1
|
Esmaeili H, Zhang Y, Ravi K, Neff K, Zhu W, Migrino RQ, Park JG, Nikkhah M. Development of an electroconductive Heart-on-a-chip model to investigate cellular and molecular response of human cardiac tissue to gold nanomaterials. Biomaterials 2025; 320:123275. [PMID: 40138961 DOI: 10.1016/j.biomaterials.2025.123275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
To date, various strategies have been developed to construct biomimetic and functional in vitro cardiac tissue models utilizing human induced pluripotent stem cells (hiPSCs). Among these approaches, microfluidic-based Heart-on-a-chip (HOC) models are promising, as they enable the engineering of miniaturized, physiologically relevant in vitro cardiac tissues with precise control over cellular constituents and tissue architecture. Despite significant advancements, previously reported HOC models often lack the electroconductivity features of the native human myocardium. In this study, we developed a 3D electroconductive HOC (referred to as eHOC) model through the co-culture of isogenic hiPSC-derived cardiomyocytes (hiCMs) and cardiac fibroblasts (hiCFs), embedded within an electroconductive hydrogel scaffold in a microfluidic-based chip system. Functional and gene expression analyses demonstrated that, compared to non-conductive HOC, the eHOC model exhibited enhanced contractile functionality, improved calcium transients, and increased expression of structural and calcium handling genes. The eHOC model was further leveraged to investigate the underlying electroconduction-induced pathway(s) associated with cardiac tissue development through single-cell RNA sequencing (scRNA-seq). Notably, scRNA-seq analyses revealed a significant downregulation of a set of cardiac genes, associated with the fetal stage of heart development, as well as upregulation of sarcomere- and conduction-related genes within the eHOC model. Additionally, upregulation of the cardiac muscle contraction and motor protein pathways were observed in the eHOC model, consistent with enhanced contractile functionality of the engineered cardiac tissues. Comparison of scRNA-seq data from the 3D eHOC model with published datasets of adult human hearts demonstrated a similar expression pattern of fetal- and adult-like cardiac genes. Overall, this study provides a unique eHOC model with improved biomimcry and organotypic features, which could be potentially used for drug testing and discovery, as well as disease modeling applications.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Yining Zhang
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Kalpana Ravi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Keagan Neff
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, 85022, USA; University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Jin G Park
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Virginia G. Piper Center for Personalized Diagnosis, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
2
|
Li J, Yang Y, Yi Z, Zhu Y, Yang H, Chen B, Lobie PE, Ma S. Microdroplet-Engineered Skeletal Muscle Organoids from Primary Tissue Recapitulate Parental Physiology with High Reproducibility. RESEARCH (WASHINGTON, D.C.) 2025; 8:0699. [PMID: 40375923 PMCID: PMC12078942 DOI: 10.34133/research.0699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/18/2025]
Abstract
Achieving high maturity and functionality in in vitro skeletal muscle models is essential for advancing our understanding of muscle biology, disease mechanisms, and drug discovery. However, current models struggle to fully recapitulate key features such as sarcomere structure, muscle fiber composition, and contractile function while also ensuring consistency and rapid production. Adult stem cells residing in muscle tissue are known for their powerful regenerative potential, yet tissue-derived skeletal muscle organoids have not been established. In this study, we introduce droplet-engineered skeletal muscle organoids derived from primary tissue using cascade-tubing microfluidics. These droplet-engineered organoids (DEOs) exhibit high maturity, including well-developed striated sarcomeres, spontaneous and stimulated contractions, and recapitulation of parental muscle fiber types. Notably, DEOs are produced in just 8 d without the need for primary cell culture-substantially accelerating the 50- to 60-d process required by classical organoid models. Additionally, the cascade-tubing microfluidics platform enables high-throughput production of hundreds of uniform DEO replicates from a small tissue sample, providing a scalable and reproducible solution for skeletal muscle research and drug screening.
