1
|
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
|
2
|
Cardona-Timoner M, Gomes RN, Nascimento DS. Dressed in Collagen: 2D and 3D Cardiac Fibrosis Models. Int J Mol Sci 2025; 26:3038. [PMID: 40243696 PMCID: PMC11988687 DOI: 10.3390/ijms26073038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/20/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025] Open
Abstract
Cardiovascular diseases (CVD), the leading cause of death worldwide, and their strong association with fibrosis highlight the pressing need for innovative antifibrotic therapies. In vitro models have emerged as valuable tools for replicating cardiac fibrosis 'in a dish', facilitating the study of disease mechanisms and serving as platforms for drug testing and development. These in vitro systems encompass 2D and 3D models, each with its own limitations and advantages. 2D models offer high reproducibility, cost-effectiveness, and high-throughput capabilities, but they oversimplify the complex fibrotic environment. On the other hand, 3D models provide greater biological relevance but are more complex, harder to reproduce, and less suited for high-throughput screening. The choice of model depends on the specific research question and the stage of drug development. Despite significant progress, challenges remain, including the integration of immune cells in cardiac fibrosis and optimizing the scalability and throughput of highly biomimetic systems. Herein, we review recent in vitro cardiac fibrosis models, with a focus on their shared characteristics and remaining challenges, and explore how in vitro fibrosis models of other organs could inspire novel approaches in cardiac research, showcasing potential strategies that could be adapted to refine myocardial fibrosis models.
Collapse
Affiliation(s)
- Maria Cardona-Timoner
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.C.-T.); (R.N.G.)
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
- Faculdade de Engenharia da Universidade do Porto (FEUP), 4200-465 Porto, Portugal
| | - Rita N. Gomes
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.C.-T.); (R.N.G.)
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Diana S. Nascimento
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal; (M.C.-T.); (R.N.G.)
- Instituto Nacional de Engenharia Biomédica (INEB), University of Porto, 4200-135 Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
3
|
Osten F, Bodenschatz AK, Ivaskevica K, Kröhn S, Piep B, Holler T, Teske J, Montag J, Iorga B, Weber N, Zweigerdt R, Kraft T, Meissner JD. Differential impact of substrates on myosin heavy and light chain expression in human stem cell-derived cardiomyocytes at single-cell level. J Muscle Res Cell Motil 2025:10.1007/s10974-025-09690-2. [PMID: 39948277 DOI: 10.1007/s10974-025-09690-2] [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: 11/21/2024] [Accepted: 02/01/2025] [Indexed: 04/20/2025]
Abstract
To fully exploit the potential of human pluripotent stem cell-derived cardiomyocytes, ideally they should acquire a mature, adult ventricular-like phenotype. Predominant expression of the β-isoform of myosin heavy chain (β-MyHC) and the ventricular isoform of myosin regulatory light chain 2 (MLC2v) is a marker of human adult cardiac ventricle. Yet predominant co-expression of these isoforms is rarely reported by current culture protocols. Here, we assessed the impact of different substrates on β-MyHC and MLC2v expression in single human embryonic stem cell-derived CMs (hESC-CMs). As substrates, surface materials with differing stiffness as defined by Young's modulus were combined with either laminin, a single-component coating, or Matrigel, a multi-component coating including growth factors. Semi-quantitative single-cell immunofluorescence analysis demonstrated that surfaces with supraphysiological stiffness in combination with laminin are sufficient for promotion of predominant β-MyHC expression, but not for predominant MLC2v expression in hESC-CMs. Accordingly, mechanical stimuli likely promote expression of β-MyHC in these cultures. Culture on matrices with a lower stiffness than glass in combination with growth factor-containing Matrigel led to only moderate increases in MLC2v expression, possibly more dependent on growth factors, suggesting different regulation of expression. Integrin-related downstream signal transducers, integrin-linked and cardiac troponin I-interacting kinase, as well as modulation of intracellular Ca2+-concentration and epigenetic signaling did not affect MyHC/MLC2 isoform expression. The data indicate that expression of adult ventricular markers β-MyHC and MLC2v depends on different stimuli like substrate stiffness and growth factors. To conclude, multiple stimuli appear to be necessary to promote an adult ventricular phenotype.
Collapse
Affiliation(s)
- Felix Osten
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany.
| | - Alea K Bodenschatz
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Karina Ivaskevica
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Simon Kröhn
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Birgit Piep
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Tim Holler
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Jana Teske
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Judith Montag
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Faculty of Medicine, MSB Medical School Berlin, Berlin, Germany
| | - Bogdan Iorga
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Bucharest, Romania
| | - Natalie Weber
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Joachim D Meissner
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
4
|
Pal K. Unravelling molecular mechanobiology using DNA-based fluorogenic tension sensors. J Mater Chem B 2024; 13:37-53. [PMID: 39564891 DOI: 10.1039/d4tb01858c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Investigations of the biological system have revealed many principles that govern regular life processes. Recently, the analysis of tiny mechanical forces associated with many biological processes revealed their significance in understanding biological functions. Consequently, this piqued the interest of researchers, and a series of technologies have been developed to understand biomechanical cues at the molecular level. Notable techniques include single-molecule force spectroscopy, traction force microscopy, and molecular tension sensors. Well-defined double-stranded DNA structures could possess programmable mechanical characteristics, and hence, they have become one of the central molecules in molecular tension sensor technology. With the advancement of DNA technology, DNA or nucleic acid-based robust tension sensors offer the possibility of understanding mechanobiology in the bulk to single-molecule level range with desired spatiotemporal resolution. This review presents a comprehensive account of molecular tension sensors with a special emphasis on DNA-based fluorogenic tension sensors. Along with a detailed discussion on irreversible and reversible DNA-based tension sensors and their application in super-resolution microscopy, a discussion on biomolecules associated with cellular mechanotransduction and key findings in the field are included. This review ends with an elaborate discussion on the current challenges and future prospects of molecular tension sensors.
Collapse
Affiliation(s)
- Kaushik Pal
- Biophysical Chemistry Laboratory, Department of Chemistry, Indian Institute of Technology Tirupati, Yerpedu, Tirupati, AP-517619, India.
| |
Collapse
|
5
|
DePalma SJ, Jilberto J, Stis AE, Huang DD, Lo J, Davidson CD, Chowdhury A, Kent RN, Jewett ME, Kobeissi H, Chen CS, Lejeune E, Helms AS, Nordsletten DA, Baker BM. Matrix Architecture and Mechanics Regulate Myofibril Organization, Costamere Assembly, and Contractility in Engineered Myocardial Microtissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309740. [PMID: 39558513 DOI: 10.1002/advs.202309740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/09/2024] [Indexed: 11/20/2024]
Abstract
The mechanical function of the myocardium is defined by cardiomyocyte contractility and the biomechanics of the extracellular matrix (ECM). Understanding this relationship remains an important unmet challenge due to limitations in existing approaches for engineering myocardial tissue. Here, they established arrays of cardiac microtissues with tunable mechanics and architecture by integrating ECM-mimetic synthetic, fiber matrices, and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), enabling real-time contractility readouts, in-depth structural assessment, and tissue-specific computational modeling. They found that the stiffness and alignment of matrix fibers distinctly affect the structural development and contractile function of pure iPSC-CM tissues. Further examination into the impact of fibrous matrix stiffness enabled by computational models and quantitative immunofluorescence implicates cell-ECM interactions in myofibril assembly, myofibril maturation, and notably costamere assembly, which correlates with improved contractile function of tissues. These results highlight how iPSC-CM tissue models with controllable architecture and mechanics can elucidate mechanisms of tissue maturation and disease.
Collapse
Affiliation(s)
- Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Javiera Jilberto
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Austin E Stis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Darcy D Huang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jason Lo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Aamilah Chowdhury
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Maggie E Jewett
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hiba Kobeissi
- Department of Mechanical Engineering, Boston University, Boston, MA, 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, 02215, USA
| | - Adam S Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David A Nordsletten
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, School of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, London, SE1 7EH, UK
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
6
|
Niro F, Fernandes S, Cassani M, Apostolico M, Oliver-De La Cruz J, Pereira-Sousa D, Pagliari S, Vinarsky V, Zdráhal Z, Potesil D, Pustka V, Pompilio G, Sommariva E, Rovina D, Maione AS, Bersanini L, Becker M, Rasponi M, Forte G. Fibrotic extracellular matrix impacts cardiomyocyte phenotype and function in an iPSC-derived isogenic model of cardiac fibrosis. Transl Res 2024; 273:58-77. [PMID: 39025226 PMCID: PMC11832458 DOI: 10.1016/j.trsl.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
Cardiac fibrosis occurs following insults to the myocardium and is characterized by the abnormal accumulation of non-compliant extracellular matrix (ECM), which compromises cardiomyocyte contractile activity and eventually leads to heart failure. This phenomenon is driven by the activation of cardiac fibroblasts (cFbs) to myofibroblasts and results in changes in ECM biochemical, structural and mechanical properties. The lack of predictive in vitro models of heart fibrosis has so far hampered the search for innovative treatments, as most of the cellular-based in vitro reductionist models do not take into account the leading role of ECM cues in driving the progression of the pathology. Here, we devised a single-step decellularization protocol to obtain and thoroughly characterize the biochemical and micro-mechanical properties of the ECM secreted by activated cFbs differentiated from human induced pluripotent stem cells (iPSCs). We activated iPSC-derived cFbs to the myofibroblast phenotype by tuning basic fibroblast growth factor (bFGF) and transforming growth factor beta 1 (TGF-β1) signalling and confirmed that activated cells acquired key features of myofibroblast phenotype, like SMAD2/3 nuclear shuttling, the formation of aligned alpha-smooth muscle actin (α-SMA)-rich stress fibres and increased focal adhesions (FAs) assembly. Next, we used Mass Spectrometry, nanoindentation, scanning electron and confocal microscopy to unveil the characteristic composition and the visco-elastic properties of the abundant, collagen-rich ECM deposited by cardiac myofibroblasts in vitro. Finally, we demonstrated that the fibrotic ECM activates mechanosensitive pathways in iPSC-derived cardiomyocytes, impacting on their shape, sarcomere assembly, phenotype, and calcium handling properties. We thus propose human bio-inspired decellularized matrices as animal-free, isogenic cardiomyocyte culture substrates recapitulating key pathophysiological changes occurring at the cellular level during cardiac fibrosis.
Collapse
Affiliation(s)
- Francesco Niro
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Soraia Fernandes
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Marco Cassani
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Monica Apostolico
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Jorge Oliver-De La Cruz
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Daniel Pereira-Sousa
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic
| | - Stefania Pagliari
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Vladimir Vinarsky
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Zbyněk Zdráhal
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - David Potesil
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Vaclav Pustka
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy; Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Davide Rovina
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Angela Serena Maione
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | | | - Marco Rasponi
- Department of Electronics, Informatics and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK.
| |
Collapse
|
7
|
Dong Y, Lu R, Cao H, Zhang J, Wu X, Deng Y, Li JD. Deficiency in Prader-Willi syndrome gene necdin leads to attenuated cardiac contractility. iScience 2024; 27:109974. [PMID: 38832028 PMCID: PMC11144731 DOI: 10.1016/j.isci.2024.109974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/02/2024] [Accepted: 05/10/2024] [Indexed: 06/05/2024] Open
Abstract
Prader-Willi syndrome (PWS) is a genetic disorder characterized by behavioral disturbances, hyperphagia, and intellectual disability. Several surveys indicate that PWS is also associated with cardiac abnormalities, possibly contributing to a high incidence of sudden death. However, the pathological mechanisms underlying cardiac dysfunction in PWS remain unclear. In this study, we found that deficiency in necdin, an intronless gene within PWS region, led to heart systolic and diastolic dysfunction in mice. Through yeast two-hybrid screening, we identified an interaction between necdin and non-muscle myosin regulatory light chain 12a/b (MYL12 A/B). We further showed that necdin stabilized MYL12 A/B via SGT1-heat shock protein 90 (HSP90) chaperone machinery. The zebrafish lacking the MYL12 A/B analog, MYL12.1, exhibited impaired heart function, while cardiac-specific overexpression of MYL12A normalized the heart dysfunction in necdin-deficient mice. Our findings revealed necdin dysfunction as a contributing factor to cardiomyopathy in PWS patients and emphasized the importance of HSP90 chaperone machinery and non-muscle myosin in heart fitness.
