1
|
House A, Santillan A, Correa E, Youssef V, Guvendiren M. Cellular Alignment and Matrix Stiffening Induced Changes in Human Induced Pluripotent Stem Cell Derived Cardiomyocytes. Adv Healthc Mater 2025; 14:e2402228. [PMID: 39468891 DOI: 10.1002/adhm.202402228] [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: 07/03/2024] [Revised: 10/15/2024] [Indexed: 10/30/2024]
Abstract
Biological processes are inherently dynamic, necessitating biomaterial platforms capable of spatiotemporal control over cellular organization and matrix stiffness for accurate study of tissue development, wound healing, and disease. However, most in vitro platforms remain static. In this study, a dynamic biomaterial platform comprising a stiffening hydrogel is introduced and achieved through a stepwise approach of addition followed by light-mediated crosslinking, integrated with an elastomeric substrate featuring strain-responsive lamellar surface patterns. Employing this platform, the response of human induced pluripotent stem cell-derived cardiomyocytes (hIPSC-CMs) is investigated to dynamic stiffening from healthy to fibrotic tissue stiffness. The results demonstrate that culturing hIPSC-CMs on physiologically relevant healthy stiffness significantly enhances their function, as evidenced by increased sarcomere fraction, wider sarcomere width, significantly higher connexin-43 content, and elevated cell beating frequency compared to cells cultured on fibrotic matrix. Conversely, dynamic matrix stiffening negatively impacts hIPSC-CM function, with earlier stiffening events exerting a more pronounced hindering effect. These findings provide valuable insights into material-based approaches for addressing existing challenges in hIPSC-CM maturation and have broader implications across various tissue models, including muscle, tendon, nerve, and cornea, where both cellular alignment and matrix stiffening play pivotal roles in tissue development and regeneration.
Collapse
Affiliation(s)
- Andrew House
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Anjeli Santillan
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Evan Correa
- Federated Department of Biological Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Victoria Youssef
- Federated Department of Biological Sciences, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| | - Murat Guvendiren
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, 07102, USA
| |
Collapse
|
2
|
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
|
3
|
Blazeski A, Garcia-Cardena G, Kamm RD. Advancing Cardiac Organoid Engineering Through Application of Biophysical Forces. IEEE Rev Biomed Eng 2024; PP:211-230. [PMID: 40030454 DOI: 10.1109/rbme.2024.3514378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Cardiac organoids represent an important bioengineering opportunity in the development of models to study human heart pathophysiology. By incorporating multiple cardiac cell types in three-dimensional culture and developmentally-guided biochemical signaling, cardiac organoids recapitulate numerous features of heart tissue. However, cardiac tissue also experiences a variety of mechanical forces as the heart develops and over the course of each contraction cycle. It is now clear that these forces impact cellular specification, phenotype, and function, and should be incorporated into the engineering of cardiac organoids in order to generate better models. In this review, we discuss strategies for engineering cardiac organoids and report the effects of organoid design on the function of cardiac cells. We then discuss the mechanical environment of the heart, including forces arising from tissue elasticity, contraction, blood flow, and stretch, and report on efforts to mimic these biophysical cues in cardiac organoids. Finally, we review emerging areas of cardiac organoid research, for the study of cardiac development, the formation of multi-organ models, and the simulation of the effects of spaceflight on cardiac tissue and consider how these investigations might benefit from the inclusion of mechanical cues.
Collapse
|
4
|
Wen Y, Yang H, Hong Y. Transcriptomic Approaches to Cardiomyocyte-Biomaterial Interactions: A Review. ACS Biomater Sci Eng 2024; 10:4175-4194. [PMID: 38934720 DOI: 10.1021/acsbiomaterials.4c00303] [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] [Indexed: 06/28/2024]
Abstract
Biomaterials, essential for supporting, enhancing, and repairing damaged tissues, play a critical role in various medical applications. This Review focuses on the interaction of biomaterials and cardiomyocytes, emphasizing the unique significance of transcriptomic approaches in understanding their interactions, which are pivotal in cardiac bioengineering and regenerative medicine. Transcriptomic approaches serve as powerful tools to investigate how cardiomyocytes respond to biomaterials, shedding light on the gene expression patterns, regulatory pathways, and cellular processes involved in these interactions. Emerging technologies such as bulk RNA-seq, single-cell RNA-seq, single-nucleus RNA-seq, and spatial transcriptomics offer promising avenues for more precise and in-depth investigations. Longitudinal studies, pathway analyses, and machine learning techniques further improve the ability to explore the complex regulatory mechanisms involved. This review also discusses the challenges and opportunities of utilizing transcriptomic techniques in cardiomyocyte-biomaterial research. Although there are ongoing challenges such as costs, cell size limitation, sample differences, and complex analytical process, there exist exciting prospects in comprehensive gene expression analyses, biomaterial design, cardiac disease treatment, and drug testing. These multimodal methodologies have the capacity to deepen our understanding of the intricate interaction network between cardiomyocytes and biomaterials, potentially revolutionizing cardiac research with the aim of promoting heart health, and they are also promising for studying interactions between biomaterials and other cell types.
