mesoderm and endoderm lineages and a loss of footprints of POU5F1/SOX2 pluripotency factors following VPA treatment. Our results provide the first genome-wide analysis of the chromatin landscape following VPA-induced differentiation in mESCs and provide new mechanistic insight into the intricate molecular processes that govern departure from pluripotency and early lineage commitment.
Collapse
|
23
|
Wang X, Pierre V, Senapati S, Park PSH, Senyo SE. Microenvironment Stiffness Amplifies Post-ischemia Heart Regeneration in Response to Exogenous Extracellular Matrix Proteins in Neonatal Mice. Front Cardiovasc Med 2021; 8:773978. [PMID: 34805326 PMCID: PMC8602555 DOI: 10.3389/fcvm.2021.773978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
The cardiogenesis of the fetal heart is absent in juveniles and adults. Cross-transplantation of decellularized extracellular matrix (dECM) can stimulate regeneration in myocardial infarct (MI) models. We have previously shown that dECM and tissue stiffness have cooperative regulation of heart regeneration in transiently regenerative day 1 neonatal mice. To investigate underlying mechanisms of mechano-signaling and dECM, we pharmacologically altered heart stiffness and administered dECM hydrogels in non-regenerative mice after MI. The dECM combined with softening exhibits preserved cardiac function, LV geometry, increased cardiomyocyte mitosis and lowered fibrosis while stiffening further aggravated ischemic damage. Transcriptome analysis identified a protein in cardiomyocytes, CLCA2, confirmed to be upregulated after MI and downregulated by dECM in a mechanosensitive manner. Synthetic knock-down of CLCA2 expression induced mitosis in primary rat cardiomyocytes in the dish. Together, our results indicate that therapeutic efficacy of extracellular molecules for heart regeneration can be modulated by heart microenvironment stiffness in vivo.
Collapse
Affiliation(s)
- Xinming Wang
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Valinteshley Pierre
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Subhadip Senapati
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Paul S.-H. Park
- Department of Ophthalmology and Visual Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Samuel E. Senyo
- Department of Biomedical Engineering, Case School of Engineering, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
24
|
Meng F, Xie B, Martin JF. Targeting the Hippo pathway in heart repair. Cardiovasc Res 2021; 118:2402-2414. [PMID: 34528077 DOI: 10.1093/cvr/cvab291] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo pathway is an evolutionarily and functionally conserved signaling pathway that controls organ size by regulating cell proliferation, apoptosis, and differentiation. Emerging evidence has shown that the Hippo pathway plays critical roles in cardiac development, homeostasis, disease, and regeneration. Targeting the Hippo pathway has tremendous potential as a therapeutic strategy for treating intractable cardiovascular diseases such as heart failure. In this review, we summarize the function of the Hippo pathway in the heart. Particularly, we highlight the posttranslational modification of Hippo pathway components, including the core kinases LATS1/2 and their downstream effectors YAP/TAZ, in different contexts, which has provided new insights and avenues in cardiac research.
Collapse
Affiliation(s)
- Fansen Meng
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Bing Xie
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030.,Texas Heart Institute, Houston, Texas, 77030
| |
Collapse
|
25
|
Behmer Hansen RA, Wang X, Kaw G, Pierre V, Senyo SE. Accounting for Material Changes in Decellularized Tissue with Underutilized Methodologies. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6696295. [PMID: 34159202 PMCID: PMC8187050 DOI: 10.1155/2021/6696295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 05/05/2021] [Accepted: 05/21/2021] [Indexed: 11/17/2022]
Abstract
Tissue decellularization has rapidly developed to be a practical approach in tissue engineering research; biological tissue is cleared of cells resulting in a protein-rich husk as a natural scaffold for growing transplanted cells as a donor organ therapy. Minimally processed, acellular extracellular matrix reproduces natural interactions with cells in vitro and for tissue engineering applications in animal models. There are many decellularization techniques that achieve preservation of molecular profile (proteins and sugars), microstructure features such as organization of ECM layers (interstitial matrix and basement membrane) and organ level macrofeatures (vasculature and tissue compartments). While structural and molecular cues receive attention, mechanical and material properties of decellularized tissues are not often discussed. The effects of decellularization on an organ depend on the tissue properties, clearing mechanism, chemical interactions, solubility, temperature, and treatment duration. Physical characterization by a few labs including work from the authors provides evidence that decellularization protocols should be tailored to specific research questions. Physical characterization beyond histology and immunohistochemistry of the decellularized matrix (dECM) extends evaluation of retained functional features of the original tissue. We direct our attention to current technologies that can be employed for structure function analysis of dECM using underutilized tools such as atomic force microscopy (AFM), cryogenic electron microscopy (cryo-EM), dynamic mechanical analysis (DMA), Fourier-transform infrared spectroscopy (FTIR), mass spectrometry, and rheometry. Structural imaging and mechanical functional testing combined with high-throughput molecular analyses opens a new approach for a deeper appreciation of how cellular behavior is influenced by the isolated microenvironment (specifically dECM). Additionally, the impact of these features with different decellularization techniques and generation of synthetic material scaffolds with desired attributes are informed. Ultimately, this mechanical profiling provides a new dimension to our understanding of decellularized matrix and its role in new applications.