Collapse
Affiliation(s)
- Jiawei Li
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
- Meatoid Biotechnology Limited, Shenzhen 518107, China
| | - Yiming Yang
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
| | - Ziqi Yi
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Yu Zhu
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Haowei Yang
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Baiming Chen
- School of Medicine,
The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Peter E. Lobie
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
- Key Lab of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education Institutes,
Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| |
Collapse
|
3
|
Tornberg K, Grötsch W, Ritari N, Haikka S, Sukki L, Aalto-Setälä K, Pekkanen-Mattila M, Kallio P. Compartmentalized perfusion for temporal control of the chemical microenvironment of iPSC-derived cardiac cells. LAB ON A CHIP 2025. [PMID: 40370144 DOI: 10.1039/d5lc00072f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Organ-on-chip structures are predicted to have a significant influence in drug research. In these structures, perfusion can provide cells a more controllable environment to receive signaling molecules. In many current organ-on-chip applications, perfusion is used for shear stress stimulus for the cells, but it can also provide a more precise way of controlling the chemical microenvironment around the cells. In this paper, we propose an open-top organ-on-chip structure with compartment-specific perfusion to introduce stimulating molecules to cells with only minimal extra unspecific stimulus. Using numerical simulations, we show that shear stress sensed by the cells within the structure is low. We further validated the flow profile experimentally. We showed that the hiPSC-CMs accommodate to the flow environment where the shear stress is kept below 0.035 mPa. We also show that the beating rate of hiPSC-CMs increases due to the stimulation provided by chemical stimulant molecules introduced through the flow.
Collapse
Affiliation(s)
- Kaisa Tornberg
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Wolfram Grötsch
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Niina Ritari
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Saara Haikka
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Lassi Sukki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | | | | | - Pasi Kallio
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
4
|
Quarta R, Cristiano E, Han MKL, Boccanegra B, Marinelli M, Gaio N, Ohana J, Mouly V, Cappellari O, De Luca A. Patient-Oriented In Vitro Studies in Duchenne Muscular Dystrophy: Validation of a 3D Skeletal Muscle Organoid Platform. Biomedicines 2025; 13:1109. [PMID: 40426938 PMCID: PMC12109395 DOI: 10.3390/biomedicines13051109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Three-dimensional skeletal muscle organoids (3D SkMO) are becoming of increasing interest for preclinical studies in Duchenne muscular dystrophy (DMD), provided that the used platform demonstrates the possibility to form functional and reproducible 3D SkMOs, to investigate on potential patient-related phenotypic differences. Methods: In this study, we employed fibrin-based 3D skeletal muscle organoids derived from immortalized myogenic precursors of DMD patients carrying either a stop codon mutation in exon 59 or a 48-50 deletion. We compared dystrophic lines with a healthy wild-type control (HWT) by assessing microtissue formation ability, contractile function at multiple timepoints along with intracellular calcium dynamics via calcium imaging, as well as expression of myogenic markers. Results: We found patient-specific structural and functional differences in the early stages of 3D SkMO development. Contractile force, measured as both single twitch and tetanic responses, was significantly lower in dystrophic 3D SkMOs compared to HWT, with the most pronounced differences observed at day 7 of differentiation. However, these disparities diminished over time under similar culturing conditions and in the absence of continuous nerve-like stimulation, suggesting that the primary deficit lies in delayed myogenic maturation, as also supported by gene expression analysis. Conclusions: Our results underline that, despite the initial maturation delay, DMD muscle precursors retain the capacity to form functional 3D SkMOs once this intrinsic lag is overcome. This suggests a critical role of dystrophin in early myogenic development, while contraction-induced stress and/or an inflammatory microenvironment are essential to fully recapitulate dystrophic phenotypes in 3D SkMOs.