Collapse
Affiliation(s)
- Yufeng Wen
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas 76207, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| |
Collapse
|
5
|
Kistamás K, Müller A, Muenthaisong S, Lamberto F, Zana M, Dulac M, Leal F, Maziz A, Costa P, Bernotiene E, Bergaud C, Dinnyés A. Multifactorial approaches to enhance maturation of human iPSC-derived cardiomyocytes. J Mol Liq 2023; 387:122668. [DOI: 10.1016/j.molliq.2023.122668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Harbuz I, Banciu DD, David R, Cercel C, Cotîrță O, Ciurea BM, Radu SM, Dinescu S, Jinga SI, Banciu A. Perspectives on Scaffold Designs with Roles in Liver Cell Asymmetry and Medical and Industrial Applications by Using a New Type of Specialized 3D Bioprinter. Int J Mol Sci 2023; 24:14722. [PMID: 37834167 PMCID: PMC10573170 DOI: 10.3390/ijms241914722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Cellular asymmetry is an important element of efficiency in the compartmentalization of intracellular chemical reactions that ensure efficient tissue function. Improving the current 3D printing methods by using cellular asymmetry is essential in producing complex tissues and organs such as the liver. The use of cell spots containing at least two cells and basement membrane-like bio support materials allows cells to be tethered at two points on the basement membrane and with another cell in order to maintain cell asymmetry. Our model is a new type of 3D bioprinter that uses oriented multicellular complexes with cellular asymmetry. This novel approach is necessary to replace the sequential and slow processes of organogenesis with rapid methods of growth and 3D organ printing. The use of the extracellular matrix in the process of bioprinting with cells allows one to preserve the cellular asymmetry in the 3D printing process and thus preserve the compartmentalization of biological processes and metabolic efficiency.
Collapse
Affiliation(s)
- Iuliana Harbuz
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Daniel Dumitru Banciu
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Rodica David
- Institute for Research on the Quality of Society and the Sciences of Education, University Constantin Brancusi of Targu Jiu, Republicii 1, 210185 Targu Jiu, Romania;
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Cristina Cercel
- University of Medicine and Pharmacy “Carol Davila” Bucharest, 37 Dionisie Lupu Street, 020021 Bucharest, Romania;
| | - Octavian Cotîrță
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Bogdan Marius Ciurea
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Sorin Mihai Radu
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Stela Dinescu
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Sorin Ion Jinga
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Adela Banciu
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| |
Collapse
|
7
|
Vuorenpää H, Björninen M, Välimäki H, Ahola A, Kroon M, Honkamäki L, Koivumäki JT, Pekkanen-Mattila M. Building blocks of microphysiological system to model physiology and pathophysiology of human heart. Front Physiol 2023; 14:1213959. [PMID: 37485060 PMCID: PMC10358860 DOI: 10.3389/fphys.2023.1213959] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
Microphysiological systems (MPS) are drawing increasing interest from academia and from biomedical industry due to their improved capability to capture human physiology. MPS offer an advanced in vitro platform that can be used to study human organ and tissue level functions in health and in diseased states more accurately than traditional single cell cultures or even animal models. Key features in MPS include microenvironmental control and monitoring as well as high biological complexity of the target tissue. To reach these qualities, cross-disciplinary collaboration from multiple fields of science is required to build MPS. Here, we review different areas of expertise and describe essential building blocks of heart MPS including relevant cardiac cell types, supporting matrix, mechanical stimulation, functional measurements, and computational modelling. The review presents current methods in cardiac MPS and provides insights for future MPS development with improved recapitulation of human physiology.