Collapse
Affiliation(s)
- Ryan A. Behmer Hansen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Xinming Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Gitanjali Kaw
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Valinteshley Pierre
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Samuel E. Senyo
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
26
|
Thomas K, Henley T, Rossi S, Costello MJ, Polacheck W, Griffith BE, Bressan M. Adherens junction engagement regulates functional patterning of the cardiac pacemaker cell lineage. Dev Cell 2021; 56:1498-1511.e7. [PMID: 33891897 PMCID: PMC8137639 DOI: 10.1016/j.devcel.2021.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 02/16/2021] [Accepted: 03/31/2021] [Indexed: 12/19/2022]
Abstract
Cardiac pacemaker cells (CPCs) rhythmically initiate the electrical impulses that drive heart contraction. CPCs display the highest rate of spontaneous depolarization in the heart despite being subjected to inhibitory electrochemical conditions that should theoretically suppress their activity. While several models have been proposed to explain this apparent paradox, the actual molecular mechanisms that allow CPCs to overcome electrogenic barriers to their function remain poorly understood. Here, we have traced CPC development at single-cell resolution and uncovered a series of cytoarchitectural patterning events that are critical for proper pacemaking. Specifically, our data reveal that CPCs dynamically modulate adherens junction (AJ) engagement to control characteristics including surface area, volume, and gap junctional coupling. This allows CPCs to adopt a structural configuration that supports their overall excitability. Thus, our data have identified a direct role for local cellular mechanics in patterning critical morphological features that are necessary for CPC electrical activity.
Collapse
Affiliation(s)
- Kandace Thomas
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Trevor Henley
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Simone Rossi
- Department of Mathematics, University of North Carolina, Chapel Hill, NC, USA
| | - M Joseph Costello
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William Polacheck
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; University of North Carolina at Chapel Hill and North Carolina State University, Joint Department of Biomedical Engineering, Chapel Hill, NC 27599, USA
| | - Boyce E Griffith
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Departments of Mathematics, Applied Physical Sciences, and Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA; Carolina Center for Interdisciplinary Applied Mathematics, University of North Carolina, Chapel Hill, NC, USA; Computational Medicine Program, University of North Carolina, Chapel Hill, NC, USA
| | - Michael Bressan
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
27
|
Decreased YAP activity reduces proliferative ability in human induced pluripotent stem cell of duchenne muscular dystrophy derived cardiomyocytes. Sci Rep 2021; 11:10351. [PMID: 33990626 PMCID: PMC8121946 DOI: 10.1038/s41598-021-89603-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 04/20/2021] [Indexed: 11/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle degeneration accompanied by dilated cardiomyopathy. Recently, abnormality of yes-associated protein (YAP) has been reported as the pathogenesis of muscle degeneration of DMD; however YAP activity remains unclear in dystrophic heart of DMD. Herein, we investigated YAP activity using disease-specific induced pluripotent stem cell (iPSC) derived cardiomyocytes (CMs) in DMD. DMD-iPSCs were generated from DMD patient with exon 48–54 deletion in DMD, and genome-edited (Ed)-DMD-iPSCs with in-frame (Ed-DMD-iPSCs) were created using CRISPR/Cas9. Nuclear translocation of YAP [nuclear (N)/cytoplasmic (C) ratio] was significantly lower in DMD-iPSC-CMs than in Ed-DMD-iPSC-CMs. In addition, Ki67 expression, indicating proliferative ability, was significantly lower in DMD-iPSC-CMs than Ed-DMD-iPSC-CMs. Therefore, immunofluorescent staining showed that actin stress fibers associated with YAP activity by mechanotransduction were disorganized in DMD-iPSC-CMs. Lysophosphatidic acid (LPA), a known lipid mediator on induction of actin polymerization, significantly increased YAP activity and actin dynamics in DMD-iPSC-CMs using live cell imaging. These results suggested that altered YAP activity due to impaired actin dynamics reduced proliferative ability in DMD-iPSC-CMs. Hence, decreased YAP activity in dystrophic heart may contribute to DMD-cardiomyopathy pathogenesis.
Collapse
|
28
|
Querdel E, Reinsch M, Castro L, Köse D, Bähr A, Reich S, Geertz B, Ulmer B, Schulze M, Lemoine MD, Krause T, Lemme M, Sani J, Shibamiya A, Stüdemann T, Köhne M, Bibra CV, Hornaschewitz N, Pecha S, Nejahsie Y, Mannhardt I, Christ T, Reichenspurner H, Hansen A, Klymiuk N, Krane M, Kupatt C, Eschenhagen T, Weinberger F. Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner. Circulation 2021; 143:1991-2006. [PMID: 33648345 PMCID: PMC8126500 DOI: 10.1161/circulationaha.120.047904] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Supplemental Digital Content is available in the text. Human engineered heart tissue (EHT) transplantation represents a potential regenerative strategy for patients with heart failure and has been successful in preclinical models. Clinical application requires upscaling, adaptation to good manufacturing practices, and determination of the effective dose.
Collapse
Affiliation(s)
- Eva Querdel
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Marina Reinsch
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Liesa Castro
- Department of Cardiovascular Surgery, University Heart Center (L.C., S.P., H.R.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.).,Now with Department of Cardiology and Intensive Care Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Germany (L.C.)
| | - Deniz Köse
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Andrea Bähr
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.).,Center for Innovative Medical Models, LMU Munich, Oberschleissheim, Germany (A.B., N.K.)
| | - Svenja Reich
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany
| | - Birgit Geertz
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany
| | - Bärbel Ulmer
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Mirja Schulze
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Marc D Lemoine
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.).,Department of Cardiology-Electrophysiology (M.D.L.), University Heart Center, Hamburg, Germany
| | - Tobias Krause
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Marta Lemme
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Jascha Sani
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Aya Shibamiya
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Tim Stüdemann
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Maria Köhne
- German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.).,Department of Pediatric Cardiac Surgery (M. Köhne), University Heart Center, Hamburg, Germany
| | - Constantin von Bibra
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Nadja Hornaschewitz
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.)
| | - Simon Pecha
- Department of Cardiovascular Surgery, University Heart Center (L.C., S.P., H.R.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Yusuf Nejahsie
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany
| | - Ingra Mannhardt
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Hermann Reichenspurner
- Department of Cardiovascular Surgery, University Heart Center (L.C., S.P., H.R.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Arne Hansen
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Nikolai Klymiuk
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.).,Center for Innovative Medical Models, LMU Munich, Oberschleissheim, Germany (A.B., N.K.)
| | - M Krane
- Department of Cardiovascular Surgery, German Heart Centre Munich (M. Krane), Technical University Munich, Germany.,INSURE (Institute for Translational Cardiac Surgery), Cardiovascular Surgery, Munich, Germany (M. Krane)
| | - C Kupatt
- I. Medizinische Klinik & Poliklinik, University Clinic Rechts der Isar (A.B., N.H., N.K., C.K.), Technical University Munich, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance Munich (A.B., N.H., N.K., C.K.)