Collapse
Affiliation(s)
- Raffaella Quarta
- Department of Pharmacy Drug Science, University of Bari Aldo Moro, 70125 Bari, Italy; (R.Q.); (B.B.); (A.D.L.)
| | - Enrica Cristiano
- Department of Pharmacy Drug Science, University of Bari Aldo Moro, 70125 Bari, Italy; (R.Q.); (B.B.); (A.D.L.)
| | | | - Brigida Boccanegra
- Department of Pharmacy Drug Science, University of Bari Aldo Moro, 70125 Bari, Italy; (R.Q.); (B.B.); (A.D.L.)
| | - Manuel Marinelli
- Department of Pharmacy Drug Science, University of Bari Aldo Moro, 70125 Bari, Italy; (R.Q.); (B.B.); (A.D.L.)
| | - Nikolas Gaio
- BIOND Solutions B.V., 2629 JD Delft, The Netherlands
| | - Jessica Ohana
- Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, 75013 Paris, France
| | - Vincent Mouly
- Institut de Myologie, Centre de Recherche en Myologie, Sorbonne Université, 75013 Paris, France
| | - Ornella Cappellari
- Department of Pharmacy Drug Science, University of Bari Aldo Moro, 70125 Bari, Italy; (R.Q.); (B.B.); (A.D.L.)
| | - Annamaria De Luca
- Department of Pharmacy Drug Science, University of Bari Aldo Moro, 70125 Bari, Italy; (R.Q.); (B.B.); (A.D.L.)
| |
Collapse
|
5
|
Ng XJ, Esser TU, Trossmann VT, Rudisch C, Fiedler M, Roshanbinfar K, Lamberger Z, Stahlhut P, Lang G, Scheibel T, Engel FB. Enhancing Form Stability: Shrink-Resistant Hydrogels Made of Interpenetrating Networks of Recombinant Spider Silk and Collagen-I. Adv Healthc Mater 2025; 14:e2500311. [PMID: 40143764 PMCID: PMC12057611 DOI: 10.1002/adhm.202500311] [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: 01/18/2025] [Revised: 03/09/2025] [Indexed: 03/28/2025]
Abstract
Tissue engineering enables the production of tissues and organ-like structures as models for drug testing and mechanistical studies or functional replacements for injured tissues. Available cytocompatible materials are limited in number, suffer from insufficient mechanical properties, and cells interacting with them often cause construct shrinkage. As shape is important for function, identifying cytocompatible, shrink-resistant materials are a major aim. Here, it is shown that hydrogels made of interpenetrating networks of collagen-I and recombinant spider silk protein eADF4(C16)-RGD nanofibrils exhibit synergistic and tunable mechanical properties. Composite hydrogels allow cell adhesion and spreading and are resistant to shrinkage mediated by fibroblasts, C2C12 myoblasts, and human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes. Myoblasts differentiate and fuse into myotubes, and hiPSC-cardiomyocytes can be cultured long-term, show spontaneous contractions, and remain drug responsive. Collectively, a novel composite material is developed to overcome the challenge of post-fabrication matrix shrinkage conferring high shape fidelity suitable for tissue engineering.