Collapse
Affiliation(s)
- Yufan Dong
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central South University, Changsha 410078, Hunan, P.R. China
| | - Renbin Lu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central South University, Changsha 410078, Hunan, P.R. China
| | - Hui Cao
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- Laboratory of Zebrafish Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jing Zhang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, P.R. China
- Hunan Key Laboratory of Animal Models for Human Diseases, Changsha 410078, Hunan, P.R. China
- Hunan Key Laboratory of Medical Genetics, Changsha 410078, Hunan, P.R. China
| | - Xiushan Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- Laboratory of Zebrafish Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yun Deng
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Normal University, Changsha, China
- Laboratory of Zebrafish Genetics, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Jia-Da Li
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, P.R. China
- Hunan Key Laboratory of Animal Models for Human Diseases, Changsha 410078, Hunan, P.R. China
- Hunan Key Laboratory of Medical Genetics, Changsha 410078, Hunan, P.R. China
- Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, Changsha 410078, Hunan, P.R. China
- National Clinical Research Center for Geriatric Disorder, Xiangya Hospital, Central South University, Changsha 410078, Hunan, P.R. China
| |
Collapse
|
8
|
Swiatlowska P, Tipping W, Marhuenda E, Severi P, Fomin V, Yang Z, Xiao Q, Graham D, Shanahan C, Iskratsch T. Hypertensive Pressure Mechanosensing Alone Triggers Lipid Droplet Accumulation and Transdifferentiation of Vascular Smooth Muscle Cells to Foam Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308686. [PMID: 38145971 PMCID: PMC10916670 DOI: 10.1002/advs.202308686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Indexed: 12/27/2023]
Abstract
Arterial Vascular smooth muscle cells (VSMCs) play a central role in the onset and progression of atherosclerosis. Upon exposure to pathological stimuli, they can take on alternative phenotypes that, among others, have been described as macrophage like, or foam cells. VSMC foam cells make up >50% of all arterial foam cells and have been suggested to retain an even higher proportion of the cell stored lipid droplets, further leading to apoptosis, secondary necrosis, and an inflammatory response. However, the mechanism of VSMC foam cell formation is still unclear. Here, it is identified that mechanical stimulation through hypertensive pressure alone is sufficient for the phenotypic switch. Hyperspectral stimulated Raman scattering imaging demonstrates rapid lipid droplet formation and changes to lipid metabolism and changes are confirmed in ABCA1, KLF4, LDLR, and CD68 expression, cell proliferation, and migration. Further, a mechanosignaling route is identified involving Piezo1, phospholipid, and arachidonic acid signaling, as well as epigenetic regulation, whereby CUT&Tag epigenomic analysis confirms changes in the cells (lipid) metabolism and atherosclerotic pathways. Overall, the results show for the first time that VSMC foam cell formation can be triggered by mechanical stimulation alone, suggesting modulation of mechanosignaling can be harnessed as potential therapeutic strategy.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - William Tipping
- Department of Pure and Applied ChemistryUniversity of StrathclydeGlasgowG1 1QAUK
| | - Emilie Marhuenda
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| | - Paolo Severi
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
- Department of Translational MedicineLaboratory for Technologies of Advanced Therapies (LTTA)University of FerraraFerrara44121Italy
| | | | - Zhisheng Yang
- William Harvey Research InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Qingzhong Xiao
- William Harvey Research InstituteQueen Mary University of LondonLondonEC1M 6BQUK
| | - Duncan Graham
- Department of Pure and Applied ChemistryUniversity of StrathclydeGlasgowG1 1QAUK
| | - Cathy Shanahan
- School of Cardiovascular Medicine and SciencesKing's College LondonLondonSE5 9NUUK
| | - Thomas Iskratsch
- School of Engineering and Materials ScienceQueen Mary University of LondonLondonE1 4NSUK
| |
Collapse
|
9
|
Faleeva M, Ahmad S, Theofilatos K, Lynham S, Watson G, Whitehead M, Marhuenda E, Iskratsch T, Cox S, Shanahan CM. Sox9 Accelerates Vascular Aging by Regulating Extracellular Matrix Composition and Stiffness. Circ Res 2024; 134:307-324. [PMID: 38179698 PMCID: PMC10826924 DOI: 10.1161/circresaha.123.323365] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Vascular calcification and increased extracellular matrix (ECM) stiffness are hallmarks of vascular aging. Sox9 (SRY-box transcription factor 9) has been implicated in vascular smooth muscle cell (VSMC) osteo/chondrogenic conversion; however, its relationship with aging and calcification has not been studied. METHODS Immunohistochemistry was performed on human aortic samples from young and aged patients. Young and senescent primary human VSMCs were induced to produce ECM, and Sox9 expression was manipulated using adenoviral overexpression and depletion. ECM properties were characterized using atomic force microscopy and proteomics, and VSMC phenotype on hydrogels and the ECM were examined using confocal microscopy. RESULTS In vivo, Sox9 was not spatially associated with vascular calcification but correlated with the senescence marker p16 (cyclin-dependent kinase inhibitor 2A). In vitro Sox9 showed mechanosensitive responses with increased expression and nuclear translocation in senescent cells and on stiff matrices. Sox9 was found to regulate ECM stiffness and organization by orchestrating changes in collagen (Col) expression and reducing VSMC contractility, leading to the formation of an ECM that mirrored that of senescent cells. These ECM changes promoted phenotypic modulation of VSMCs, whereby senescent cells plated on ECM synthesized from cells depleted of Sox9 returned to a proliferative state, while proliferating cells on a matrix produced by Sox9 expressing cells showed reduced proliferation and increased DNA damage, reiterating features of senescent cells. LH3 (procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3) was identified as an Sox9 target and key regulator of ECM stiffness. LH3 is packaged into extracellular vesicles and Sox9 promotes extracellular vesicle secretion, leading to increased LH3 deposition within the ECM. CONCLUSIONS These findings highlight the crucial role of ECM structure and composition in regulating VSMC phenotype. We identify a positive feedback cycle, whereby cellular senescence and increased ECM stiffening promote Sox9 expression, which, in turn, drives further ECM modifications to further accelerate stiffening and senescence.
Collapse
Affiliation(s)
- Maria Faleeva
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Sadia Ahmad
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Konstantinos Theofilatos
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Steven Lynham
- Proteomics Facility, Centre of Excellence for Mass Spectrometry (S.L.) King’s College London, United Kingdom
| | - Gabriel Watson
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Meredith Whitehead
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Emilie Marhuenda
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Thomas Iskratsch
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Susan Cox
- Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine (S.C.) King’s College London, United Kingdom
| | - Catherine M. Shanahan
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| |
Collapse
|
10
|
Grimes KM, Maillet M, Swoboda CO, Bowers SLK, Millay DP, Molkentin JD. MEK1-ERK1/2 signaling regulates the cardiomyocyte non-sarcomeric actin cytoskeletal network. Am J Physiol Heart Circ Physiol 2024; 326:H180-H189. [PMID: 37999644 PMCID: PMC11551003 DOI: 10.1152/ajpheart.00612.2023] [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: 09/29/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
During select pathological conditions, the heart can hypertrophy and remodel in either a dilated or concentric ventricular geometry, which is associated with lengthening or widening of cardiomyocytes, respectively. The mitogen-activated protein kinase kinase 1 (MEK1) and extracellular signal-related kinase 1 and 2 (ERK1/2) pathway has been implicated in these differential types of growth such that cardiac overexpression of activated MEK1 causes profound concentric hypertrophy and cardiomyocyte thickening, while genetic ablation of the genes encoding ERK1/2 in the mouse heart causes dilation and cardiomyocyte lengthening. However, the mechanisms by which this kinase signaling pathway controls cardiomyocyte directional growth as well as its downstream effectors are poorly understood. To investigate this, we conducted an unbiased phosphoproteomic screen in cultured neonatal rat ventricular myocytes treated with an activated MEK1 adenovirus, the MEK1 inhibitor U0126, or an eGFP adenovirus control. Bioinformatic analysis identified cytoskeletal-related proteins as the largest subset of differentially phosphorylated proteins. Phos-tag and traditional Western blotting were performed to confirm that many cytoskeletal proteins displayed changes in phosphorylation with manipulations in MEK1-ERK1/2 signaling. From this, we hypothesized that the actin cytoskeleton would be changed in vivo in the mouse heart. Indeed, we found that activated MEK1 transgenic mice and gene-deleted mice lacking ERK1/2 protein had enhanced non-sarcomeric actin expression in cardiomyocytes compared with wild-type control hearts. Consistent with these results, cytoplasmic β- and γ-actin were increased at the subcortical intracellular regions of adult cardiomyocytes. Together, these data suggest that MEK1-ERK1/2 signaling influences the non-sarcomeric cytoskeletal actin network, which may be important for facilitating the growth of cardiomyocytes in length and/or width.NEW & NOTEWORTHY Here, we performed an unbiased analysis of the total phosphoproteome downstream of MEK1-ERK1/2 kinase signaling in cardiomyocytes. Pathway analysis suggested that proteins of the non-sarcomeric cytoskeleton were the most differentially affected. We showed that cytoplasmic β-actin and γ-actin isoforms, regulated by MEK1-ERK1/2, are localized to the subcortical space at both lateral membranes and intercalated discs of adult cardiomyocytes suggesting how MEK1-ERK1/2 signaling might underlie directional growth of adult cardiomyocytes.
Collapse
Affiliation(s)
- Kelly M Grimes
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States
| | - Marjorie Maillet
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States
| | - Casey O Swoboda
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States
| | - Stephanie L K Bowers
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States
| | - Doug P Millay
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States
| |
Collapse
|
11
|
Li Q, Chen Z, Zhang Y, Ding S, Ding H, Wang L, Xie Z, Fu Y, Wei M, Liu S, Chen J, Wang X, Gu Z. Imaging cellular forces with photonic crystals. Nat Commun 2023; 14:7369. [PMID: 37963911 PMCID: PMC10646022 DOI: 10.1038/s41467-023-43090-9] [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: 01/27/2022] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
Current techniques for visualizing and quantifying cellular forces have limitations in live cell imaging, throughput, and multi-scale analysis, which impede progress in cell force research and its practical applications. We developed a photonic crystal cellular force microscopy (PCCFM) to image vertical cell forces over a wide field of view (1.3 mm ⨯ 1.0 mm, a 10 ⨯ objective image) at high speed (about 20 frames per second) without references. The photonic crystal hydrogel substrate (PCS) converts micro-nano deformations into perceivable color changes, enabling in situ visualization and quantification of tiny vertical cell forces with high throughput. It enabled long-term, cross-scale monitoring from subcellular focal adhesions to tissue-level cell sheets and aggregates.
Collapse
Affiliation(s)
- Qiwei Li
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Zaozao Chen
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
- Institute of Biomaterials and Medical Devices, Southeast University, 215163, Suzhou, Jiangsu, China
| | - Ying Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Shuang Ding
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Haibo Ding
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Luping Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
- Faculty of Sports Science, Ningbo University, 315211, Ningbo, China
| | - Zhuoying Xie
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Yifu Fu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Mengxiao Wei
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Shengnan Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Jialun Chen
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Xuan Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 210096, Nanjing, Jiangsu, China.
- Institute of Biomaterials and Medical Devices, Southeast University, 215163, Suzhou, Jiangsu, China.
| |
Collapse
|
12
|
DePalma SJ, Jillberto J, Stis AE, Huang DD, Lo J, Davidson CD, Chowdhury A, Jewett ME, Kobeissi H, Chen CS, Lejeune E, Helms AS, Nordsletten DA, Baker BM. Matrix architecture and mechanics regulate myofibril organization, costamere assembly, and contractility of engineered myocardial microtissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.20.563346. [PMID: 37961415 PMCID: PMC10634701 DOI: 10.1101/2023.10.20.563346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
The mechanical function of the myocardium is defined by cardiomyocyte contractility and the biomechanics of the extracellular matrix (ECM). Understanding this relationship remains an important unmet challenge due to limitations in existing approaches for engineering myocardial tissue. Here, we established arrays of cardiac microtissues with tunable mechanics and architecture by integrating ECM-mimetic synthetic, fiber matrices and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), enabling real-time contractility readouts, in-depth structural assessment, and tissue-specific computational modeling. We find that the stiffness and alignment of matrix fibers distinctly affect the structural development and contractile function of pure iPSC-CM tissues. Further examination into the impact of fibrous matrix stiffness enabled by computational models and quantitative immunofluorescence implicates cell-ECM interactions in myofibril assembly and notably costamere assembly, which correlates with improved contractile function of tissues. These results highlight how iPSC-CM tissue models with controllable architecture and mechanics can inform the design of translatable regenerative cardiac therapies.