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology (E.Q., M.R., D.K., S.R., B.G., B.U., M.S., T.K., M.L., J.S., A.S., T.S., C.v.B., Y.N., I.M., T.C., A.H., T.E., F.W.), University Medical Center, Hamburg-Eppendorf, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck (E.Q., M.R., L.C., D.K., B.U., M.S., M.D.L., T.K., M.L., J.S., A.S., T.S., M. Köhne, C.v.B., S.P., I.M., T.C., H.R., A.H., T.E., F.W.)
| |
Collapse
|
29
|
Esmaeili H, Li C, Fu X, Jung JP. Engineering Extracellular Matrix Proteins to Enhance Cardiac Regeneration After Myocardial Infarction. Front Bioeng Biotechnol 2021; 8:611936. [PMID: 33553118 PMCID: PMC7855456 DOI: 10.3389/fbioe.2020.611936] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/18/2020] [Indexed: 01/09/2023] Open
Abstract
Engineering microenvironments for accelerated myocardial repair is a challenging goal. Cell therapy has evolved over a few decades to engraft therapeutic cells to replenish lost cardiomyocytes in the left ventricle. However, compelling evidence supports that tailoring specific signals to endogenous cells rather than the direct integration of therapeutic cells could be an attractive strategy for better clinical outcomes. Of many possible routes to instruct endogenous cells, we reviewed recent cases that extracellular matrix (ECM) proteins contribute to enhanced cardiomyocyte proliferation from neonates to adults. In addition, the presence of ECM proteins exerts biophysical regulation in tissue, leading to the control of microenvironments and adaptation for enhanced cardiomyocyte proliferation. Finally, we also summarized recent clinical trials exclusively using ECM proteins, further supporting the notion that engineering ECM proteins would be a critical strategy to enhance myocardial repair without taking any risks or complications of applying therapeutic cardiac cells.
Collapse
Affiliation(s)
- Hamid Esmaeili
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, United States
| | - Chaoyang Li
- School of Animal Sciences, Louisiana State University AgCenter, Baton Rouge, LA, United States
| | - Xing Fu
- School of Animal Sciences, Louisiana State University AgCenter, Baton Rouge, LA, United States
| | - Jangwook P Jung
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
30
|
Liu Y, Li J, Yao B, Wang Y, Wang R, Yang S, Li Z, Zhang Y, Huang S, Fu X. The stiffness of hydrogel-based bioink impacts mesenchymal stem cells differentiation toward sweat glands in 3D-bioprinted matrix. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 118:111387. [DOI: 10.1016/j.msec.2020.111387] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 07/08/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
|
31
|
Lee SH, Hadipour-Lakmehsari S, Kim DH, Di Paola M, Kuzmanov U, Shah S, Lee JJH, Kislinger T, Sharma P, Oudit GY, Gramolini AO. Bioinformatic analysis of membrane and associated proteins in murine cardiomyocytes and human myocardium. Sci Data 2020; 7:425. [PMID: 33262348 PMCID: PMC7708497 DOI: 10.1038/s41597-020-00762-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
In the current study we examined several proteomic- and RNA-Seq-based datasets of cardiac-enriched, cell-surface and membrane-associated proteins in human fetal and mouse neonatal ventricular cardiomyocytes. By integrating available microarray and tissue expression profiles with MGI phenotypic analysis, we identified 173 membrane-associated proteins that are cardiac-enriched, conserved amongst eukaryotic species, and have not yet been linked to a 'cardiac' Phenotype-Ontology. To highlight the utility of this dataset, we selected several proteins to investigate more carefully, including FAM162A, MCT1, and COX20, to show cardiac enrichment, subcellular distribution and expression patterns in disease. We performed three-dimensional confocal imaging analysis to validate subcellular localization and expression in adult mouse ventricular cardiomyocytes. FAM162A, MCT1, and COX20 were expressed differentially at the transcriptomic and proteomic levels in multiple models of mouse and human heart diseases and may represent potential diagnostic and therapeutic targets for human dilated and ischemic cardiomyopathies. Altogether, we believe this comprehensive cardiomyocyte membrane proteome dataset will prove instrumental to future investigations aimed at characterizing heart disease markers and/or therapeutic targets for heart failure.
Collapse
Affiliation(s)
- Shin-Haw Lee
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, M5G 1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada
| | - Sina Hadipour-Lakmehsari
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, M5G 1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada
| | - Da Hye Kim
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, M5G 1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada
| | - Michelle Di Paola
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, M5G 1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada
| | - Uros Kuzmanov
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, M5G 1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada
| | - Saumya Shah
- Department of Medicine, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, Edmonton, Alberta, T6G 2B7, Canada
| | - Joseph Jong-Hwan Lee
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, M5G 1M1, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada
| | - Thomas Kislinger
- Princess Margaret Cancer Research Centre, Toronto, Ontario, M5G 1L8, Canada
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| | - Parveen Sharma
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada
- Department of Cardiovascular & Metabolic Medicine, University of Liverpool, Liverpool, L69 3GE, UK
| | - Gavin Y Oudit
- Department of Medicine, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
- Mazankowski Alberta Heart Institute, Edmonton, Alberta, T6G 2B7, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Ontario, M5G 1M1, Canada.