Collapse
Affiliation(s)
- Xuen J. Ng
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Tilman U. Esser
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Vanessa T. Trossmann
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Christoph Rudisch
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Maren Fiedler
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Zan Lamberger
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Gregor Lang
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Thomas Scheibel
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
- Bayreuth Center for Colloids and Interfaces (BZKG)University of Bayreuth95447BayreuthGermany
- Bavarian Polymer Institute (BPI)University of Bayreuth95447BayreuthGermany
- Bayreuth Center for Molecular Biosciences (BZMB)University of Bayreuth95447BayreuthGermany
- Bayreuth Center for Material Science (BayMAT)University of Bayreuth95447BayreuthGermany
- Faculty of MedicineUniversity of Würzburg97080WürzburgGermany
| | - Felix B. Engel
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| |
Collapse
|
6
|
Ma J, Ladd DM, Kaval N, Wang HS. Toxicity of long term exposure to low dose polystyrene microplastics and nanoplastics in human iPSC-derived cardiomyocytes. Food Chem Toxicol 2025; 202:115489. [PMID: 40312000 DOI: 10.1016/j.fct.2025.115489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/27/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Microplastics and nanoplastics (MNPs) are widespread environmental pollutants with potential risks to human health including cardiovascular effects. However, the impact of MNPs on the heart, particularly in human-relevant cardiac models, remains poorly understood. In this study, we investigated the long term effects of polystyrene (PS) MNPs-1 μm (PS-1) and 0.05 μm (PS-0.05) in human iPSC-derived cardiomyocytes (hiPSC-CMs). PS MNPs exposure reduced myocyte viability in a time- and dose-dependent manner. At a low dose of 0.1 μg/L, both PS-0.05 and PS-1 suppressed myocyte contractility, reduced Ca2+ transient amplitude, and altered contraction and Ca2+ transient dynamics. In hypertrophic hiPSC-CMs, PS-0.05 exposure exacerbated hypertrophy, increasing cell size and proBNP expression, a marker of myocyte hypertrophy. The mechanism of PS MNPs-induced cardiotoxicity likely involved mitochondrial dysfunction, as indicated by decreased mitochondrial membrane potential, increased mitochondrial ROS, and elevated intracellular ROS levels. This is the first study to assess the long term impact of low dose MNPs in human cardiomyocytes, providing crucial insight into the potential cardiac toxicity of MNPs and their implications for human heart health.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Drew M Ladd
- Department of Chemistry, College of Arts and Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Necati Kaval
- Department of Chemistry, College of Arts and Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
7
|
Bissoli I, Alabiso F, Cosentino C, Seragnoli Chystyakova A, Ferré F, Alviano F, Marrazzo P, Pignatti C, Agnetti G, Regazzi R, Flamigni F, D'Adamo S, Cetrullo S. Modeling heart failure by induced pluripotent stem cell-derived organoids. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167861. [PMID: 40254266 DOI: 10.1016/j.bbadis.2025.167861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cardiac organoids offer significant advantages for in vitro studies, as their 3D structure and cellular composition more closely replicate tissue complexity compared to 2D models. This is particularly relevant for studying complex diseases like heart failure (HF), which involve multiple cell types and cardiac structures. Thus, the primary aim of this study was to produce self-assembled, scaffold-free cardiac organoids from induced pluripotent stem cells (iPSCs), capable of simulating key aspects of HF in vitro. Gene expression analysis confirmed a transition from stemness markers (OCT4, NANOG) to cardiac markers (TNNT2, DES), validating their cardiac phenotype. To induce hallmark HF features, endothelin-1 (ET-1) treatment was applied. Key findings indicate that this experimental model successfully reproduced HF pathological markers, including the upregulation of genes encoding atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and the cytoskeletal protein α-skeletal actin (ACTA1), along with changes in microRNA (miR) expression profiles. Functionally, ET-1 treatment reduced organoid contractility, indicating a decline in contractile function-a hallmark of HF. Furthermore, histological analyses by Thioflavin T (ThT) staining, ThT fluorescence assay and filter trap assay on protein extracts demonstrated protein aggregation following ET-1 treatment. Co-administration of various nutraceuticals was shown to mitigate these effects. These findings underscore the value of this ET-1-stimulated cardiac organoid model as a powerful platform for studying HF mechanisms and evaluating novel therapeutic approaches.