Collapse
Affiliation(s)
- Hanna Vuorenpää
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Miina Björninen
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hannu Välimäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti Ahola
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mart Kroon
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Honkamäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jussi T. Koivumäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
8
|
Van de Sande D, Ghasemi M, Watters T, Burton F, Pham L, Altrocchi C, Gallacher DJ, Lu H, Smith G. Does Enhanced Structural Maturity of hiPSC-Cardiomyocytes Better for the Detection of Drug-Induced Cardiotoxicity? Biomolecules 2023; 13:676. [PMID: 37189424 PMCID: PMC10135569 DOI: 10.3390/biom13040676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are currently used following the Comprehensive in vitro Proarrhythmic Assay (CiPA) initiative and subsequent recommendations in the International Council for Harmonization (ICH) guidelines S7B and E14 Q&A, to detect drug-induced cardiotoxicity. Monocultures of hiPSC-CMs are immature compared to adult ventricular cardiomyocytes and might lack the native heterogeneous nature. We investigated whether hiPSC-CMs, treated to enhance structural maturity, are superior in detecting drug-induced changes in electrophysiology and contraction. This was achieved by comparing hiPSC-CMs cultured in 2D monolayers on the current standard (fibronectin matrix, FM), to monolayers on a coating known to promote structural maturity (CELLvo™ Matrix Plus, MM). Functional assessment of electrophysiology and contractility was made using a high-throughput screening approach involving the use of both voltage-sensitive fluorescent dyes for electrophysiology and video technology for contractility. Using 11 reference drugs, the response of the monolayer of hiPSC-CMs was comparable in the two experimental settings (FM and MM). The data showed no functionally relevant differences in electrophysiology between hiPSC-CMs in standard FM and MM, while contractility read-outs indicated an altered amplitude of contraction but not changes in time course. RNA profiling for cardiac proteins shows similarity of the RNA expression across the two forms of 2D culture, suggesting that cell-to-matrix adhesion differences may explain account for differences in contraction amplitude. The results support the view that hiPSC-CMs in both 2D monolayer FM and MM that promote structural maturity are equally effective in detecting drug-induced electrophysiological effects in functional safety studies.
Collapse
Affiliation(s)
- Dieter Van de Sande
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Mohammadreza Ghasemi
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| | - Taylor Watters
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| | - Francis Burton
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| | - Ly Pham
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Cristina Altrocchi
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - David J. Gallacher
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Huarong Lu
- Global Safety Pharmacology, Nonclinical Safety, Janssen Pharmaceutical NV, B-2340 Beerse, Belgium
| | - Godfrey Smith
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
- Clyde Biosciences Limited, BioCity Scotland, Lanarkshire ML1 5UH, Scotland, UK
| |
Collapse
|
9
|
Dvornikov AV, Bunch TA, Lepak VC, Colson BA. Fluorescence lifetime-based assay reports structural changes in cardiac muscle mediated by effectors of contractile regulation. J Gen Physiol 2023; 155:e202113054. [PMID: 36633587 PMCID: PMC9859762 DOI: 10.1085/jgp.202113054] [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: 11/11/2021] [Revised: 09/23/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Cardiac muscle contraction is regulated by Ca2+-induced structural changes of the thin filaments to permit myosin cross-bridge cycling driven by ATP hydrolysis in the sarcomere. In congestive heart failure, contraction is weakened, and thus targeting the contractile proteins of the sarcomere is a promising approach to therapy. However, development of novel therapeutic interventions has been challenging due to a lack of precise discovery tools. We have developed a fluorescence lifetime-based assay using an existing site-directed probe, N,N'-dimethyl-N-(iodoacetyl)-N'-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)ethylenediamine (IANBD) attached to human cardiac troponin C (cTnC) mutant cTnCT53C, exchanged into porcine cardiac myofibrils. We hypothesized that IANBD-cTnCT53C fluorescence lifetime measurements provide insight into the activation state of the thin filament. The sensitivity and precision of detecting structural changes in cTnC due to physiological and therapeutic modulators of thick and thin filament functions were determined. The effects of Ca2+ binding to cTnC and myosin binding to the thin filament were readily detected by this assay in mock high-throughput screen tests using a fluorescence lifetime plate reader. We then evaluated known effectors of altered cTnC-Ca2+ binding, W7 and pimobendan, and myosin-binding drugs, mavacamten and omecamtiv mecarbil, used to treat cardiac diseases. Screening assays were determined to be of high quality as indicated by the Z' factor. We conclude that cTnC lifetime-based probes allow for precise evaluation of the thin filament activation in functioning myofibrils that can be used in future high-throughput screens of small-molecule modulators of function of the thin and thick filaments.