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, M5S 1M8, Canada.
| |
Collapse
|
32
|
Yu Y, Su X, Qin Q, Hou Y, Zhang X, Zhang H, Jia M, Chen Y. Yes-associated protein and transcriptional coactivator with PDZ-binding motif as new targets in cardiovascular diseases. Pharmacol Res 2020; 159:105009. [DOI: 10.1016/j.phrs.2020.105009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/14/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
|
33
|
Wang X, Senapati S, Akinbote A, Gnanasambandam B, Park PSH, Senyo SE. Microenvironment stiffness requires decellularized cardiac extracellular matrix to promote heart regeneration in the neonatal mouse heart. Acta Biomater 2020; 113:380-392. [PMID: 32590172 PMCID: PMC7428869 DOI: 10.1016/j.actbio.2020.06.032] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 02/08/2023]
Abstract
The transient period of regeneration potential in the postnatal heart suggests molecular changes with maturation influence the cardiac response to damage. We have previously demonstrated that injury and exercise can stimulate cardiomyocyte proliferation in the adult heart suggesting a sensitivity to exogenous signals. Here, we consider whether exogenous fetal ECM and mechanically unloading interstitial matrix can drive regeneration after myocardial infarction (MI) surgery in low-regenerative hearts of day5 mice. Compared to controls, exogenous fetal ECM increases cardiac function and lowers fibrosis at 3 weeks post-injury and this effect can be augmented by softening heart tissue. In vitro experiments support a mechano-sensitivity to exogenous ECM signaling. We tested potential mechanisms and observed that fetal ECM increases nuclear YAP localization which could be enhanced by pharmacological stabilization of the cytoskeleton. Blocking YAP expression lowered fetal ECM effects though not completely. Lastly we observed mechanically unloading heart interstitial matrix increased agrin expression, an extracellular node in the YAP signaling pathway. Collectively, these data support a combined effect of exogenous factors and mechanical activity in altering agrin expression, cytoskeletal remodeling, and YAP signaling in driving cardiomyocyte cell cycle activity and regeneration in postnatal non-regenerative mice. STATEMENT OF SIGNIFICANCE: With the purpose of developing regenerative strategies, we investigate the influence of the local niche on the cardiac injury response. We conclude tissue stiffness, as anticipated in aging or disease, impairs regenerative therapeutics. Most novel, mechanical unloading facilitates enhanced cardiac regeneration only after cells are pushed into a permissive state by fetal biomolecules. Specifically, mechanical unloading appears to increase extracellular agrin expression that amplifies fetal-stimulation of nuclear YAP signaling which correlates with observed increases of cell cycle activity in cardiomyocytes. The results further suggest the cytoskeleton is critical to this interaction between mechanical unloading and independently actived YAP signaling. Using animal models, tissue explants, and cells, this work indicates that local mechanical stimuli can augment proliferating-permissive cardiomyocytes in the natural cardiac niche.
Collapse
Affiliation(s)
- Xinming Wang
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Subhadip Senapati
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University, United States
| | - Akinola Akinbote
- Department of Macromolecular Science & Engineering, Case Western Reserve University, United States
| | - Bhargavee Gnanasambandam
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Paul S-H Park
- Department of Ophthalmology & Visual Sciences, Case Western Reserve University, United States
| | - Samuel E Senyo
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States.
| |
Collapse
|
34
|
Flinn MA, Otten C, Brandt ZJ, Bostrom JR, Kenarsary A, Wan TC, Auchampach JA, Abdelilah-Seyfried S, O'Meara CC, Link BA. Llgl1 regulates zebrafish cardiac development by mediating Yap stability in cardiomyocytes. Development 2020; 147:147/16/dev193581. [PMID: 32843528 DOI: 10.1242/dev.193581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/10/2020] [Indexed: 01/19/2023]
Abstract
The Hippo-Yap pathway regulates multiple cellular processes in response to mechanical and other stimuli. In Drosophila, the polarity protein Lethal (2) giant larvae [L(2)gl], negatively regulates Hippo-mediated transcriptional output. However, in vertebrates, little is known about its homolog Llgl1. Here, we define a novel role for vertebrate Llgl1 in regulating Yap stability in cardiomyocytes, which impacts heart development. In contrast to the role of Drosophila L(2)gl, Llgl1 depletion in cultured rat cardiomyocytes decreased Yap protein levels and blunted target gene transcription without affecting Yap transcript abundance. Llgl1 depletion in zebrafish resulted in larger and dysmorphic cardiomyocytes, pericardial effusion, impaired blood flow and aberrant valvulogenesis. Cardiomyocyte Yap protein levels were decreased in llgl1 morphants, whereas Notch, which is regulated by hemodynamic forces and participates in valvulogenesis, was more broadly activated. Consistent with the role of Llgl1 in regulating Yap stability, cardiomyocyte-specific overexpression of Yap in Llgl1-depleted embryos ameliorated pericardial effusion and restored blood flow velocity. Altogether, our data reveal that vertebrate Llgl1 is crucial for Yap stability in cardiomyocytes and its absence impairs cardiac development.