Collapse
|
13
|
Ross JA, Arcos-Villacis N, Battey E, Boogerd C, Orellana CA, Marhuenda E, Swiatlowska P, Hodzic D, Prin F, Mohun T, Catibog N, Tapia O, Gerace L, Iskratsch T, Shah AM, Stroud MJ. Lem2 is essential for cardiac development by maintaining nuclear integrity. Cardiovasc Res 2023; 119:2074-2088. [PMID: 37067297 PMCID: PMC10478753 DOI: 10.1093/cvr/cvad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 01/19/2023] [Accepted: 02/08/2023] [Indexed: 04/18/2023] Open
Abstract
AIMS Nuclear envelope integrity is essential for the compartmentalization of the nucleus and cytoplasm. Importantly, mutations in genes encoding nuclear envelope (NE) and associated proteins are the second highest cause of familial dilated cardiomyopathy. One such NE protein that causes cardiomyopathy in humans and affects mouse heart development is Lem2. However, its role in the heart remains poorly understood. METHODS AND RESULTS We generated mice in which Lem2 was specifically ablated either in embryonic cardiomyocytes (Lem2 cKO) or in adult cardiomyocytes (Lem2 iCKO) and carried out detailed physiological, tissue, and cellular analyses. High-resolution episcopic microscopy was used for three-dimensional reconstructions and detailed morphological analyses. RNA-sequencing and immunofluorescence identified altered pathways and cellular phenotypes, and cardiomyocytes were isolated to interrogate nuclear integrity in more detail. In addition, echocardiography provided a physiological assessment of Lem2 iCKO adult mice. We found that Lem2 was essential for cardiac development, and hearts from Lem2 cKO mice were morphologically and transcriptionally underdeveloped. Lem2 cKO hearts displayed high levels of DNA damage, nuclear rupture, and apoptosis. Crucially, we found that these defects were driven by muscle contraction as they were ameliorated by inhibiting myosin contraction and L-type calcium channels. Conversely, reducing Lem2 levels to ∼45% in adult cardiomyocytes did not lead to overt cardiac dysfunction up to 18 months of age. CONCLUSIONS Our data suggest that Lem2 is critical for integrity at the nascent NE in foetal hearts, and protects the nucleus from the mechanical forces of muscle contraction. In contrast, the adult heart is not detectably affected by partial Lem2 depletion, perhaps owing to a more established NE and increased adaptation to mechanical stress. Taken together, these data provide insights into mechanisms underlying cardiomyopathy in patients with mutations in Lem2 and cardio-laminopathies in general.
Collapse
Affiliation(s)
- Jacob A Ross
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Nathaly Arcos-Villacis
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Edmund Battey
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
- Centre of Human and Applied Physiological Sciences, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, UK
| | - Cornelis Boogerd
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), University Medical Center Utrecht, Utrecht 3584 CT, The Netherlands
| | - Constanza Avalos Orellana
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Emilie Marhuenda
- Division of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Pamela Swiatlowska
- Division of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Didier Hodzic
- Department of Developmental Biology, Washington University School of Medicine, 660S. Euclid Avenue, St Louis, MO 63110, USA
| | - Fabrice Prin
- Crick Advanced Light Microscopy Facility, The Francis Crick Institute, London NW1 1AT, UK
| | - Tim Mohun
- Crick Advanced Light Microscopy Facility, The Francis Crick Institute, London NW1 1AT, UK
| | - Norman Catibog
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Olga Tapia
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
- Research Group on Foods, Nutritional Biochemistry and Health, Universidad Europea del Atlántico, Santander 39011, Spain
- Department of Basic Medical Sciences, Institute of Biomedical Technologies, University of La Laguna, Tenerife 38200, Spain
| | - Larry Gerace
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Ajay M Shah
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Matthew J Stroud
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular & Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
14
|
Chen Y, Yuan Y, Chen Y, Jiang X, Hua X, Chen Z, Wang J, Liu H, Zhou Q, Yu Y, Yang Z, Yu Y, Wang Y, Wang Q, Li Y, Chen J, Wang Y. Novel signaling axis of FHOD1-RNF213-Col1α/Col3α in the pathogenesis of hypertension-induced tunica media thickening. J Mol Cell Cardiol 2023; 182:57-72. [PMID: 37482037 DOI: 10.1016/j.yjmcc.2023.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/05/2023] [Accepted: 07/15/2023] [Indexed: 07/25/2023]
Abstract
Hypertension-induced tunica media thickening (TMT) is the most important fundamental for the subsequent complications like stroke and cardiovascular diseases. Pathogenically, TMT originates from both vascular smooth muscle cells (VSMCs) hypertrophy due to synthesizing more amount of intracellular contractile proteins and excess secretion of extracellular matrix. However, what key molecules are involved in the pathogenesis of TMT is unknown. We hypothesize that formin homology 2 domain-containing protein 1 (FHOD1), an amply expressed mediator for assembly of thin actin filament in VSMCs, is a key regulator for the pathogenesis of TMT. In this study, we found that FHOD1 expression and its phosphorylation/activation were both upregulated in the arteries of three kinds of hypertensive rats. Ang-II induced actin filament formation and hypertrophy through activation and upregulation of FHOD1 in VSMCs. Active FHOD1-mediated actin filament assembly and secretions of collagen-1α/collagen-3α played crucial roles in Ang-II-induced VSMCs hypertrophy in vitro and hypertensive TMT in vivo. Proteomics demonstrated that activated FL-FHOD1 or its C-terminal diaphanous-autoregulatory domain significantly upregulated RNF213 (ring finger protein 213), a 591-kDa cytosolic E3 ubiquitin ligase with its loss-of-functional mutations being a susceptibility gene for Moyamoya disease which has prominent tunica media thinning in both intracranial and systemic arteries. Mechanistically, activated FHOD1 upregulated its downstream effector RNF213 independently of its classical pathway of decreasing G-actin/F-actin ratio, transcription, and translation, but dependently on its C-terminus-mediated stabilization of RNF213 protein. FHOD1-RNF213 signaling dramatically promoted collagen-1α/collagen-3α syntheses in VSMCs. Our results discovered a novel signaling axis of FHOD1-RNF213-collagen-1α/collagen-3α and its key role in the pathogenesis of hypertensive TMT.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuchan Yuan
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yuhan Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xueze Jiang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Xuesheng Hua
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhiyong Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Julie Wang
- Department of Computer Science, Brown University, Providence, RI 02912, USA
| | - Hua Liu
- Department of Intensive Care Med, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Qing Zhou
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Ying Yu
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Zhenwei Yang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yi Yu
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yongqin Wang
- Division of Rheumatology and Immunology, University of Toledo Medical center, 3120 Glendale Avenue, Toledo, OH 43614, USA
| | - Qunshan Wang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yigang Li
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Jie Chen
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| | - Yuepeng Wang
- Molecular Cardiology Research Laboratory, Department of Cardiology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
15
|
De Silva S, Fan Z, Kang B, Shanahan CM, Zhang Q. Nesprin-1: novel regulator of striated muscle nuclear positioning and mechanotransduction. Biochem Soc Trans 2023; 51:1331-1345. [PMID: 37171063 PMCID: PMC10317153 DOI: 10.1042/bst20221541] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023]
Abstract
Nesprins (nuclear envelope spectrin repeat proteins) are multi-isomeric scaffolding proteins. Giant nesprin-1 and -2 localise to the outer nuclear membrane, interact with SUN (Sad1p/UNC-84) domain-containing proteins at the inner nuclear membrane to form the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex, which, in association with lamin A/C and emerin, mechanically couples the nucleus to the cytoskeleton. Despite ubiquitous expression of nesprin giant isoforms, pathogenic mutations in nesprin-1 and -2 are associated with tissue-specific disorders, particularly related to striated muscle such as dilated cardiomyopathy and Emery-Dreifuss muscular dystrophy. Recent evidence suggests this muscle-specificity might be attributable in part, to the small muscle specific isoform, nesprin-1α2, which has a novel role in striated muscle function. Our current understanding of muscle-specific functions of nesprin-1 and its isoforms will be summarised in this review to provide insight into potential pathological mechanisms of nesprin-related muscle disease and may inform potential targets of therapeutic modulation.
Collapse
Affiliation(s)
- Shanelle De Silva
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| | - Zhijuan Fan
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
- Clinical Laboratory, Tianjin Third Central Hospital, Tianjin 300170, China
| | - Baoqiang Kang
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| | - Catherine M. Shanahan
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| | - Qiuping Zhang
- King's College London British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, London SE5 9NU, U.K
| |
Collapse
|
16
|
Peng L, Matellan C, Bosch‐Fortea M, Gonzalez‐Molina J, Frigerio M, Salentinig S, del Rio Hernandez A, Gautrot JE. Mesenchymal Stem Cells Sense the Toughness of Nanomaterials and Interfaces. Adv Healthc Mater 2023; 12:e2203297. [PMID: 36717365 PMCID: PMC11468436 DOI: 10.1002/adhm.202203297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/20/2023] [Indexed: 02/01/2023]
Abstract
Stem cells are known to sense and respond to the mechanical properties of biomaterials. In turn, cells exert forces on their environment that can lead to striking changes in shape, size and contraction of associated tissues, and may result in mechanical disruption and functional failure. However, no study has so far correlated stem cell phenotype and biomaterials toughness. Indeed, disentangling toughness-mediated cell response from other mechanosensing processes has remained elusive as it is particularly challenging to uncouple Youngs' or shear moduli from toughness, within a range relevant to cell-generated forces. In this report, it is shown how the design of the macromolecular architecture of polymer nanosheets regulates interfacial toughness, independently of interfacial shear storage modulus, and how this controls the expansion of mesenchymal stem cells at liquid interfaces. The viscoelasticity and toughness of poly(l-lysine) nanosheets assembled at liquid-liquid interfaces is characterised via interfacial shear rheology. The local (microscale) mechanics of nanosheets are characterised via magnetic tweezer-assisted interfacial microrheology and the thickness of these assemblies is determined from in situ ellipsometry. Finally, the response of mesenchymal stem cells to adhesion and culture at corresponding interfaces is investigated via immunostaining and confocal microscopy.
Collapse
Affiliation(s)
- Lihui Peng
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| | - Carlos Matellan
- School of Engineering and Materials ScienceQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
| | - Minerva Bosch‐Fortea
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| | - Jordi Gonzalez‐Molina
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| | - Matteo Frigerio
- Department of ChemistryUniversity of FribourgChemin du Musée 9Fribourg1700Switzerland
| | - Stefan Salentinig
- Department of ChemistryUniversity of FribourgChemin du Musée 9Fribourg1700Switzerland
| | - Armando del Rio Hernandez
- School of Engineering and Materials ScienceQueen MaryUniversity of LondonMile End RoadLondonE1 4NSUK
| | - Julien E. Gautrot
- Institute of BioengineeringQueen Mary University of LondonMile End RoadE1 4NSLondonUK
- Cellular and Molecular Biomechanical LaboratoryDepartment of BioengineeringImperial College LondonLondonSW7 2AZUK
| |
Collapse
|
17
|
Meng H, Fu S, Ferreira MB, Hou Y, Pearce OM, Gavara N, Knight MM. YAP activation inhibits inflammatory signalling and cartilage breakdown associated with reduced primary cilia expression. Osteoarthritis Cartilage 2023; 31:600-612. [PMID: 36368426 DOI: 10.1016/j.joca.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/14/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To clarify the role of YAP in modulating cartilage inflammation and degradation and the involvement of primary cilia and associated intraflagellar transport (IFT). METHODS Isolated primary chondrocytes were cultured on substrates of different stiffness (6-1000 kPa) or treated with YAP agonist lysophosphatidic acid (LPA) or YAP antagonist verteporfin (VP), or genetically modified by YAP siRNA, all ± IL1β. Nitric oxide (NO) and prostaglandin E2 (PGE2) release were measured to monitor IL1β response. YAP activity was quantified by YAP nuclear/cytoplasmic ratio and percentage of YAP-positive cells. Mechanical properties of cartilage explants were tested to confirm cartilage degradation. The involvement of primary cilia and IFT was analysed using IFT88 siRNA and ORPK cells with hypomorphic mutation of IFT88. RESULTS Treatment with LPA, or increasing polydimethylsiloxane (PDMS) substrate stiffness, activated YAP nuclear expression and inhibited IL1β-induced release of NO and PGE2, in isolated chondrocytes. Treatment with LPA also inhibited IL1β-mediated inflammatory signalling in cartilage explants and prevented matrix degradation and the loss of cartilage biomechanics. YAP activation reduced expression of primary cilia, knockdown of YAP in the absence of functional cilia/IFT failed to induce an inflammatory response. CONCLUSIONS We demonstrate that both pharmaceutical and mechanical activation of YAP blocks pro-inflammatory signalling induced by IL1β and prevents cartilage breakdown and the loss of biomechanical functionality. This is associated with reduced expression of primary cilia revealing a potential anti-inflammatory mechanism with novel therapeutic targets for treatment of osteoarthritis (OA).