Collapse
Affiliation(s)
- Irene Bissoli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
| | - Francesco Alabiso
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Cristina Cosentino
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | | - Fabrizio Ferré
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Francesco Alviano
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Pasquale Marrazzo
- Department of Biomolecular Sciences, Università degli Studi di Urbino Carlo Bo, Urbino, Italy
| | - Carla Pignatti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giulio Agnetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland; Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Flavio Flamigni
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Stefania D'Adamo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Silvia Cetrullo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy; Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy; Health Sciences and Technologies-Interdepartmental Center for Industrial Research (CIRI-SDV), University of Bologna, Bologna, Italy.
| |
Collapse
|
8
|
Choi YH, Leng J, Fan J, Ramirez RJ, Cho HC. Tissue elasticity modulates cardiac pacemaker cell automaticity. Am J Physiol Heart Circ Physiol 2025; 328:H978-H990. [PMID: 40080390 DOI: 10.1152/ajpheart.00813.2024] [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: 11/19/2024] [Revised: 12/17/2024] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
Tissue elasticity is essential to a broad spectrum of cell biology and organ function including the heart. Routine cell culture models on rigid polystyrene dishes are limited in studying the impact of tissue elasticity in distinct regions of the myocardium such as the cardiac conduction system. Gelatin, a derivative of collagen, is a simple and tunable platform for modeling tissue elasticity. We sought to study the effects of increasing tissue stiffness on cardiac pacemaker cell function by using transcription factor-reprogrammed pacemaker cells cultured on gelatin hydrogels with specific elasticity. Our data indicate that automaticity of the pacemaker cells, measured in rhythmic contractions and oscillating intracellular Ca2+ transients, was enhanced when cultured on a stiffer matrix of 14 kPa. This was accompanied by increased expression of cardiac pacemaker ion channel, Hcn4, and a reciprocal decrease in Cx43 expression compared with control conditions. Propagation of Ca2+ transients was slower in the pacemaker cell monolayers compared with control, which recapitulates a hallmark feature in the native pacemaker tissue. Ca2+ transient propagation of pacemaker cell monolayer was slower on stiffer than on softer hydrogel, and this was dependent on enhanced proliferation of cardiac fibroblasts rather than differences in gap junctional coupling. Culturing the pacemaker cells on rigid plastic plates led to irregular or loss of synchronous contractions as well as unusually long Ca2+ transient durations. Taken together, our data demonstrate that automaticity of pacemaker cells is augmented by stiffer extracellular matrix substrates within the elasticity range of the healthy myocardium. This simple approach presents a physiological in vitro model to study mechanoelectric feedback of cardiomyocytes including the conduction system cells.NEW & NOTEWORTHY The major achievement of this work is development of a robust and straightforward approach to model cardiac conduction system cells with a range of cardiac tissue elasticity with a goal to understand the impact of tissue stiffness on cardiac pacing. Our data provide a framework for further investigation of the heart rhythm in health and disease in the context of fibrosis.
Collapse
Affiliation(s)
- Young Hwan Choi
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Division of Pediatric Cardiac Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Jing Leng
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Division of Pediatric Cardiac Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Jinqi Fan
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Division of Pediatric Cardiac Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Rafael J Ramirez
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Division of Pediatric Cardiac Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| | - Hee Cheol Cho
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins Children's Center, Baltimore, Maryland, United States
- Division of Pediatric Cardiac Surgery, Department of Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
- Department of Biomedical Engineering, Johns Hopkins Whiting School of Engineering, Baltimore, Maryland, United States
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
9
|
Wang YH, Liu TT, Guo YP, Zhu SJ, Liao ZM, Song JM, Zhu XM, Liang JL, Nasser MI, Liu NB, Chang DH, Zhu P, Yao B. Integrating melt electrospinning writing and microfluidics to engineer a human cardiac microenvironment for high-fidelity drug screening. Bioact Mater 2025; 45:551-566. [PMID: 39759533 PMCID: PMC11696762 DOI: 10.1016/j.bioactmat.2024.11.037] [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: 10/28/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 01/07/2025] Open
Abstract
The preclinical evaluation of drug-induced cardiotoxicity is critical for developing novel drug, helping to avoid drug wastage and post-marketing withdrawal. Although human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and the engineered heart organoid have been used for drug screening and mimicking disease models, they are always limited by the immaturity and lack of functionality of the cardiomyocytes. In this study, we constructed a Cardiomyocytes-on-a-Chip (CoC) that combines micro-grooves (MGs) and circulating mechanical stimulation to recapitulate the well-organized structure and stable beating of myocardial tissue. The phenotypic changes and maturation of CMs cultured on the CoC have been verified and can be used for the evaluation of cardiotoxicity and cardioprotective drug responses. Taken together, these results highlight the ability of our myocardial microarray platform to accurately reflect clinical behaviour, underscoring its potential as a powerful pre-clinical tool for assessing drug response and toxicity.