Collapse
Affiliation(s)
- Michael A Flinn
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cécile Otten
- Institute for Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
| | - Zachary J Brandt
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jonathan R Bostrom
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aria Kenarsary
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tina C Wan
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Pharmacology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - John A Auchampach
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Pharmacology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Salim Abdelilah-Seyfried
- Institute for Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany.,Institute for Molecular Biology, Hannover Medical School, 30625 Hannover, Germany
| | - Caitlin C O'Meara
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.,Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Brian A Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA .,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
35
|
Adeniba OO, Corbin EA, Ganguli A, Kim Y, Bashir R. Simultaneous time-varying viscosity, elasticity, and mass measurements of single adherent cancer cells across cell cycle. Sci Rep 2020; 10:12803. [PMID: 32733047 PMCID: PMC7393350 DOI: 10.1038/s41598-020-69638-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 07/16/2020] [Indexed: 01/04/2023] Open
Abstract
Biophysical studies on single cells have linked cell mechanics to physiology, functionality and disease. Evaluation of mass and viscoelasticity versus cell cycle can provide further insights into cell cycle progression and the uncontrolled proliferation of cancer. Using our pedestal microelectromechanical systems resonant sensors, we have developed a non-contact interferometric measurement technique that simultaneously tracks the dynamic changes in the viscoelastic moduli and mass of adherent colon (HT-29) and breast cancer (MCF-7) cells from the interphase through mitosis and then to the cytokinesis stages of their growth cycle. We show that by combining three optomechanical parameters in an optical path length equation and a two-degree-of-freedom model, we can simultaneously extract the viscoelasticity and mass as a function of the nano-scaled membrane fluctuation of each adherent cell. Our measurements are able to discern between soft and stiff cells across the cell cycle and demonstrated sharp viscoelastic changes due to cortical stiffening around mitosis. Cell rounding before division can be detected by measurement of mechanical coupling between the cells and the sensors. Our measurement device and method can provide for new insights into the mechanics of single adherent cells versus time.
Collapse
Affiliation(s)
- Olaoluwa O Adeniba
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Elise A Corbin
- Biomedical Engineering Department, University of Delaware, Newark, DE, 19716, USA
- Materials Science and Engineering Department, University of Delaware, Newark, DE, 19716, USA
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, 19803, USA
| | - Anurup Ganguli
- Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yongdeok Kim
- Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Materials Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Rashid Bashir
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Micro and Nanotechnology Laboratory, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
- Carle Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
36
|
Selig M, Lauer JC, Hart ML, Rolauffs B. Mechanotransduction and Stiffness-Sensing: Mechanisms and Opportunities to Control Multiple Molecular Aspects of Cell Phenotype as a Design Cornerstone of Cell-Instructive Biomaterials for Articular Cartilage Repair. Int J Mol Sci 2020; 21:E5399. [PMID: 32751354 PMCID: PMC7432012 DOI: 10.3390/ijms21155399] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/23/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Since material stiffness controls many cell functions, we reviewed the currently available knowledge on stiffness sensing and elucidated what is known in the context of clinical and experimental articular cartilage (AC) repair. Remarkably, no stiffness information on the various biomaterials for clinical AC repair was accessible. Using mRNA expression profiles and morphology as surrogate markers of stiffness-related effects, we deduced that the various clinically available biomaterials control chondrocyte (CH) phenotype well, but not to equal extents, and only in non-degenerative settings. Ample evidence demonstrates that multiple molecular aspects of CH and mesenchymal stromal cell (MSC) phenotype are susceptible to material stiffness, because proliferation, migration, lineage determination, shape, cytoskeletal properties, expression profiles, cell surface receptor composition, integrin subunit expression, and nuclear shape and composition of CHs and/or MSCs are stiffness-regulated. Moreover, material stiffness modulates MSC immuno-modulatory and angiogenic properties, transforming growth factor beta 1 (TGF-β1)-induced lineage determination, and CH re-differentiation/de-differentiation, collagen type II fragment production, and TGF-β1- and interleukin 1 beta (IL-1β)-induced changes in cell stiffness and traction force. We then integrated the available molecular signaling data into a stiffness-regulated CH phenotype model. Overall, we recommend using material stiffness for controlling cell phenotype, as this would be a promising design cornerstone for novel future-oriented, cell-instructive biomaterials for clinical high-quality AC repair tissue.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Jasmin C. Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Melanie L. Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center—Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; (M.S.); (J.C.L.); (M.L.H.)
| |
Collapse
|
37
|
Liu Y, Li Z, Li J, Yang S, Zhang Y, Yao B, Song W, Fu X, Huang S. Stiffness-mediated mesenchymal stem cell fate decision in 3D-bioprinted hydrogels. BURNS & TRAUMA 2020; 8:tkaa029. [PMID: 32733974 PMCID: PMC7382973 DOI: 10.1093/burnst/tkaa029] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/10/2020] [Indexed: 02/07/2023]
Abstract
Background Hydrogels with tuneable mechanical properties are an attractive material platform for 3D bioprinting. Thus far, numerous studies have confirmed that the biophysical cues of hydrogels, such as stiffness, are known to have a profound impact on mesenchymal stem cell (MSC) differentiation; however, their differentiation potential within 3D-bioprinted hydrogels is not completely understood. Here, we propose a protocol for the exploration of how the stiffness of alginate-gelatin (Alg-Gel) composite hydrogels (the widely used bioink) affects the differentiation of MSCs in the presence or absence of differentiation inducing factors. Methods Two types of Alg-Gel composite hydrogels (Young's modulus: 50 kPa vs. 225 kPa) were bioprinted independently of porosity. Then, stiffness-induced biases towards adipogenic and osteogenic differentiation of the embedded MSCs were analysed by co-staining with alkaline phosphatase (ALP) and oil red O. The expression of specific markers at the gene level was detected after a 3-day culture. Results Confocal microscopy indicated that all tested hydrogels supported MSC growth and viability during the culture period. Higher expression of adipogenic and osteogenic markers (ALP and lipoprotein lipase (LPL)) in stiffer 3D-bioprinted matrices demonstrated a more significant response of MSCs to stiffer hydrogels with respect to differentiation, which was more robust in differentiation-inducing medium. However, the LPL expression in stiffer 3D-bioprinted constructs was reduced at day 3 regardless of the presence of differentiation-inducing factors. Although MSCs embedded in softer hydrogels to some extent proceeded toward adipogenic and osteogenic lineages within a few days, their differentiation seemed to be slower and more limited. Interestingly, the hydrogel itself (without differentiation-inducing factors) exhibited a slight effect on whether MSCs differentiated towards an adipogenic or an osteogenic fate. Considering that the mechano-regulated protein Yes-associated protein (YAP) is involved in MSC fate decisions, we further found that inhibition of YAP significantly downregulated the expression of ALP and LPL in MSCs in stiffer constructs regardless of the induced growth factors present. Conclusions These results demonstrate that the differentiation of MSCs in 3D-bioprinted matrices is dependent on hydrogel stiffness, which emphasizes the importance of biophysical cues as a determinant of cellular behaviour.