Collapse
Affiliation(s)
- H Meng
- School of Engineering and Materials Science, Queen Mary University of London, London, UK.
| | - S Fu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - M B Ferreira
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Y Hou
- School of Engineering and Materials Science, Queen Mary University of London, London, UK; Centre for Predictive in Vitro Models, Queen Mary University of London, London, UK
| | - O M Pearce
- Barts Cancer Institute, School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - N Gavara
- Serra-Hunter Program, Biophysics and Bioengineering Unit, Department of Biomedicine, Medical School, University of Barcelona, Barcelona, Spain
| | - M M Knight
- School of Engineering and Materials Science, Queen Mary University of London, London, UK; Centre for Predictive in Vitro Models, Queen Mary University of London, London, UK
| |
Collapse
|
18
|
Shoykhet M, Dervishi O, Menauer P, Hiermaier M, Moztarzadeh S, Osterloh C, Ludwig RJ, Williams T, Gerull B, Kääb S, Clauss S, Schüttler D, Waschke J, Yeruva S. EGFR inhibition leads to enhanced desmosome assembly and cardiomyocyte cohesion via ROCK activation. JCI Insight 2023; 8:163763. [PMID: 36795511 PMCID: PMC10070108 DOI: 10.1172/jci.insight.163763] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/15/2023] [Indexed: 02/17/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial heart disease partly caused by impaired desmosome turnover. Thus, stabilization of desmosome integrity may provide new treatment options. Desmosomes, apart from cellular cohesion, provide the structural framework of a signaling hub. Here, we investigated the role of the epidermal growth factor receptor (EGFR) in cardiomyocyte cohesion. We inhibited EGFR under physiological and pathophysiological conditions using the murine plakoglobin-KO AC model, in which EGFR was upregulated. EGFR inhibition enhanced cardiomyocyte cohesion. Immunoprecipitation showed an interaction of EGFR and desmoglein 2 (DSG2). Immunostaining and atomic force microscopy (AFM) revealed enhanced DSG2 localization and binding at cell borders upon EGFR inhibition. Enhanced area composita length and desmosome assembly were observed upon EGFR inhibition, confirmed by enhanced DSG2 and desmoplakin (DP) recruitment to cell borders. PamGene Kinase assay performed in HL-1 cardiomyocytes treated with erlotinib, an EGFR inhibitor, revealed upregulation of Rho-associated protein kinase (ROCK). Erlotinib-mediated desmosome assembly and cardiomyocyte cohesion were abolished upon ROCK inhibition. Thus, inhibiting EGFR and, thereby, stabilizing desmosome integrity via ROCK might provide treatment options for AC.
Collapse
Affiliation(s)
- Maria Shoykhet
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Orsela Dervishi
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Philipp Menauer
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sina Moztarzadeh
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Colin Osterloh
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Dominik Schüttler
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
19
|
Nordmeyer S, Kraus M, Ziehm M, Kirchner M, Schafstedde M, Kelm M, Niquet S, Stephen MM, Baczko I, Knosalla C, Schapranow MP, Dittmar G, Gotthardt M, Falcke M, Regitz-Zagrosek V, Kuehne T, Mertins P. Disease- and sex-specific differences in patients with heart valve disease: a proteome study. Life Sci Alliance 2023; 6:e202201411. [PMID: 36627164 PMCID: PMC9834574 DOI: 10.26508/lsa.202201411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Pressure overload in patients with aortic valve stenosis and volume overload in mitral valve regurgitation trigger specific forms of cardiac remodeling; however, little is known about similarities and differences in myocardial proteome regulation. We performed proteome profiling of 75 human left ventricular myocardial biopsies (aortic stenosis = 41, mitral regurgitation = 17, and controls = 17) using high-resolution tandem mass spectrometry next to clinical and hemodynamic parameter acquisition. In patients of both disease groups, proteins related to ECM and cytoskeleton were more abundant, whereas those related to energy metabolism and proteostasis were less abundant compared with controls. In addition, disease group-specific and sex-specific differences have been observed. Male patients with aortic stenosis showed more proteins related to fibrosis and less to energy metabolism, whereas female patients showed strong reduction in proteostasis-related proteins. Clinical imaging was in line with proteomic findings, showing elevation of fibrosis in both patient groups and sex differences. Disease- and sex-specific proteomic profiles provide insight into cardiac remodeling in patients with heart valve disease and might help improve the understanding of molecular mechanisms and the development of individualized treatment strategies.
Collapse
Affiliation(s)
- Sarah Nordmeyer
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Milena Kraus
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Matthias Ziehm
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marie Schafstedde
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Kelm
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sylvia Niquet
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mariet Mathew Stephen
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Istvan Baczko
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Christoph Knosalla
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart, Department of Cardiothoracic and Vascular Surgery, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Matthieu-P Schapranow
- Hasso Plattner Institute for Digital Engineering, Digital Health Center, University of Potsdam, Potsdam, Germany
| | - Gunnar Dittmar
- Proteomics of Cellular Signaling, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Michael Gotthardt
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Neuromuscular and Cardiovascular Cell Biology, Berlin, Germany
| | - Martin Falcke
- Max Delbrück Center for Molecular Medicine, Mathematical Cell Physiology, Berlin, Germany
| | - Vera Regitz-Zagrosek
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Cardiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Titus Kuehne
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Institute for Cardiovascular Computer-Assisted Medicine, Berlin, Germany
- Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Department of Congenital Heart Disease - Pediatric Cardiology, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Philipp Mertins
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Proteomics Platform, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
20
|
Ringström N, Edling C, Nalesso G, Jeevaratnam K. Framing Heartaches: The Cardiac ECM and the Effects of Age. Int J Mol Sci 2023; 24:4713. [PMID: 36902143 PMCID: PMC10003270 DOI: 10.3390/ijms24054713] [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: 01/12/2023] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 03/05/2023] Open
Abstract
The cardiac extracellular matrix (ECM) is involved in several pathological conditions, and age itself is also associated with certain changes in the heart: it gets larger and stiffer, and it develops an increased risk of abnormal intrinsic rhythm. This, therefore, makes conditions such as atrial arrythmia more common. Many of these changes are directly related to the ECM, yet the proteomic composition of the ECM and how it changes with age is not fully resolved. The limited research progress in this field is mainly due to the intrinsic challenges in unravelling tightly bound cardiac proteomic components and also the time-consuming and costly dependency on animal models. This review aims to give an overview of the composition of the cardiac ECM, how different components aid the function of the healthy heart, how the ECM is remodelled and how it is affected by ageing.
Collapse
Affiliation(s)
| | | | | | - Kamalan Jeevaratnam
- Faculty of Health and Medical Science, University of Surrey, Guildford GU2 7AL, UK
| |
Collapse
|
21
|
Chrysanthou A, Kanso H, Zhong W, Shang L, Gautrot JE. Supercharged Protein Nanosheets for Cell Expansion on Bioemulsions. ACS APPLIED MATERIALS & INTERFACES 2023; 15:2760-2770. [PMID: 36598358 PMCID: PMC9869332 DOI: 10.1021/acsami.2c20188] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/21/2022] [Indexed: 05/27/2023]
Abstract
Cell culture at liquid-liquid interfaces, for example, at the surface of oil microdroplets, is an attractive strategy to scale up adherent cell manufacturing while replacing the use of microplastics. Such a process requires the adhesion of cells at interfaces stabilized and reinforced by protein nanosheets displaying not only high elasticity but also presenting cell adhesive ligands able to bind integrin receptors. In this report, supercharged albumins are found to form strong elastic protein nanosheets when co-assembling with the co-surfactant pentafluorobenzoyl chloride (PFBC) and mediate extracellular matrix (ECM) protein adsorption and cell adhesion. The interfacial mechanical properties and elasticity of supercharged nanosheets are characterized by interfacial rheology, and behaviors are compared to those of native bovine serum albumin, human serum albumin, and α-lactalbumin. The impact of PFBC on such assembly is investigated. ECM protein adsorption to resulting supercharged nanosheets is then quantified via surface plasmon resonance and fluorescence microscopy, demonstrating that the dual role supercharged albumins are proposed to play as scaffold protein structuring liquid-liquid interfaces and substrates for the capture of ECM molecules. Finally, the adhesion and proliferation of primary human epidermal stem cells are investigated, at pinned droplets, as well as on bioemulsions stabilized by corresponding supercharged nanosheets. This study demonstrates the potential of supercharged proteins for the engineering of biointerfaces for stem cell manufacturing and draws structure-property relationships that will guide further engineering of associated systems.
Collapse
Affiliation(s)
- Alexandra Chrysanthou
- Institute
of Bioengineering, Queen Mary, University
of London, Mile End Road, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary, University of London, Mile End Road, London E1 4NS, U.K.
| | - Hassan Kanso
- Institute
of Bioengineering, Queen Mary, University
of London, Mile End Road, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary, University of London, Mile End Road, London E1 4NS, U.K.
| | - Wencheng Zhong
- State
Key Laboratory of Solidification Processing, School of Materials Science
and Engineering, Northwestern Polytechnical
University and Shaanxi Joint Laboratory of Graphene (NPU), Xi’an 710072, China
| | - Li Shang
- State
Key Laboratory of Solidification Processing, School of Materials Science
and Engineering, Northwestern Polytechnical
University and Shaanxi Joint Laboratory of Graphene (NPU), Xi’an 710072, China
- NPU-QMUL
Joint Research Institute of Advanced Materials and Structures (JRI-AMAS), Northwestern Polytechnical University, Xi’an 710072, China
| | - Julien E. Gautrot
- Institute
of Bioengineering, Queen Mary, University
of London, Mile End Road, London E1 4NS, U.K.
- School
of Engineering and Materials Science, Queen
Mary, University of London, Mile End Road, London E1 4NS, U.K.
| |
Collapse
|
22
|
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
|
23
|
Hawkes W, Marhuenda E, Reynolds P, O'Neill C, Pandey P, Samuel Wilson DG, Freeley M, Huang D, Hu J, Gondarenko S, Hone J, Gadegaard N, Palma M, Iskratsch T. Regulation of cardiomyocyte adhesion and mechanosignalling through distinct nanoscale behaviour of integrin ligands mimicking healthy or fibrotic extracellular matrix. Philos Trans R Soc Lond B Biol Sci 2022; 377:20220021. [PMID: 36189804 PMCID: PMC9527911 DOI: 10.1098/rstb.2022.0021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/12/2022] [Indexed: 12/21/2022] Open
Abstract
The stiffness of the cardiovascular environment changes during ageing and in disease and contributes to disease incidence and progression. Changing collagen expression and cross-linking regulate the rigidity of the cardiac extracellular matrix (ECM). Additionally, basal lamina glycoproteins, especially laminin and fibronectin regulate cardiomyocyte adhesion formation, mechanics and mechanosignalling. Laminin is abundant in the healthy heart, but fibronectin is increasingly expressed in the fibrotic heart. ECM receptors are co-regulated with the changing ECM. Owing to differences in integrin dynamics, clustering and downstream adhesion formation this is expected to ultimately influence cardiomyocyte mechanosignalling; however, details remain elusive. Here, we sought to investigate how different cardiomyocyte integrin/ligand combinations affect adhesion formation, traction forces and mechanosignalling, using a combination of uniformly coated surfaces with defined stiffness, polydimethylsiloxane nanopillars, micropatterning and specifically designed bionanoarrays for precise ligand presentation. Thereby we found that the adhesion nanoscale organization, signalling and traction force generation of neonatal rat cardiomyocytes (which express both laminin and fibronectin binding integrins) are strongly dependent on the integrin/ligand combination. Together our data indicate that the presence of fibronectin in combination with the enhanced stiffness in fibrotic areas will strongly impact on the cardiomyocyte behaviour and influence disease progression. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- William Hawkes
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
- Department of Chemistry, Queen Mary University of London, London E1 4NS, UK
| | - Emilie Marhuenda
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Paul Reynolds
- School of Engineering, University of Glasgow, Glasgow G12 8QQ, UK
| | - Caoimhe O'Neill
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | - Pragati Pandey
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| | | | - Mark Freeley
- Department of Chemistry, Queen Mary University of London, London E1 4NS, UK
| | - Da Huang
- Department of Chemistry, Queen Mary University of London, London E1 4NS, UK
| | - Junquiang Hu
- Department of Mechanical Engineering, Columbia University, New York, NY 10027, USA
| | - Sasha Gondarenko
- Department of Mechanical Engineering, Columbia University, New York, NY 10027, USA
| | - James Hone
- Department of Mechanical Engineering, Columbia University, New York, NY 10027, USA
| | | | - Matteo Palma
- Department of Chemistry, Queen Mary University of London, London E1 4NS, UK
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK
| |
Collapse
|
24
|
Cardiac fibroblasts and mechanosensation in heart development, health and disease. Nat Rev Cardiol 2022; 20:309-324. [PMID: 36376437 DOI: 10.1038/s41569-022-00799-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/04/2022] [Indexed: 11/16/2022]
Abstract
The term 'mechanosensation' describes the capacity of cells to translate mechanical stimuli into the coordinated regulation of intracellular signals, cellular function, gene expression and epigenetic programming. This capacity is related not only to the sensitivity of the cells to tissue motion, but also to the decryption of tissue geometric arrangement and mechanical properties. The cardiac stroma, composed of fibroblasts, has been historically considered a mechanically passive component of the heart. However, the latest research suggests that the mechanical functions of these cells are an active and necessary component of the developmental biology programme of the heart that is involved in myocardial growth and homeostasis, and a crucial determinant of cardiac repair and disease. In this Review, we discuss the general concept of cell mechanosensation and force generation as potent regulators in heart development and pathology, and describe the integration of mechanical and biohumoral pathways predisposing the heart to fibrosis and failure. Next, we address the use of 3D culture systems to integrate tissue mechanics to mimic cardiac remodelling. Finally, we highlight the potential of mechanotherapeutic strategies, including pharmacological treatment and device-mediated left ventricular unloading, to reverse remodelling in the failing heart.