Collapse
Affiliation(s)
- Yu-hong Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong, 510100, China
| | - Ting-ting Liu
- Department of Laboratory Diagnosis, The 971th Hospital, Qingdao, China
| | - Yan-ping Guo
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong, 510100, China
- School of Medicine, South China University of Technology, 510641, China
| | - Shuo-ji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong, 510100, China
- Department of Cardiac Surgery, The University of Tokyo Hospital, Tokyo, 113-8654, Japan
| | - Zi-ming Liao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, China
| | - Jia-mei Song
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, China
| | - Xi-ming Zhu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, China
| | - Jia-liang Liang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong, 510100, China
| | - Moussa Ide Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong, 510100, China
| | - Nan-bo Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong, 510100, China
| | - De-hua Chang
- Department of Cell Therapy in Regenerative Medicine, University of Tokyo Hospital, Tokyo, 113-8654, Japan
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong, 510100, China
| | - Bin Yao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, 300072, China
| |
Collapse
|
10
|
Vaziri N, Shutt TE, Karim W, Raedler TJ, Pantelis C, Thomas N, Jayaram M, Greenway SC, Bousman CA. Examination of mitochondria- and inflammasome-mediated mechanisms of clozapine-induced myocarditis using patient-derived iPSC cardiomyocytes. Mol Psychiatry 2025. [DOI: 10.1038/s41380-025-02935-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 01/22/2025] [Accepted: 02/12/2025] [Indexed: 03/20/2025]
|
11
|
Rivera-Arbeláez JM, Dostanić M, Windt LM, Stein JM, Cofiño-Fabres C, Boonen T, Wiendels M, van den Berg A, Segerink LI, Mummery CL, Sarro PM, van Meer BJ, Ribeiro MC, Mastrangeli M, Passier R. FORCETRACKER: A versatile tool for standardized assessment of tissue contractile properties in 3D Heart-on-Chip platforms. PLoS One 2025; 20:e0314985. [PMID: 39946364 PMCID: PMC11825004 DOI: 10.1371/journal.pone.0314985] [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: 05/28/2024] [Accepted: 11/19/2024] [Indexed: 02/16/2025] Open
Abstract
Engineered heart tissues (EHTs) have shown great potential in recapitulating tissue organization, functions, and cell-cell interactions of the human heart in vitro. Currently, multiple EHT platforms are used by both industry and academia for different applications, such as drug discovery, disease modelling, and fundamental research. The tissues' contractile force, one of the main hallmarks of tissue function and maturation level of cardiomyocytes, can be read out from EHT platforms by optically tracking the movement of elastic pillars induced by the contractile tissues. However, existing optical tracking algorithms which focus on calculating the contractile force are customized and platform-specific, often not available to the broad research community, and thus hamper head-to-head comparison of the model output. Therefore, there is the need for robust, standardized and platform-independent software for tissues' force assessment. To meet this need, we developed ForceTracker: a standalone and computationally efficient software for analyzing contractile properties of tissues in different EHT platforms. The software uses a shape-detection algorithm to single out and track the movement of pillars' tips for the most common shapes of EHT platforms. In this way, we can obtain information about tissues' contractile performance. ForceTracker is coded in Python and uses a multi-threading approach for time-efficient analysis of large data sets in multiple formats. The software efficiency to analyze circular and rectangular pillar shapes is successfully tested by analyzing different format videos from two EHT platforms, developed by different research groups. We demonstrate robust and reproducible performance of the software in the analysis of tissues over time and in various conditions. ForceTracker's detection and tracking shows low sensitivity to common incidental defects, such as alteration of tissue shape or air bubbles. Detection accuracy is determined via comparison with manual measurements using the software ImageJ. We developed ForceTracker as a tool for standardized analysis of contractile performance in EHT platforms to facilitate research on disease modeling and drug discovery in academia and industry.