Collapse
Affiliation(s)
- Yufan Liu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Zhao Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Jianjun Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Siming Yang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Yijie Zhang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Bin Yao
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Wei Song
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| | - Sha Huang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Department, PLA General Hospital and PLA Medical College, 28 Fu Xing Road, Beijing 100853, P. R. China.,PLA Key Laboratory of Tissue Repair and Regenerative Medicine and Beijing Key Research Laboratory of Skin Injury, Repair and Regeneration; Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 2019RU051, 51 Fu Cheng Road, Beijing 100048, P. R. China
| |
Collapse
|
38
|
Targeting the Hippo pathway in cancer, fibrosis, wound healing and regenerative medicine. Nat Rev Drug Discov 2020; 19:480-494. [PMID: 32555376 DOI: 10.1038/s41573-020-0070-z] [Citation(s) in RCA: 539] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 02/07/2023]
Abstract
The Hippo pathway is an evolutionarily conserved signalling pathway with key roles in organ development, epithelial homeostasis, tissue regeneration, wound healing and immune modulation. Many of these roles are mediated by the transcriptional effectors YAP and TAZ, which direct gene expression via control of the TEAD family of transcription factors. Dysregulated Hippo pathway and YAP/TAZ-TEAD activity is associated with various diseases, most notably cancer, making this pathway an attractive target for therapeutic intervention. This Review highlights the key findings from studies of Hippo pathway signalling across biological processes and diseases, and discusses new strategies and therapeutic implications of targeting this pathway.
Collapse
|
39
|
Angulo-Urarte A, van der Wal T, Huveneers S. Cell-cell junctions as sensors and transducers of mechanical forces. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183316. [PMID: 32360073 DOI: 10.1016/j.bbamem.2020.183316] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/02/2020] [Accepted: 04/15/2020] [Indexed: 12/16/2022]
Abstract
Epithelial and endothelial monolayers are multicellular sheets that form barriers between the 'outside' and 'inside' of tissues. Cell-cell junctions, made by adherens junctions, tight junctions and desmosomes, hold together these monolayers. They form intercellular contacts by binding their receptor counterparts on neighboring cells and anchoring these structures intracellularly to the cytoskeleton. During tissue development, maintenance and pathogenesis, monolayers encounter a range of mechanical forces from the cells themselves and from external systemic forces, such as blood pressure or tissue stiffness. The molecular landscape of cell-cell junctions is diverse, containing transmembrane proteins that form intercellular bonds and a variety of cytoplasmic proteins that remodel the junctional connection to the cytoskeleton. Many junction-associated proteins participate in mechanotransduction cascades to confer mechanical cues into cellular responses that allow monolayers to maintain their structural integrity. We will discuss force-dependent junctional molecular events and their role in cell-cell contact organization and remodeling.
Collapse
Affiliation(s)
- Ana Angulo-Urarte
- Amsterdam UMC, University of Amsterdam, Location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Tanne van der Wal
- Amsterdam UMC, University of Amsterdam, Location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Stephan Huveneers
- Amsterdam UMC, University of Amsterdam, Location AMC, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands.
| |
Collapse
|
40
|
Cardiac regeneration as an environmental adaptation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118623. [DOI: 10.1016/j.bbamcr.2019.118623] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/02/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022]
|
41
|
Abstract
Arrhythmogenic cardiomyopathy is a genetic disorder characterized by the risk of life-threatening arrhythmias, myocardial dysfunction and fibrofatty replacement of myocardial tissue. Mutations in genes that encode components of desmosomes, the adhesive junctions that connect cardiomyocytes, are the predominant cause of arrhythmogenic cardiomyopathy and can be identified in about half of patients with the condition. However, the molecular mechanisms leading to myocardial destruction, remodelling and arrhythmic predisposition remain poorly understood. Through the development of animal, induced pluripotent stem cell and other models of disease, advances in our understanding of the pathogenic mechanisms of arrhythmogenic cardiomyopathy over the past decade have brought several signalling pathways into focus. These pathways include canonical and non-canonical WNT signalling, the Hippo-Yes-associated protein (YAP) pathway and transforming growth factor-β signalling. These studies have begun to identify potential therapeutic targets whose modulation has shown promise in preclinical models. In this Review, we summarize and discuss the reported molecular mechanisms underlying the pathogenesis of arrhythmogenic cardiomyopathy.