Collapse
|
25
|
Zuela-Sopilniak N, Lammerding J. Can't handle the stress? Mechanobiology and disease. Trends Mol Med 2022; 28:710-725. [PMID: 35717527 PMCID: PMC9420767 DOI: 10.1016/j.molmed.2022.05.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/22/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
Mechanobiology is a rapidly growing research area focused on how mechanical forces and properties influence biological systems at the cell, molecular, and tissue level, and how those biological systems, in turn, control mechanical parameters. Recently, it has become apparent that disrupted mechanobiology has a significant role in many diseases, from cardiovascular disease to muscular dystrophy and cancer. An improved understanding of this intricate process could be harnessed toward developing alternative and more targeted treatment strategies, and to advance the fields of regenerative and personalized medicine. Modulating the mechanical properties of the cellular microenvironment has already been used successfully to boost antitumor immune responses and to induce cardiac and spinal regeneration, providing inspiration for further research in this area.
Collapse
Affiliation(s)
- Noam Zuela-Sopilniak
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
26
|
Montero-Calle P, Flandes-Iparraguirre M, Mountris K, S de la Nava A, Laita N, Rosales RM, Iglesias-García O, De-Juan-Pardo EM, Atienza F, Fernández-Santos ME, Peña E, Doblaré M, Gavira JJ, Fernández-Avilés F, Prosper F, Pueyo E, Mazo Vega MM. Fabrication of human myocardium using multidimensional modelling of engineered tissues. Biofabrication 2022; 14. [PMID: 36007502 DOI: 10.1088/1758-5090/ac8cb3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/25/2022] [Indexed: 11/12/2022]
Abstract
Biofabrication of human tissues has seen a meteoric growth triggered by recent technical advancements such as human induced pluripotent stem cells (hiPSCs) and additive manufacturing. However, generation of cardiac tissue is still hampered by lack of addequate mechanical properties and crucially by the often unpredictable post-fabrication evolution of biological components. In this study we employ melt electrowriting (MEW) and hiPSC-derived cardiac cells to generate fibre-reinforced human cardiac minitissues. These are thoroughly characterized in order to build computational models and simulations able to predict their post-fabrication evolution. Our results show that MEW-based human minitissues display advanced maturation 28 post-generation, with a significant increase in the expression of cardiac genes such as MYL2, GJA5, SCN5A and the MYH7/MYH6 and MYL2/MYL7 ratios. Human iPSC-cardiomyocytes are significantly more aligned within the MEW-based 3D tissues, as compared to conventional 2D controls, and also display greater expression of CX43. These are also correlated with a more mature functionality in the form of faster conduction velocity. We used these data to develop simulations capable of accurately reproducing the experimental performance. In-depth gauging of the structural disposition (cellular alignment) and intercellular connectivity (CX43) allowed us to develop an improved computational model able to predict the relationship between cardiac cell alignment and functional performance. This study lays down the path for advancing in the development of in silico tools to predict cardiac biofabricated tissue evolution after generation, and maps the route towards more accurate and biomimetic tissue manufacture.
Collapse
Affiliation(s)
| | | | - Konstantinos Mountris
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018 , SPAIN
| | - Ana S de la Nava
- Hospital General Universitario Gregorio Marañón, 46, Dr. Esquerdo, Madrid, Madrid, 28007, SPAIN
| | - Nicolás Laita
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | - Ricardo M Rosales
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | | | - Elena M De-Juan-Pardo
- Mechanical Engineering, University of Western Australia Faculty of Engineering Computing and Mathematics, M050, B.Block, 1.36, 35 Stirling Highway, Perth, Perth, Western Australia, 6009, AUSTRALIA
| | - Felipe Atienza
- Hospital General Universitario Gregorio Marañón, 46, Dr. Esquerdo st, Madrid, Madrid, 28007, SPAIN
| | | | - Estefanía Peña
- Aragón Institute for Engineering Research, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | - Manuel Doblaré
- Instituto de Investigación en Ingeniería de Aragón, Mariano Esquillor Gómez, Zaragoza, 50018, SPAIN
| | - Juan J Gavira
- Department of Cardiology, Clínica Universidad de Navarra, Pio XII av, Pamplona, 31008, SPAIN
| | | | - Felipe Prosper
- Hematology, Universidad de Navarra, Pio XII, 36, Pamplona, Navarra, 31008, SPAIN
| | - Esther Pueyo
- Instituto de Investigación en Ingeniería de Aragón, Calle Mariano Esquillor s/n, Zaragoza, 50018, SPAIN
| | | |
Collapse
|
27
|
Chen Y, Chan JPY, Wu J, Li R, Santerre JP. Compatibility and function of human induced pluripotent stem cell derived cardiomyocytes on an electrospun nanofibrous scaffold, generated from an ionomeric polyurethane composite. J Biomed Mater Res A 2022; 110:1932-1943. [PMID: 35851742 DOI: 10.1002/jbm.a.37428] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 11/06/2022]
Abstract
Synthetic scaffolds are needed for generating organized neo-myocardium constructs to promote functional tissue repair. This study investigated the biocompatibility of an elastomeric electrospun degradable polar/hydrophobic/ionic polyurethane (D-PHI) composite scaffold with human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The composite material was electrospun to generate scaffolds, with nanofibres oriented in aligned or random directions. These features enabled the authors to evaluate the effect of characteristic elements which mimic that of the native extracellular matrix (alignment, chemical heterogeneity, and fiber topography) on hiPSC-CMs activity. The functional nature of the hiPSC-CM cultured on gelatin and Matrigel-coated scaffolds were assessed, investigating the influence of protein interactions with the synthetic substrate on subsequent cell phenotype. After 7 days of culture, high hiPSC-CM viability was observed on the scaffolds. The cells on the aligned scaffold were elongated and demonstrated aligned sarcomeres that oriented parallel to the direction of the fibers, while the cells on random scaffolds and a tissue culture polystyrene (TCPS) control did not exhibit such an organized morphology. The hiPSC-CMs cultured on the scaffolds and TCPS expressed similar levels of cardiac troponin-T, but there was a higher expression of ventricular myosin light chain-2 on the D-PHI composite scaffolds versus TCPS, indicating a higher proportion of hiPSC-CM exhibiting a ventricular cardiomyocyte like phenotype. Within 7 days, the hiPSC-CMs on aligned scaffolds and TCPS beat synchronously and had similar conductive velocities. These preliminary results show that aligned D-PHI elastomeric scaffolds allow hiPSC-CMs to demonstrate important cardiomyocytes characteristics, critical to enabling their future potential use for cardiac tissue regeneration.
Collapse
Affiliation(s)
- Yizhou Chen
- Institute of Biomedical Engineering, University of Toronto Toronto Ontario Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research University of Toronto Toronto Ontario Canada
| | - Jennifer P. Y. Chan
- Institute of Biomedical Engineering, University of Toronto Toronto Ontario Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research University of Toronto Toronto Ontario Canada
- Baylis Medical Mississauga Ontario Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network Toronto Ontario Canada
| | - Ren‐Ke Li
- Toronto General Hospital Research Institute, University Health Network Toronto Ontario Canada
| | - J. Paul Santerre
- Institute of Biomedical Engineering, University of Toronto Toronto Ontario Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research University of Toronto Toronto Ontario Canada
- Toronto General Hospital Research Institute, University Health Network Toronto Ontario Canada
- Faculty of Dentistry University of Toronto Toronto Ontario Canada
| |
Collapse
|
28
|
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
|
29
|
Swiatlowska P, Sit B, Feng Z, Marhuenda E, Xanthis I, Zingaro S, Ward M, Zhou X, Xiao Q, Shanahan C, Jones GE, Yu CH, Iskratsch T. Pressure and stiffness sensing together regulate vascular smooth muscle cell phenotype switching. SCIENCE ADVANCES 2022; 8:eabm3471. [PMID: 35427166 PMCID: PMC9012473 DOI: 10.1126/sciadv.abm3471] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 03/01/2022] [Indexed: 06/14/2023]
Abstract
Vascular smooth muscle cells (VSMCs) play a central role in the progression of atherosclerosis, where they switch from a contractile to a synthetic phenotype. Because of their role as risk factors for atherosclerosis, we sought here to systematically study the impact of matrix stiffness and (hemodynamic) pressure on VSMCs. Thereby, we find that pressure and stiffness individually affect the VSMC phenotype. However, only the combination of hypertensive pressure and matrix compliance, and as such mechanical stimuli that are prevalent during atherosclerosis, leads to a full phenotypic switch including the formation of matrix-degrading podosomes. We further analyze the molecular mechanism in stiffness and pressure sensing and identify a regulation through different but overlapping pathways culminating in the regulation of the actin cytoskeleton through cofilin. Together, our data show how different pathological mechanical signals combined but through distinct pathways accelerate a phenotypic switch that will ultimately contribute to atherosclerotic disease progression.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Brian Sit
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
- School of Biomedical Sciences, Hong Kong University, Hong Kong, Hong Kong
| | - Zhen Feng
- School of Biomedical Sciences, Hong Kong University, Hong Kong, Hong Kong
| | - Emilie Marhuenda
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Ioannis Xanthis
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Simona Zingaro
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
| | - Matthew Ward
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Xinmiao Zhou
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Qingzhong Xiao
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Cathy Shanahan
- School of Cardiovascular Medicine and Sciences, King’s College London, London, UK
| | - Gareth E. Jones
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
| | - Cheng-han Yu
- School of Biomedical Sciences, Hong Kong University, Hong Kong, Hong Kong
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
- Randall Centre for Cell and Molecular Biophysics, King’s College London, London, UK
| |
Collapse
|
30
|
Shi H, Wu X, Sun S, Wang C, Vangelatos Z, Ash-Shakoor A, Grigoropoulos CP, Mather PT, Henderson JH, Ma Z. Profiling the responsiveness of focal adhesions of human cardiomyocytes to extracellular dynamic nano-topography. Bioact Mater 2022; 10:367-377. [PMID: 34901553 PMCID: PMC8636819 DOI: 10.1016/j.bioactmat.2021.08.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/17/2021] [Accepted: 08/25/2021] [Indexed: 01/02/2023] Open
Abstract
Focal adhesion complexes function as the mediators of cell-extracellular matrix interactions to sense and transmit the extracellular signals. Previous studies have demonstrated that cardiomyocyte focal adhesions can be modulated by surface topographic features. However, the response of focal adhesions to dynamic surface topographic changes remains underexplored. To study this dynamic responsiveness of focal adhesions, we utilized a shape memory polymer-based substrate that can produce a flat-to-wrinkle surface transition triggered by an increase of temperature. Using this dynamic culture system, we analyzed three proteins (paxillin, vinculin and zyxin) from different layers of the focal adhesion complex in response to dynamic extracellular topographic change. Hence, we quantified the dynamic profile of cardiomyocyte focal adhesion in a time-dependent manner, which provides new understanding of dynamic cardiac mechanobiology.
Collapse
Affiliation(s)
- Huaiyu Shi
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Xiangjun Wu
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Shiyang Sun
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Chenyan Wang
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Zacharias Vangelatos
- Department of Mechanical Engineering, University of California, Berkeley, PA, 94720, USA
| | - Ariel Ash-Shakoor
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
| | | | - Patrick T. Mather
- Department of Chemical Engineering, Bucknell University, Lewisburg, PA, 17837, USA
| | - James H. Henderson
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Syracuse Institute for Materials and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| |
Collapse
|
31
|
Neel BL, Nisler CR, Walujkar S, Araya-Secchi R, Sotomayor M. Elastic versus brittle mechanical responses predicted for dimeric cadherin complexes. Biophys J 2022; 121:1013-1028. [PMID: 35151631 PMCID: PMC8943749 DOI: 10.1016/j.bpj.2022.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/02/2022] [Accepted: 02/07/2022] [Indexed: 12/15/2022] Open
Abstract
Cadherins are a superfamily of adhesion proteins involved in a variety of biological processes that include the formation of intercellular contacts, the maintenance of tissue integrity, and the development of neuronal circuits. These transmembrane proteins are characterized by ectodomains composed of a variable number of extracellular cadherin (EC) repeats that are similar but not identical in sequence and fold. E-cadherin, along with desmoglein and desmocollin proteins, are three classical-type cadherins that have slightly curved ectodomains and engage in homophilic and heterophilic interactions through an exchange of conserved tryptophan residues in their N-terminal EC1 repeat. In contrast, clustered protocadherins are straighter than classical cadherins and interact through an antiparallel homophilic binding interface that involves overlapped EC1 to EC4 repeats. Here we present molecular dynamics simulations that model the adhesive domains of these cadherins using available crystal structures, with systems encompassing up to 2.8 million atoms. Simulations of complete classical cadherin ectodomain dimers predict a two-phased elastic response to force in which these complexes first softly unbend and then stiffen to unbind without unfolding. Simulated α, β, and γ clustered protocadherin homodimers lack a two-phased elastic response, are brittle and stiffer than classical cadherins and exhibit complex unbinding pathways that in some cases involve transient intermediates. We propose that these distinct mechanical responses are important for function, with classical cadherin ectodomains acting as molecular shock absorbers and with stiffer clustered protocadherin ectodomains facilitating overlap that favors binding specificity over mechanical resilience. Overall, our simulations provide insights into the molecular mechanics of single cadherin dimers relevant in the formation of cellular junctions essential for tissue function.