Collapse
Affiliation(s)
- José M. Rivera-Arbeláez
- MESA+Institute for Nanotechnology, BIOS Lab on a Chip Group, Technical Medical Centre, Max Planck Center for Com-plex Fluid Dynamics, University of Twente, Enschede, The Netherlands
- Department of Bioengineering Technologies, Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Milica Dostanić
- Microelectronics, Delft University of Technology, Delft, The Netherlands
| | - Laura M. Windt
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jeroen M. Stein
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Carla Cofiño-Fabres
- Department of Bioengineering Technologies, Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Tom Boonen
- River BioMedics, Enschede, The Netherlands
| | - Maury Wiendels
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Albert van den Berg
- MESA+Institute for Nanotechnology, BIOS Lab on a Chip Group, Technical Medical Centre, Max Planck Center for Com-plex Fluid Dynamics, University of Twente, Enschede, The Netherlands
| | - Loes I. Segerink
- MESA+Institute for Nanotechnology, BIOS Lab on a Chip Group, Technical Medical Centre, Max Planck Center for Com-plex Fluid Dynamics, University of Twente, Enschede, The Netherlands
| | - Christine L. Mummery
- Department of Bioengineering Technologies, Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | - Robert Passier
- Department of Bioengineering Technologies, Applied Stem Cell Technologies Group, Technical Medical Centre, University of Twente, Enschede, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
12
|
Licata JP, Gerstenhaber JA, Lelkes PI. Novel, low-cost bioreactor for in vitro electrical stimulation of cardiac cells. Front Bioeng Biotechnol 2025; 13:1531731. [PMID: 39963172 PMCID: PMC11830680 DOI: 10.3389/fbioe.2025.1531731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction The successful implantation of laboratory-grown cardiac tissue requires phenotypically mature cardiomyocytes capable of electrophysiological integration with native heart tissue. Pulsed electrical stimulation (ES) has been identified as a promising strategy for enhancing cardiomyocyte maturation. However, there are discrepancies in the literature as to best practices for promoting cardiac differentiation using ES. Methods This study presents a novel, 3D printed bioreactor that delivers in vitro ES to human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), promoting cell maturity and functional readiness for implantation. Finite element analysis and mathematical modeling were used to model the fluid dynamics and to characterize in detail the delivery of pulsatile electrical signals, providing precise control over stimulation parameters such as voltage, current, and charge. Results The bioreactor developed here provides an easy-to-use, inexpensive platform for culturing hiPSC-CMs under the influence of ES and low-shear fluid flow for enhanced nutrient availability, while its "drop-in" design facilitates real-time observation of cultured cells. The electrical stimulation provided is controlled, modeled, and predictable, enabling reproducible experimental conditions and promoting comparability across future studies. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) grown in the bioreactor with ES showed improved differentiation and an enhanced ability to respond to external electrical pacing signals. Discussion By offering a standardized platform for ES-based cardiomyocyte maturation, this bioreactor aims to accelerate advancements in cardiac tissue engineering. Future research will explore how variations in ES parameters influence cardiomyocyte phenotype and maturation, contributing to a deeper understanding of cardiac cell development and optimization for therapeutic applications.