Collapse
|
42
|
Vargas RE, Duong VT, Han H, Ta AP, Chen Y, Zhao S, Yang B, Seo G, Chuc K, Oh S, El Ali A, Razorenova OV, Chen J, Luo R, Li X, Wang W. Elucidation of WW domain ligand binding specificities in the Hippo pathway reveals STXBP4 as YAP inhibitor. EMBO J 2020; 39:e102406. [PMID: 31782549 PMCID: PMC6939200 DOI: 10.15252/embj.2019102406] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 11/06/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
The Hippo pathway, which plays a critical role in organ size control and cancer, features numerous WW domain-based protein-protein interactions. However, ~100 WW domains and 2,000 PY motif-containing peptide ligands are found in the human proteome, raising a "WW-PY" binding specificity issue in the Hippo pathway. In this study, we have established the WW domain binding specificity for Hippo pathway components and uncovered a unique amino acid sequence required for it. By using this criterion, we have identified a WW domain-containing protein, STXBP4, as a negative regulator of YAP. Mechanistically, STXBP4 assembles a protein complex comprising α-catenin and a group of Hippo PY motif-containing components/regulators to inhibit YAP, a process that is regulated by actin cytoskeleton tension. Interestingly, STXBP4 is a potential tumor suppressor for human kidney cancer, whose downregulation is correlated with YAP activation in clear cell renal cell carcinoma. Taken together, our study not only elucidates the WW domain binding specificity for the Hippo pathway, but also reveals STXBP4 as a player in actin cytoskeleton tension-mediated Hippo pathway regulation.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Hippo Signaling Pathway
- Humans
- Kidney Neoplasms/genetics
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Prognosis
- Protein Binding
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Signal Transduction
- Survival Rate
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transcription, Genetic
- Tumor Cells, Cultured
- Vesicular Transport Proteins/genetics
- Vesicular Transport Proteins/metabolism
- WW Domains
- Xenograft Model Antitumor Assays
- YAP-Signaling Proteins
Collapse
Affiliation(s)
- Rebecca E Vargas
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Vy Thuy Duong
- Department of ChemistryUniversity of California, IrvineIrvineCAUSA
| | - Han Han
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Albert Paul Ta
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Yuxuan Chen
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Shiji Zhao
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Bing Yang
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Gayoung Seo
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Kimberly Chuc
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Sunwoo Oh
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| | - Amal El Ali
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCAUSA
| | - Olga V Razorenova
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCAUSA
| | - Junjie Chen
- Department of Experimental Radiation OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Ray Luo
- Department of Molecular Biology and BiochemistryUniversity of California, IrvineIrvineCAUSA
- Department of Chemical and Biomolecular EngineeringUniversity of California, IrvineIrvineCAUSA
- Department of Materials Science and EngineeringUniversity of California, IrvineIrvineCAUSA
- Department of Biomedical EngineeringUniversity of California, IrvineIrvineCAUSA
| | - Xu Li
- School of Life SciencesWestlake UniversityHangzhouZhejiangChina
| | - Wenqi Wang
- Department of Developmental and Cell BiologyUniversity of California, IrvineIrvineCAUSA
| |
Collapse
|
43
|
Flinn MA, Link BA, O'Meara CC. Upstream regulation of the Hippo-Yap pathway in cardiomyocyte regeneration. Semin Cell Dev Biol 2019; 100:11-19. [PMID: 31606277 DOI: 10.1016/j.semcdb.2019.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022]
Abstract
The response of the adult mammalian heart to injury such as myocardial infarction has long been described as primarily fibrotic scarring and adverse remodeling with little to no regeneration of cardiomyocytes. Emerging studies have challenged this paradigm by demonstrating that, indeed, adult mammalian cardiomyocytes are capable of completing cytokinesis albeit at levels vastly insufficient to compensate for the loss of functional cardiomyocytes following ischemic injury. Thus, there is great interest in identifying mechanisms to guide adult cardiomyocyte cell cycle re-entry and facilitate endogenous heart regeneration. The Hippo signaling pathway is a core kinase cascade that functions to suppress the transcriptional co-activators Yap and Taz by phosphorylation and therefore cytoplasmic retention or phospho-degradation. This pathway has recently sparked interest in the field of cardiac regeneration as inhibition of Hippo kinase signaling or overdriving the transcriptional co-activator, Yap, significantly promotes proliferation of terminally differentiated adult mammalian cardiomyocytes and can restore function in failing mouse hearts. Thus, the Hippo pathway is an attractive therapeutic target for promoting cardiomyocyte renewal and cardiac regeneration. Although the core kinases and transcriptional activators of the Hippo pathway have been studied extensively over the last twenty years, the regulatory inputs of this pathway, particularly in vertebrates, are poorly understood. Recent studies have elucidated several upstream regulatory inputs to the Hippo pathway in adult mammalian cardiomyocytes that influence cell proliferation and heart regeneration. Considering upstream inputs to the Hippo pathway are thought to be context and cell type specific, targeting these various components could serve as a therapeutic approach for refining Hippo-Yap signaling in the heart. Here, we provide an overview of the emerging regulatory inputs to the Hippo pathway as they relate to mammalian cardiomyocytes and heart regeneration.
Collapse
Affiliation(s)
- Michael A Flinn
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Brian A Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Caitlin C O'Meara
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA; Genomics Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
44
|
Abstract
The Hippo-YAP (Yes-associated protein) pathway is an evolutionarily and functionally conserved regulator of organ size and growth with crucial roles in cell proliferation, apoptosis, and differentiation. This pathway has great potential for therapeutic manipulation in different disease states and to promote organ regeneration. In this Review, we summarize findings from the past decade revealing the function and regulation of the Hippo-YAP pathway in cardiac development, growth, homeostasis, disease, and regeneration. In particular, we highlight the roles of the Hippo-YAP pathway in endogenous heart muscle renewal, including the pivotal role of the Hippo-YAP pathway in regulating cardiomyocyte proliferation and differentiation, stress response, and mechanical signalling. The human heart lacks the capacity to self-repair; therefore, the loss of cardiomyocytes after injury such as myocardial infarction can result in heart failure and death. Despite substantial advances in the treatment of heart failure, an enormous unmet clinical need exists for alternative treatment options. Targeting the Hippo-YAP pathway has tremendous potential for developing therapeutic strategies for cardiac repair and regeneration for currently intractable cardiovascular diseases such as heart failure. The lessons learned from cardiac repair and regeneration studies will also bring new insights into the regeneration of other tissues with limited regenerative capacity.