Collapse
Affiliation(s)
- Brandon L Neel
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio
| | - Collin R Nisler
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; Biophysics Graduate Program, The Ohio State University, Columbus, Ohio
| | - Sanket Walujkar
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; Chemical Physics Graduate Program, The Ohio State University, Columbus, Ohio
| | - Raul Araya-Secchi
- Facultad de Ingeniería y Tecnología, Universidad San Sebastián, Santiago, Chile
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, Ohio; The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio; Biophysics Graduate Program, The Ohio State University, Columbus, Ohio; Chemical Physics Graduate Program, The Ohio State University, Columbus, Ohio.
| |
Collapse
|
32
|
Mechanobiology of muscle and myofibril morphogenesis. Cells Dev 2021; 168:203760. [PMID: 34863916 DOI: 10.1016/j.cdev.2021.203760] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/05/2023]
Abstract
Muscles generate forces for animal locomotion. The contractile apparatus of muscles is the sarcomere, a highly regular array of large actin and myosin filaments linked by gigantic titin springs. During muscle development many sarcomeres assemble in series into long periodic myofibrils that mechanically connect the attached skeleton elements. Thus, ATP-driven myosin forces can power movement of the skeleton. Here we review muscle and myofibril morphogenesis, with a particular focus on their mechanobiology. We describe recent progress on the molecular structure of sarcomeres and their mechanical connections to the skeleton. We discuss current models predicting how tension coordinates the assembly of key sarcomeric components to periodic myofibrils that then further mature during development. This requires transcriptional feedback mechanisms that may help to coordinate myofibril assembly and maturation states with the transcriptional program. To fuel the varying energy demands of muscles we also discuss the close mechanical interactions of myofibrils with mitochondria and nuclei to optimally support powerful or enduring muscle fibers.
Collapse
|
33
|
Peng L, Gautrot JE. Long term expansion profile of mesenchymal stromal cells at protein nanosheet-stabilised bioemulsions for next generation cell culture microcarriers. Mater Today Bio 2021; 12:100159. [PMID: 34841241 PMCID: PMC8605361 DOI: 10.1016/j.mtbio.2021.100159] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/10/2021] [Accepted: 11/13/2021] [Indexed: 12/27/2022] Open
Abstract
Tremendous progress in the identification, isolation and expansion of stem cells has allowed their application in regenerative medicine and tissue engineering, and their use as advanced in vitro models. As a result, stem cell manufacturing increasingly requires scale up, parallelisation and automation. However, solid substrates currently used for the culture of adherent cells are poorly adapted for such applications, owing to their difficult processing from cell products, relatively high costs and their typical reliance on difficult to recycle plastics and microplastics. In this work, we show that bioemulsions formed of microdroplets stabilised by protein nanosheets displaying strong interfacial mechanics are well-suited for the scale up of adherent stem cells such as mesenchymal stromal cells (MSCs). We demonstrate that, over multiple passages (up to passage 10), MSCs retain comparable phenotypes when cultured on such bioemulsions, solid microcarriers (Synthemax II) and classic 2D tissue culture polystyrene. Phenotyping (cell proliferation, morphometry, flow cytometry and differentiation assays) of MSCs cultured for multiple passages on these systems indicate that, although stemness is lost at late passages when cultured on these different substrates, stem cell phenotypes remained comparable between different culture conditions, at any given passage. Hence our study validates the use of bioemulsions for the long term expansion of adherent stem cells and paves the way to the design of novel 3D bioreactors based on microdroplet microcarriers.
Collapse
Affiliation(s)
- Lihui Peng
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| | - Julien E Gautrot
- Institute of Bioengineering and, UK.,School of Engineering and Materials Science, Queen Mary, University of London, Mile End Road, London, E1 4NS, UK
| |
Collapse
|
34
|
Singh JP, Young JL. The cardiac nanoenvironment: form and function at the nanoscale. Biophys Rev 2021; 13:625-636. [PMID: 34765045 PMCID: PMC8555021 DOI: 10.1007/s12551-021-00834-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Mechanical forces in the cardiovascular system occur over a wide range of length scales. At the whole organ level, large scale forces drive the beating heart as a synergistic unit. On the microscale, individual cells and their surrounding extracellular matrix (ECM) exhibit dynamic reciprocity, with mechanical feedback moving bidirectionally. Finally, in the nanometer regime, molecular features of cells and the ECM show remarkable sensitivity to mechanical cues. While small, these nanoscale properties are in many cases directly responsible for the mechanosensitive signaling processes that elicit cellular outcomes. Given the inherent challenges in observing, quantifying, and reconstituting this nanoscale environment, it is not surprising that this landscape has been understudied compared to larger length scales. Here, we aim to shine light upon the cardiac nanoenvironment, which plays a crucial role in maintaining physiological homeostasis while also underlying pathological processes. Thus, we will highlight strategies aimed at (1) elucidating the nanoscale components of the cardiac matrix, and (2) designing new materials and biosystems capable of mimicking these features in vitro.
Collapse
Affiliation(s)
- Jashan P Singh
- Mechanobiology Institute, National University of Singapore, 117411 Singapore, Singapore
| | - Jennifer L Young
- Mechanobiology Institute, National University of Singapore, 117411 Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, 117575 Singapore, Singapore
| |
Collapse
|
35
|
Abstract
The contraction-relaxation cycle of the heart is one of the most robust mechanical systems in the body that adapts rapidly to the body's needs by changing mechanical parameters. In many respects, we can consider the cardiac system as a complex machine and can use engineering approaches to describe its function. The classical physiology of the heart also focused on understanding function but the new molecular level tools in light microscopy and nanoengineering now enable a deeper understanding of the physiology. The field of mechanobiology has emerged with a focus on how mechanical activity alters biological systems at the molecular level and how those systems in turn control mechanical parameters. In the case of mechanical activity, there are clearly benefits of exercise for the heart, for cancer patients, and for aging but we do not understand the links at a molecular level. Why does regular exercise benefit the heart? We have some preliminary clues at a molecular level about the benefits of physical activity in the cases of cancer and aging; however, there is less known about how exercise affects cardiovascular performance. Unlike the omics approaches which generally link proteins to processes, a mechanobiological understanding of a process explains how forces and mechanical activity will regulate the process through modifications of protein activities. In other words, mechanical activity is an essential component of most biological systems that is transduced into biochemical changes in protein activity. Further, it follows logically that if a mechanical parameter of the cardiac system is typically controlled, then cellular mechanosensing systems must be able to directly or indirectly measure that parameter. The challenge is to understand how changes in activity of the heart are controlled in the short term and then how the system adapts to the integrated level of activity over the longer term. By way of introduction to molecular mechanobiology, I will present examples of mechanosensing from the molecular to the cellular scale and how they may be integrated at the cell and tissue levels. An important element of Mechanobiology at the system level is the physiological state of the cell: i.e., the cell in a senescent state, a cancer state, or a normal cell state (Sheetz 2019). The background for the mechanobiological approach is discussed in "The Cell as a Machine" (Sheetz and Yu, Cambridge Univ Press, 2018), which considers cell states and the molecular systems underlying the important cellular functions. A major challenge in mechanobiology is the understanding of the transduction of mechanical activity into changes in cell function. Of particular relevance here is the benefit of exercise to cardiac performance. This has been seen in many cases and there are a variety of factors that contribute. Further, exercise will benefit cancer patients and will reverse some of the adverse effects of aging. Exercise will cause increased cardiac activity that will be sensed by many mechanosensory systems from a molecular to a cellular level both in the heart and in the vasculature. At a molecular level in cardiac systems, proteins are able to measure stress and strain and to generate appropriate signals of the magnitude of stress and strain that can regulate the cellular contractility and other parameters. The protein sensors are generally passive systems that give a transient measure of local parameters such as the stress at cell-cell junctions during contraction and the strain of the sarcomeres during relaxation. Large stresses at the junctions can activate signaling systems that can reduce contractility or over time activate remodeling of the junctions to better support larger stresses. The proteins involved and their sensory mechanisms are not known currently; however, the mechanosensitive channel, Piezo1, has been implicated in the transduction process in the vasculature (Beech 2018). In the case of strain sensors, large stretches of titin during relaxation can unfold more titin domains that can send signals to the cell. Two different mechanisms of strain sensing are likely in titin. The titin kinase domain is activated by strain but the substrates of the kinase are not know in vivo (Linke 2018). In the backbone of titin are many Ig domains that unfold at different forces and unfolding could cause the binding of proteins that would then activate enzymatic pathways to alter the contractile cycle to give the proper level of strain (Ait-Mou et al. 2017; Granzier et al. 2014; Granzier et al. 2009). The cell-matrix adhesion protein, talin, has eleven cryptic binding sites for another adhesion protein, vinculin, that are revealed by the unfolding of domains in the talin molecule (Yao et al. 2016). Since some domains unfold at lower forces than others, small strains will preferentially unfold those domains, making the system an excellent sensor of the extent of stretch as expected for titin. Because there is an ordered array of many titin molecules, the sensing of strain can be very sensitive to small changes in sarcomere length. Needless to say, titin is only one part of the regulatory system that controls sarcomere length. As one goes more deeply into the working of the system, it is evident that many additional mechanosensory elements are involved in maintaining a functioning cardiac system.
Collapse
|
36
|
Huethorst E, Mortensen P, Simitev RD, Gao H, Pohjolainen L, Talman V, Ruskoaho H, Burton FL, Gadegaard N, Smith GL. Conventional rigid 2D substrates cause complex contractile signals in monolayers of human induced pluripotent stem cell-derived cardiomyocytes. J Physiol 2021; 600:483-507. [PMID: 34761809 PMCID: PMC9299844 DOI: 10.1113/jp282228] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/03/2021] [Indexed: 11/21/2022] Open
Abstract
Abstract Human induced pluripotent stem cell‐derived cardiomyocytes (hiPSC‐CM) in monolayers interact mechanically via cell–cell and cell–substrate adhesion. Spatiotemporal features of contraction were analysed in hiPSC‐CM monolayers (1) attached to glass or plastic (Young's modulus (E) >1 GPa), (2) detached (substrate‐free) and (3) attached to a flexible collagen hydrogel (E = 22 kPa). The effects of isoprenaline on contraction were compared between rigid and flexible substrates. To clarify the underlying mechanisms, further gene expression and computational studies were performed. HiPSC‐CM monolayers exhibited multiphasic contractile profiles on rigid surfaces in contrast to hydrogels, substrate‐free cultures or single cells where only simple twitch‐like time‐courses were observed. Isoprenaline did not change the contraction profile on either surface, but its lusitropic and chronotropic effects were greater in hydrogel compared with glass. There was no significant difference between stiff and flexible substrates in regard to expression of the stress‐activated genes NPPA and NPPB. A computational model of cell clusters demonstrated similar complex contractile interactions on stiff substrates as a consequence of cell‐to‐cell functional heterogeneity. Rigid biomaterial surfaces give rise to unphysiological, multiphasic contractions in hiPSC‐CM monolayers. Flexible substrates are necessary for normal twitch‐like contractility kinetics and interpretation of inotropic interventions.
![]() Key points Spatiotemporal contractility analysis of human induced pluripotent stem cell‐derived cardiomyocyte (hiPSC‐CM) monolayers seeded on conventional, rigid surfaces (glass or plastic) revealed the presence of multiphasic contraction patterns across the monolayer with a high variability, despite action potentials recorded in the same areas being identical. These multiphasic patterns are not present in single cells, in detached monolayers or in monolayers seeded on soft substrates such as a hydrogel, where only ‘twitch’‐like transients are observed. HiPSC‐CM monolayers that display a high percentage of regions with multiphasic contraction have significantly increased contractile duration and a decreased lusotropic drug response. There is no indication that the multiphasic contraction patterns are associated with significant activation of the stress‐activated NPPA or NPPB signalling pathways. A computational model of cell clusters supports the biological findings that the rigid surface and the differential cell–substrate adhesion underly multiphasic contractile behaviour of hiPSC‐CMs.