Collapse
Affiliation(s)
- Alexey V. Dvornikov
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Thomas A. Bunch
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Victoria C. Lepak
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Brett A. Colson
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
10
|
Pioner JM, Santini L, Palandri C, Langione M, Grandinetti B, Querceto S, Martella D, Mazzantini C, Scellini B, Giammarino L, Lupi F, Mazzarotto F, Gowran A, Rovina D, Santoro R, Pompilio G, Tesi C, Parmeggiani C, Regnier M, Cerbai E, Mack DL, Poggesi C, Ferrantini C, Coppini R. Calcium handling maturation and adaptation to increased substrate stiffness in human iPSC-derived cardiomyocytes: The impact of full-length dystrophin deficiency. Front Physiol 2022; 13:1030920. [PMID: 36419836 PMCID: PMC9676373 DOI: 10.3389/fphys.2022.1030920] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
Cardiomyocytes differentiated from human induced Pluripotent Stem Cells (hiPSC- CMs) are a unique source for modelling inherited cardiomyopathies. In particular, the possibility of observing maturation processes in a simple culture dish opens novel perspectives in the study of early-disease defects caused by genetic mutations before the onset of clinical manifestations. For instance, calcium handling abnormalities are considered as a leading cause of cardiomyocyte dysfunction in several genetic-based dilated cardiomyopathies, including rare types such as Duchenne Muscular Dystrophy (DMD)-associated cardiomyopathy. To better define the maturation of calcium handling we simultaneously measured action potential and calcium transients (Ca-Ts) using fluorescent indicators at specific time points. We combined micropatterned substrates with long-term cultures to improve maturation of hiPSC-CMs (60, 75 or 90 days post-differentiation). Control-(hiPSC)-CMs displayed increased maturation over time (90 vs 60 days), with longer action potential duration (APD), increased Ca-T amplitude, faster Ca-T rise (time to peak) and Ca-T decay (RT50). The progressively increased contribution of the SR to Ca release (estimated by post-rest potentiation or Caffeine-induced Ca-Ts) appeared as the main determinant of the progressive rise of Ca-T amplitude during maturation. As an example of severe cardiomyopathy with early onset, we compared hiPSC-CMs generated from a DMD patient (DMD-ΔExon50) and a CRISPR-Cas9 genome edited cell line isogenic to the healthy control with deletion of a G base at position 263 of the DMD gene (c.263delG-CMs). In DMD-hiPSC-CMs, changes of Ca-Ts during maturation were less pronounced: indeed, DMD cells at 90 days showed reduced Ca-T amplitude and faster Ca-T rise and RT50, as compared with control hiPSC-CMs. Caffeine-Ca-T was reduced in amplitude and had a slower time course, suggesting lower SR calcium content and NCX function in DMD vs control cells. Nonetheless, the inotropic and lusitropic responses to forskolin were preserved. CRISPR-induced c.263delG-CM line recapitulated the same developmental calcium handling alterations observed in DMD-CMs. We then tested the effects of micropatterned substrates with higher stiffness. In control hiPSC-CMs, higher stiffness leads to higher amplitude of Ca-T with faster decay kinetics. In hiPSC-CMs lacking full-length dystrophin, however, stiffer substrates did not modify Ca-Ts but only led to higher SR Ca content. These findings highlighted the inability of dystrophin-deficient cardiomyocytes to adjust their calcium homeostasis in response to increases of extracellular matrix stiffness, which suggests a mechanism occurring during the physiological and pathological development (i.e. fibrosis).
Collapse
Affiliation(s)
| | - Lorenzo Santini
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Chiara Palandri
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Marianna Langione
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Bruno Grandinetti
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, Florence, Italy
| | - Silvia Querceto
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, Florence, Italy
| | - Daniele Martella
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, Florence, Italy
- Istituto Nazionale di Ricerca Metrologica (INRiM), Turin, Italy
| | | | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Flavia Lupi
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, Florence, Italy
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Aoife Gowran
- 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
| | - Rosaria Santoro
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
| | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Camilla Parmeggiani
- European Laboratory for Non-Linear Spectroscopy (LENS), University of Florence, Florence, Italy
- Department of Chemistry “Ugo Schiff”, University of Florence, Florence, Italy
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | | | - David L. Mack
- Department of Bioengineering, University of Washington, Seattle, WA, United States
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Raffaele Coppini
- Department of Neurofarba, University of Florence, Florence, Italy
| |
Collapse
|