Collapse
|
45
|
Corbin EA, Vite A, Peyster EG, Bhoopalam M, Brandimarto J, Wang X, Bennett AI, Clark AT, Cheng X, Turner KT, Musunuru K, Margulies KB. Tunable and Reversible Substrate Stiffness Reveals a Dynamic Mechanosensitivity of Cardiomyocytes. ACS APPLIED MATERIALS & INTERFACES 2019; 11:20603-20614. [PMID: 31074953 DOI: 10.1021/acsami.9b02446] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
New directions in material applications have allowed for the fresh insight into the coordination of biophysical cues and regulators. Although the role of the mechanical microenvironment on cell responses and mechanics is often studied, most analyses only consider static environments and behavior, however, cells and tissues are themselves dynamic materials that adapt in myriad ways to alterations in their environment. Here, we introduce an approach, through the addition of magnetic inclusions into a soft poly(dimethylsiloxane) elastomer, to fabricate a substrate that can be stiffened nearly instantaneously in the presence of cells through the use of a magnetic gradient to investigate short-term cellular responses to dynamic stiffening or softening. This substrate allows us to observe time-dependent changes, such as spreading, stress fiber formation, Yes-associated protein translocation, and sarcomere organization. The identification of temporal dynamic changes on a short time scale suggests that this technology can be more broadly applied to study targeted mechanisms of diverse biologic processes, including cell division, differentiation, tissue repair, pathological adaptations, and cell-death pathways. Our method provides a unique in vitro platform for studying the dynamic cell behavior by better mimicking more complex and realistic microenvironments. This platform will be amenable to future studies aimed at elucidating the mechanisms underlying mechanical sensing and signaling that influence cellular behaviors and interactions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Andy T Clark
- Department of Physics , Bryn Mawr College , Bryn Mawr , Pennsylvania 19010 , United States
| | - Xuemei Cheng
- Department of Physics , Bryn Mawr College , Bryn Mawr , Pennsylvania 19010 , United States
| | | | | | | |
Collapse
|
46
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
47
|
Cui M, Wang Z, Bassel-Duby R, Olson EN. Genetic and epigenetic regulation of cardiomyocytes in development, regeneration and disease. Development 2018; 145:145/24/dev171983. [PMID: 30573475 DOI: 10.1242/dev.171983] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Embryonic and postnatal life depend on the uninterrupted function of cardiac muscle cells. These cells, termed cardiomyocytes, display many fascinating behaviors, including complex morphogenic movements, interactions with other cell types of the heart, persistent contractility and quiescence after birth. Each of these behaviors depends on complex interactions between both cardiac-restricted and widely expressed transcription factors, as well as on epigenetic modifications. Here, we review recent advances in our understanding of the genetic and epigenetic control of cardiomyocyte differentiation and proliferation during heart development, regeneration and disease. We focus on those regulators that are required for both heart development and disease, and highlight the regenerative principles that might be manipulated to restore function to the injured adult heart.
Collapse
Affiliation(s)
- Miao Cui
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| |
Collapse
|
48
|
Liu S, Martin JF. The regulation and function of the Hippo pathway in heart regeneration. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 8:e335. [PMID: 30169913 DOI: 10.1002/wdev.335] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/30/2018] [Accepted: 07/25/2018] [Indexed: 12/31/2022]
Abstract
Heart failure caused by cardiomyocyte loss and fibrosis is a leading cause of death worldwide. Although current treatments for heart failure such as heart transplantation and left ventricular assist device implantation have obvious value, new approaches are needed. Endogenous adult cardiomyocyte renewal is measurable but inefficient and inadequate in response to extensive acute heart damage. Stimulating self-renewal of endogenous cardiomyocytes holds great promise for heart repair. Uncovering the genetic mechanisms underlying cardiomyocyte renewal is a critical step in developing new approaches to repairing the heart. Recent studies have revealed that the inhibition of the Hippo pathway is sufficient to promote the proliferation of endogenous cardiomyocytes, indicating that the manipulation of the Hippo pathway in the heart may be a promising treatment for heart failure in the future. We summarize recent findings that have shed light on the function of the Hippo pathway in heart regeneration. We also discuss the mechanisms by which Hippo pathway inhibition promotes heart regeneration and how the Hippo pathway responds to different types of injury or stress during the regenerative process. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Regeneration.
Collapse
Affiliation(s)
- Shijie Liu
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas
| | - James F Martin
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, Texas.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
49
|
Abstract
Death of adult cardiac myocytes and supportive tissues resulting from cardiovascular diseases such as myocardial infarction is the proximal driver of pathological ventricular remodeling that often culminates in heart failure. Unfortunately, no currently available therapeutic barring heart transplantation can directly replenish myocytes lost from the injured heart. For decades, the field has struggled to define the intrinsic capacity and cellular sources for endogenous myocyte turnover in pursuing more innovative therapeutic strategies aimed at regenerating the injured heart. Although controversy persists to this day as to the best therapeutic regenerative strategy to use, a growing consensus has been reached that the very limited capacity for new myocyte formation in the adult mammalian heart is because of proliferation of existing cardiac myocytes but not because of the activity of an endogenous progenitor cell source of some sort. Hence, future therapeutic approaches should take into consideration the fundamental biology of myocyte renewal in designing strategies to potentially replenish these cells in the injured heart.
Collapse
Affiliation(s)
| | - Jeffery D Molkentin
- From the Department of Pediatrics (R.J.V., J.D.M.)
- Howard Hughes Medical Institute (J.D.M.)
| | - Steven R Houser
- Cincinnati Children's Hospital Medical Center, OH; and the Lewis Katz School of Medicine, Cardiovascular Research Center, Temple University, Philadelphia, PA (S.R.H.)
| |
Collapse
|