Collapse
Affiliation(s)
- Eline Huethorst
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.,Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - Peter Mortensen
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| | - Radostin D Simitev
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| | - Hao Gao
- School of Mathematics and Statistics, University of Glasgow, Glasgow, UK
| | - Lotta Pohjolainen
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Virpi Talman
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heikki Ruskoaho
- Drug Research Program and Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Francis L Burton
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - Godfrey L Smith
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
37
|
Chin IL, Hool L, Choi YS. Interrogating cardiac muscle cell mechanobiology on stiffness gradient hydrogels. Biomater Sci 2021; 9:6795-6806. [PMID: 34542112 DOI: 10.1039/d1bm01061a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Extracellular matrix (ECM) remodeling is a major facet of cardiac development and disease, yet our understanding of cardiomyocyte mechanotransduction remains limited. To enhance our understanding of cardiomyocyte mechanosensation, we studied stiffness-driven changes to cell morphology and mechanomarker expression in H9C2 cells and neonatal rat cardiomyocytes (NRCMs). Linear stiffness gradient polyacrylamide hydrogels (2-33 kPa) coated with ECM proteins including Collagen I (Col), Fibronectin (Fn) or Laminin (Ln) were used to represent necrotic, healthy, and infarcted cardiac tissue on a continuous stiffness gradient. Cell size, cell shape and nuclear size were found to be mechanosensitive in H9C2 cells, as was the expression or nuclear translocalization of the mechanomarkers Lamin-A, YAP, and MRTF-A. Minor differences were observed between the different ECM coatings, with the same overarching stiffness-dependent trends being observed across Col, Fn and Ln coated hydrogels. Inhibition of mechanotransduction in H9C2 cells using blebbistatin or Y27632 resulted in disruptions to cell shape, nuclear shape, and nuclear size, however, trends in cell size and mechanomarker expression were not significantly attenuated. Mechanosensation in NRCMs was much less marked, with no significant changes in cell morphology being detected, although YAP did become increasingly nuclear localized with increasing stiffness. In α-actinin positive cells, striations formed with regular structure and frequency at all stiffnesses for Col and Fn coated hydrogels, but not Ln coated gels. In this study, we used our stiffness gradient hydrogels to comprehensively map the relationship between ECM stiffness and cardiac cell phenotype and found that less mature H9C2 cardiac cells are more sensitive to ECM changes than the more developed neonatal cardiomyocytes.
Collapse
Affiliation(s)
- Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| | - Livia Hool
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia. .,Victor Chang Cardiac Research Institute, Sydney, NSW, Australia
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
38
|
Swiatlowska P, Iskratsch T. Tools for studying and modulating (cardiac muscle) cell mechanics and mechanosensing across the scales. Biophys Rev 2021; 13:611-623. [PMID: 34765044 PMCID: PMC8553672 DOI: 10.1007/s12551-021-00837-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/24/2021] [Indexed: 12/26/2022] Open
Abstract
Cardiomyocytes generate force for the contraction of the heart to pump blood into the lungs and body. At the same time, they are exquisitely tuned to the mechanical environment and react to e.g. changes in cell and extracellular matrix stiffness or altered stretching due to reduced ejection fraction in heart disease, by adapting their cytoskeleton, force generation and cell mechanics. Both mechanical sensing and cell mechanical adaptations are multiscale processes. Receptor interactions with the extracellular matrix at the nanoscale will lead to clustering of receptors and modification of the cytoskeleton. This in turn alters mechanosensing, force generation, cell and nuclear stiffness and viscoelasticity at the microscale. Further, this affects cell shape, orientation, maturation and tissue integration at the microscale to macroscale. A variety of tools have been developed and adapted to measure cardiomyocyte receptor-ligand interactions and forces or mechanics at the different ranges, resulting in a wealth of new information about cardiomyocyte mechanobiology. Here, we take stock at the different tools for exploring cardiomyocyte mechanosensing and cell mechanics at the different scales from the nanoscale to microscale and macroscale.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| |
Collapse
|
39
|
Solís C, Russell B. Striated muscle proteins are regulated both by mechanical deformation and by chemical post-translational modification. Biophys Rev 2021; 13:679-695. [PMID: 34777614 PMCID: PMC8555064 DOI: 10.1007/s12551-021-00835-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 01/09/2023] Open
Abstract
All cells sense force and build their cytoskeleton to optimize function. How is this achieved? Two major systems are involved. The first is that load deforms specific protein structures in a proportional and orientation-dependent manner. The second is post-translational modification of proteins as a consequence of signaling pathway activation. These two processes work together in a complex way so that local subcellular assembly as well as overall cell function are controlled. This review discusses many cell types but focuses on striated muscle. Detailed information is provided on how load deforms the structure of proteins in the focal adhesions and filaments, using α-actinin, vinculin, talin, focal adhesion kinase, LIM domain-containing proteins, filamin, myosin, titin, and telethonin as examples. Second messenger signals arising from external triggers are distributed throughout the cell causing post-translational or chemical modifications of protein structures, with the actin capping protein CapZ and troponin as examples. There are numerous unanswered questions of how mechanical and chemical signals are integrated by muscle proteins to regulate sarcomere structure and function yet to be studied. Therefore, more research is needed to see how external triggers are integrated with local tension generated within the cell. Nonetheless, maintenance of tension in the sarcomere is the essential and dominant mechanism, leading to the well-known phrase in exercise physiology: "use it or lose it."
Collapse
Affiliation(s)
- Christopher Solís
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Brenda Russell
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
40
|
Mulvany E, McMahan S, Xu J, Yazdani N, Willits R, Liao J, Zhang G, Hong Y. In vitro comparison of harvesting site effects on cardiac extracellular matrix hydrogels. J Biomed Mater Res A 2021; 109:1922-1930. [PMID: 33822464 PMCID: PMC9789793 DOI: 10.1002/jbm.a.37184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 03/04/2021] [Accepted: 03/24/2021] [Indexed: 12/26/2022]
Abstract
Cardiac extracellular matrix (cECM) derived hydrogel has been investigated to treat myocardial infarction through animal studies and clinical trials. The tissue harvesting site commonly selects porcine left ventricle (LV) because heart attack majorly takes place in LV. However, little is known about whether the region of cardiac tissue harvesting is critical for downstream applications. In this work, in vitro studies to compare cECM hydrogels derived from adult porcine whole heart (WH), LV, and right ventricle (RV) were performed. The cECM from WH has similar chemical composition compared with cECM from LV and RV. All three types of cECM hydrogels share many similarities in terms of their microstructure, gelation time, and mechanical properties. WH-derived cECM hydrogels have larger variations in storage modulus (G') and complex modulus (G*) compared with the other two types of cECM hydrogels. Both human cardiomyocytes and mesenchymal stem cells could maintain high cell viability on all hydrogels without significant difference. In terms of above results, the cECM hydrogels from WH, LV and RV exhibited similarity in material properties and cell response in vitro. Thus, future fabrication of cECM hydrogels from WH would increase the yield, which would decrease processing time and production cost.
Collapse
Affiliation(s)
- Emily Mulvany
- Department of Biomedical Engineering, The University of Akron, Ohio, OH 44325
| | - Sara McMahan
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| | - Jiazhu Xu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| | - Narges Yazdani
- Department of Biomedical Engineering, The University of Akron, Ohio, OH 44325
| | - Rebecca Willits
- Department of Biomedical Engineering, The University of Akron, Ohio, OH 44325
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Ohio, OH 44325,Corresponding authors: Yi Hong, , Phone: 01-817-272-0562; Ge Zhang, , phone: 01-330-972-5237
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76019,Corresponding authors: Yi Hong, , Phone: 01-817-272-0562; Ge Zhang, , phone: 01-330-972-5237
| |
Collapse
|
41
|
Miyashita Y, Tsukamoto O, Matsuoka K, Kamikubo K, Kuramoto Y, Ying Fu H, Tsubota T, Hasuike H, Takayama T, Ito H, Hitsumoto T, Okamoto C, Kioka H, Oya R, Shinomiya H, Hakui H, Shintani Y, Kato H, Kitakaze M, Sakata Y, Asano Y, Takashima S. The CR9 element is a novel mechanical load-responsive enhancer that regulates natriuretic peptide genes expression. FASEB J 2021; 35:e21495. [PMID: 33689182 DOI: 10.1096/fj.202002111rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 11/11/2022]
Abstract
Enhancers regulate gene expressions in a tissue- and pathology-specific manner by altering its activities. Plasma levels of atrial and brain natriuretic peptides, encoded by the Nppa and Nppb, respectively, and synthesized predominantly in cardiomyocytes, vary depending on the severity of heart failure. We previously identified the noncoding conserved region 9 (CR9) element as a putative Nppb enhancer at 22-kb upstream from the Nppb gene. However, its regulatory mechanism remains unknown. Here, we therefore investigated the mechanism of CR9 activation in cardiomyocytes using different kinds of drugs that induce either cardiac hypertrophy or cardiac failure accompanied by natriuretic peptides upregulation. Chronic treatment of mice with either catecholamines or doxorubicin increased CR9 activity during the progression of cardiac hypertrophy to failure, which is accompanied by proportional increases in Nppb expression. Conversely, for cultured cardiomyocytes, doxorubicin decreased CR9 activity and Nppb expression, while catecholamines increased both. However, exposing cultured cardiomyocytes to mechanical loads, such as mechanical stretch or hydrostatic pressure, upregulate CR9 activity and Nppb expression even in the presence of doxorubicin. Furthermore, the enhancement of CR9 activity and Nppa and Nppb expressions by either catecholamines or mechanical loads can be blunted by suppressing mechanosensing and mechanotransduction pathways, such as muscle LIM protein (MLP) or myosin tension. Finally, the CR9 element showed a more robust and cell-specific response to mechanical loads than the -520-bp BNP promoter. We concluded that the CR9 element is a novel enhancer that responds to mechanical loads by upregulating natriuretic peptides expression in cardiomyocytes.
Collapse
Affiliation(s)
- Yohei Miyashita
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan.,Department of Legal Medicine, Graduate School of Medicine, Osaka University, Suita, Japan.,Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Ken Matsuoka
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Kenta Kamikubo
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Yuki Kuramoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hai Ying Fu
- Department of Cardiorenal and Cerebrovascular Medicine, Faculty of Medicine, Kagawa University, Takamatsu, Japan
| | - Tomoya Tsubota
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Hirona Hasuike
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Toshio Takayama
- School of Engineering, Department of Mechanical Engineering, Tokyo Institute of Technology, Tokyo, Japan
| | - Hiroaki Ito
- Department of Physics, Graduate School of Science, Chiba University, Chiba, Japan
| | - Tatsuro Hitsumoto
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Chisato Okamoto
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Hidetaka Kioka
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ryohei Oya
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | - Haruki Shinomiya
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hideyuki Hakui
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yasunori Shintani
- Department of Molecular Pharmacology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| | | | - Yasushi Sakata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Graduate School of Medicine/Frontier Biosciences, Osaka University, Suita, Japan
| |
Collapse
|
42
|
Ross JA, Stroud MJ. THE NUCLEUS: Mechanosensing in cardiac disease. Int J Biochem Cell Biol 2021; 137:106035. [PMID: 34242685 DOI: 10.1016/j.biocel.2021.106035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022]
Abstract
The nucleus provides a physical and selective chemical boundary to segregate the genome from the cytoplasm. The contents of the nucleus are surrounded by the nuclear envelope, which acts as a hub of mechanosensation, transducing forces from the external cytoskeleton to the nucleus, thus impacting on nuclear morphology, genome organisation, gene transcription and signalling pathways. Muscle tissues such as the heart are unique in that they actively generate large contractile forces, resulting in a distinctive mechanical environment which impacts nuclear properties, function and mechanosensing. In light of this, mutations that affect the function of the nuclear envelope (collectively known as nuclear envelopathies and laminopathies) disproportionately result in striated muscle diseases, which include dilated and arrhythmogenic cardiomyopathies. Here we review the nucleus and its role in mechanotransduction, as well as associated defects that lead to cardiac dysfunction.
Collapse
Affiliation(s)
- Jacob A Ross
- British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London, UK
| | - Matthew J Stroud
- British Heart Foundation Centre of Excellence, School of Cardiovascular Medicine and Sciences, King's College London, London, UK.
| |
Collapse
|
43
|
Latham SL, Weiß N, Schwanke K, Thiel C, Croucher DR, Zweigerdt R, Manstein DJ, Taft MH. Myosin-18B Regulates Higher-Order Organization of the Cardiac Sarcomere through Thin Filament Cross-Linking and Thick Filament Dynamics. Cell Rep 2021; 32:108090. [PMID: 32877672 DOI: 10.1016/j.celrep.2020.108090] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/07/2020] [Accepted: 08/07/2020] [Indexed: 12/20/2022] Open
Abstract
MYO18B loss-of-function mutations and depletion significantly compromise the structural integrity of striated muscle sarcomeres. The molecular function of the encoded protein, myosin-18B (M18B), within the developing muscle is unknown. Here, we demonstrate that recombinant M18B lacks motor ATPase activity and harbors previously uncharacterized N-terminal actin-binding domains, properties that make M18B an efficient actin cross-linker and molecular brake capable of regulating muscle myosin-2 contractile forces. Spatiotemporal analysis of M18B throughout cardiomyogenesis and myofibrillogenesis reveals that this structural myosin undergoes nuclear-cytoplasmic redistribution during myogenic differentiation, where its incorporation within muscle stress fibers coincides with actin striation onset. Furthermore, this analysis shows that M18B is directly integrated within the muscle myosin thick filament during myofibril maturation. Altogether, our data suggest that M18B has evolved specific biochemical properties that allow it to define and maintain sarcomeric organization from within the thick filament via its dual actin cross-linking and motor modulating capabilities.
Collapse
Affiliation(s)
- Sharissa L Latham
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany; The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Hospital Clinical School, UNSW Sydney, NSW 2052, Australia
| | - Nadine Weiß
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany
| | - Kristin Schwanke
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover 30625, Germany
| | - Claudia Thiel
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany
| | - David R Croucher
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia; St Vincent's Hospital Clinical School, UNSW Sydney, NSW 2052, Australia
| | - Robert Zweigerdt
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiac, Thoracic, Transplantation and Vascular Surgery, REBIRTH-Cluster of Excellence, Hannover Medical School, Hannover 30625, Germany
| | - Dietmar J Manstein
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany
| | - Manuel H Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover 30625, Germany.
| |
Collapse
|
44
|
Maturation of human pluripotent stem cell derived cardiomyocytes in vitro and in vivo. Semin Cell Dev Biol 2021; 118:163-171. [PMID: 34053865 DOI: 10.1016/j.semcdb.2021.05.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 01/15/2023]
Abstract
Human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) represent an inexhaustible cell source for in vitro disease modeling, drug discovery and toxicity screening, and potential therapeutic applications. However, currently available differentiation protocols yield populations of hPSC-CMs with an immature phenotype similar to cardiomyocytes in the early fetal heart. In this review, we consider the developmental processes and signaling cues involved in normal human cardiac maturation, as well as how these insights might be applied to the specific maturation of hPSC-CMs. We summarize the state-of-the-art and relative merits of reported hPSC-CM maturation strategies including prolonged duration in culture, metabolic manipulation, treatment with soluble or substrate-based cues, and tissue engineering approaches. Finally, we review the evidence that hPSC-CMs mature after implantation in injured hearts as such in vivo remodeling will likely affect the safety and efficacy of a potential hPSC-based cardiac therapy.
Collapse
|
45
|
Kit-Anan W, Mazo MM, Wang BX, Leonardo V, Pence IJ, Gopal S, Gelmi A, Nagelkerke A, Becce M, Chiappini C, Harding SE, Terracciano CM, Stevens MM. Multiplexing physical stimulation on single human induced pluripotent stem cell-derived cardiomyocytes for phenotype modulation. Biofabrication 2021; 13:025004. [PMID: 33710972 PMCID: PMC7610872 DOI: 10.1088/1758-5090/abce0a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/11/2020] [Accepted: 11/25/2020] [Indexed: 12/02/2022]
Abstract
Traditional in vitro bioengineering approaches whereby only individual biophysical cues are manipulated at any one time are highly inefficient, falling short when recapitulating the complexity of the cardiac environment. Multiple biophysical cues are present in the native myocardial niche and are essential during development, as well as in maintenance of adult cardiomyocyte (CM) phenotype in both health and disease. This study establishes a novel biofabrication workflow to study and manipulate hiPSC-CMs and to understand how these cells respond to a multiplexed biophysical environment, namely 3D shape and substrate stiffness, at a single cell level. Silicon masters were fabricated and developed to generate inverse patterns of the desired 3D shapes in bas relief, which then were used to mold the designed microwell arrays into a hydrogel. Polyacrylamide (PAAm) was modified with the incorporation of acrylic acid to provide a carboxylic group conjugation site for adhesion motifs, without compromising capacity to modulate stiffness. In this manner, two individual parameters can be finely tuned independently within the hydrogel: the shape of the 3D microwell and its stiffness. The design allows the platform to isolate single hiPSC-CMs to study solely biophysical cues in the absence of cell-cell physical interaction. Under physiologic-like physical conditions (3D shape resembling that of adult CM and 9.83 kPa substrate stiffness that mimics muscle stiffness), isolated single hiPSC-CMs exhibit increased Cx-43 density, cell membrane stiffness and calcium transient amplitude; co-expression of the subpopulation-related MYL2-MYL7 proteins; and higher anisotropism than cells in pathologic-like conditions (flat surface and 112 kPa substrate stiffness). This demonstrates that supplying a physiologic or pathologic microenvironment to an isolated single hiPSC-CM in the absence of any physical cell-to-cell communication in this biofabricated platform leads to a significantly different set of cellular features, thus presenting a differential phenotype. Importantly, this demonstrates the high plasticity of hiPSC-CMs even in isolation. The ability of multiple biophysical cues to significantly influence isolated single hiPSC-CM phenotype and functionality highlights the importance of fine-tuning such cues for specific applications. This has the potential to produce more fit-for-purpose hiPSC-CMs. Further understanding of human cardiac development is enabled by the robust, versatile and reproducible biofabrication techniques applied here. We envision that this system could be easily applied to other tissues and cell types where the influence of cellular shape and stiffness of the surrounding environment is hypothesized to play an important role in physiology.
Collapse
Affiliation(s)
- Worrapong Kit-Anan
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Manuel M Mazo
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Brian X Wang
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Vincent Leonardo
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Isaac J Pence
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Sahana Gopal
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Amy Gelmi
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
- Current Address: Applied Chemistry and Environmental Science, School of Science, RMIT University, Melbourne, VIC 3001, Australia
| | - Anika Nagelkerke
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Michele Becce
- Department of Materials, Imperial College London, London, United Kingdom
| | - Ciro Chiappini
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Sian E Harding
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Cesare M Terracciano
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Molly M Stevens
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
46
|
Münch J, Abdelilah-Seyfried S. Sensing and Responding of Cardiomyocytes to Changes of Tissue Stiffness in the Diseased Heart. Front Cell Dev Biol 2021; 9:642840. [PMID: 33718383 PMCID: PMC7952448 DOI: 10.3389/fcell.2021.642840] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocytes are permanently exposed to mechanical stimulation due to cardiac contractility. Passive myocardial stiffness is a crucial factor, which defines the physiological ventricular compliance and volume of diastolic filling with blood. Heart diseases often present with increased myocardial stiffness, for instance when fibrotic changes modify the composition of the cardiac extracellular matrix (ECM). Consequently, the ventricle loses its compliance, and the diastolic blood volume is reduced. Recent advances in the field of cardiac mechanobiology revealed that disease-related environmental stiffness changes cause severe alterations in cardiomyocyte cellular behavior and function. Here, we review the molecular mechanotransduction pathways that enable cardiomyocytes to sense stiffness changes and translate those into an altered gene expression. We will also summarize current knowledge about when myocardial stiffness increases in the diseased heart. Sophisticated in vitro studies revealed functional changes, when cardiomyocytes faced a stiffer matrix. Finally, we will highlight recent studies that described modulations of cardiac stiffness and thus myocardial performance in vivo. Mechanobiology research is just at the cusp of systematic investigations related to mechanical changes in the diseased heart but what is known already makes way for new therapeutic approaches in regenerative biology.
Collapse
Affiliation(s)
- Juliane Münch
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany.,Institute of Molecular Biology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
47
|
Thomas D, Shenoy S, Sayed N. Building Multi-Dimensional Induced Pluripotent Stem Cells-Based Model Platforms to Assess Cardiotoxicity in Cancer Therapies. Front Pharmacol 2021; 12:607364. [PMID: 33679396 PMCID: PMC7930625 DOI: 10.3389/fphar.2021.607364] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular disease (CVD) complications have contributed significantly toward poor survival of cancer patients worldwide. These complications that result in myocardial and vascular damage lead to long-term multisystemic disorders. In some patient cohorts, the progression from acute to symptomatic CVD state may be accelerated due to exacerbation of underlying comorbidities such as obesity, diabetes and hypertension. In such situations, cardio-oncologists are often left with a clinical predicament in finding the optimal therapeutic balance to minimize cardiovascular risks and maximize the benefits in treating cancer. Hence, prognostically there is an urgent need for cost-effective, rapid, sensitive and patient-specific screening platform to allow risk-adapted decision making to prevent cancer therapy related cardiotoxicity. In recent years, momentous progress has been made toward the successful derivation of human cardiovascular cells from induced pluripotent stem cells (iPSCs). This technology has not only provided deeper mechanistic insights into basic cardiovascular biology but has also seamlessly integrated within the drug screening and discovery programs for early efficacy and safety evaluation. In this review, we discuss how iPSC-derived cardiovascular cells have been utilized for testing oncotherapeutics to pre-determine patient predisposition to cardiovascular toxicity. Lastly, we highlight the convergence of tissue engineering technologies and precision medicine that can enable patient-specific cardiotoxicity prognosis and treatment on a multi-organ level.
Collapse
Affiliation(s)
- Dilip Thomas
- Stanford Cardiovascular Institute, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, United States
| | - Sushma Shenoy
- Stanford Cardiovascular Institute, Stanford, CA, United States
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, United States.,Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
48
|
Daliri K, Pfannkuche K, Garipcan B. Effects of physicochemical properties of polyacrylamide (PAA) and (polydimethylsiloxane) PDMS on cardiac cell behavior. SOFT MATTER 2021; 17:1156-1172. [PMID: 33427281 DOI: 10.1039/d0sm01986k] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In vitro cell culture is commonly applied in laboratories around the world. Cultured cells are either of primary origin or established cell lines. Such transformed cell lines are increasingly replaced by pluripotent stem cell derived organotypic cells with more physiological properties. The quality of the culture conditions and matrix environment is of considerable importance in this regard. In fact, mechanical cues of the extracellular matrix have substantial effects on the cellular physiology. This is especially true if contractile cells such as cardiomyocytes are cultured. Therefore, elastic biomaterials have been introduced as scaffolds in 2D and 3D culture models for different cell types, cardiac cells among them. In this review, key aspects of cell-matrix interaction are highlighted with focus on cardiomyocytes and chemical properties as well as strengths and potential pitfalls in using two commonly applied polymers for soft matrix engineering, polyacrylamide (PAA) and polydimethylsiloxane (PDMS) are discussed.
Collapse
Affiliation(s)
- Karim Daliri
- Institute for Neurophysiology, University of Cologne, Medical Faculty, Robert Koch Str. 39, 50931 Cologne, Germany.
| | - Kurt Pfannkuche
- Institute for Neurophysiology, University of Cologne, Medical Faculty, Robert Koch Str. 39, 50931 Cologne, Germany. and Department for Pediatric Cardiology, University Hospital Cologne, Cologne, Germany and Marga-and-Walter-Boll Laboratory for Cardiac Tissue Engineering, University of Cologne, Germany and Center for Molecular Medicine, University of Cologne, Germany
| | - Bora Garipcan
- Institute of Biomedical Engineering, Bogazici University, Cengelkoy, 34684, Istanbul, Turkey.
| |
Collapse
|
49
|
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
|
50
|
Roman B, Kumar SA, Allen SC, Delgado M, Moncayo S, Reyes AM, Suggs LJ, Chintalapalle R, Li C, Joddar B. A Model for Studying the Biomechanical Effects of Varying Ratios of Collagen Types I and III on Cardiomyocytes. Cardiovasc Eng Technol 2021; 12:311-324. [PMID: 33432515 PMCID: PMC8972084 DOI: 10.1007/s13239-020-00514-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/22/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE To develop a novel model composed solely of Col I and Col III with the lower and upper limits set to include the ratios of Col I and Col III at 3:1 and 9:1 in which the structural and mechanical behavior of the resident CM can be studied. Further, the progression of fibrosis due to change in ratios of Col I:Col III was tested. METHODS Collagen gels with varying Col I:Col III ratios to represent a healthy (3:1) and diseased myocardial tissue were prepared by manually casting them in wells. Absorbance assay was performed to confirm the gelation of the gels. Rheometric analysis was performed on each of the collagen gels prepared to determine the varying stiffnesses and rheological parameters of the gels made with varying ratios of Col I:Col III. Second Harmonic Generation (SHG) was performed to observe the 3D characterization of the collagen samples. Scanning Electron microscopy was used for acquiring cross sectional images of the lyophilized collagen gels. AC16 CM (human) cell lines were cultured in the prepared gels to study cell morphology and behavior as a result of the varying collagen ratios. Cellular proliferation was studied by performing a Cell Trace Violet Assay and the applied force on each cell was measured by means of Finite Element Analysis (FEA) on CM from each sample. RESULTS Second harmonic generation microscopy used to image Col I, displayed a decrease in acquired image intensity with an increase in the non-second harmonic Col III in 3:1 gels. SEM showed a fiber-rich structure in the 3:1 gels with well-distributed pores unlike the 9:1 gels or the 1:0 controls. Rheological analysis showed a decrease in substrate stiffness with an increase of Col III, in comparison with other cases. CM cultured within 3:1 gels exhibited an elongated rod-like morphology with an average end-to-end length of 86 ± 28.8 µm characteristic of healthy CM, accompanied by higher cell growth in comparison with other cases. Finite element analysis used to estimate the forces exerted on CM cultured in the 3:1 gels, showed that the forces were well dispersed, and not concentrated within the center of cells, in comparison with other cases. CONCLUSION This study model can be adopted to simulate various biomechanical environments in which cells crosstalk with the Collagen-matrix in diseased pathologies to generate insights on strategies for prevention of fibrosis.
Collapse
Affiliation(s)
- Brian Roman
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Shweta Anil Kumar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Shane C Allen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Monica Delgado
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Sabastian Moncayo
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA
- Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Andres M Reyes
- Department of Physics, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Laura J Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ramana Chintalapalle
- Department of Mechanical Engineering, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Chunqiang Li
- Department of Physics, The University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), El Paso, USA.
- Border Biomedical Research Center, University of Texas at El Paso, 500 W University Avenue, El Paso, TX, 79968, USA.
| |
Collapse
|