Collapse
Affiliation(s)
- Joseph P. Licata
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | | | - Peter I. Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| |
Collapse
|
13
|
Jiang Y, Xue M, Ou L, Wu H, Yang J, Zhang W, Zhou Z, Gao Q, Lin B, Kong W, Chen S, Sun D. Rapid Video Analysis for Contraction Synchrony of Human Induced Pluripotent Stem Cells-Derived Cardiac Tissues. Tissue Eng Regen Med 2025; 22:211-224. [PMID: 39804547 PMCID: PMC11794902 DOI: 10.1007/s13770-024-00688-4] [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: 08/24/2024] [Revised: 11/05/2024] [Accepted: 11/30/2024] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND The contraction behaviors of cardiomyocytes (CMs), especially contraction synchrony, are crucial factors reflecting their maturity and response to drugs. A wider field of view helps to observe more pronounced synchrony differences, but the accompanied greater computational load, requiring more computing power or longer computational time. METHODS We proposed a method that directly correlates variations in optical field brightness with cardiac tissue contraction status (CVB method), based on principles from physics and photometry, for rapid video analysis in wide field of view to obtain contraction parameters, such as period and contraction propagation direction and speed. RESULTS Through video analysis of human induced pluripotent stem cell (hiPSC)-derived CMs labeled with green fluorescent protein (GFP) cultured on aligned and random nanofiber scaffolds, the CVB method was demonstrated to obtain contraction parameters and quantify the direction and speed of contraction within regions of interest (ROIs) in wide field of view. The CVB method required less computation time compared to one of the contour tracking methods, the Lucas-Kanade (LK) optical flow method, and provided better stability and accuracy in the results. CONCLUSION This method has a smaller computational load, is less affected by motion blur and out-of-focus conditions, and provides a potential tool for accurate and rapid analysis of cardiac tissue contraction synchrony in wide field of view without the need for more powerful hardware.
Collapse
Affiliation(s)
- Yuqing Jiang
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| | - Mingcheng Xue
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| | - Lu Ou
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| | - Huiquan Wu
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| | - Jianhui Yang
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| | - Wangzihan Zhang
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| | - Zhuomin Zhou
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| | - Qiang Gao
- Guangdong Provincial People's Hospital, Guangzhou, 510080, Guangdong, China
| | - Bin Lin
- Guangdong Beating Origin Regenerative Medicine Co. Ltd., Foshan, 528231, Guangdong, China
| | - Weiwei Kong
- Guangdong Beating Origin Regenerative Medicine Co. Ltd., Foshan, 528231, Guangdong, China
| | - Songyue Chen
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China.
| | - Daoheng Sun
- Pen-Tung Sah Institute of Micro-Nano Science and Technology, Xiamen University, Xiamen, 361102, Fujian, China
| |
Collapse
|
14
|
Huang Z, Jia K, Tan Y, Yu Y, Xiao W, Zhou X, Yi J, Zhang C. Advances in cardiac organoid research: implications for cardiovascular disease treatment. Cardiovasc Diabetol 2025; 24:25. [PMID: 39827092 PMCID: PMC11743075 DOI: 10.1186/s12933-025-02598-8] [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: 11/19/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Globally, cardiovascular diseases remain among the leading causes of mortality, highlighting the urgent need for innovative research models. Consequently, the development of accurate models that simulate cardiac function holds significant scientific and clinical value for both disease research and therapeutic interventions. Cardiac organoids, which are three-dimensional structures derived from the induced differentiation of stem cells, are particularly promising. These organoids not only replicate the autonomous beating and essential electrophysiological properties of the heart but are also widely employed in studies related to cardiac diseases, drug efficacy testing, and regenerative medicine. This review comprehensively surveys the various fabrication techniques used to create cardiac organoids and their diverse applications in modeling a range of cardiac diseases. We emphasize the role of advanced technologies in enhancing the maturation and functionality of cardiac cells, ensuring that these models closely resemble native cardiac tissue. Furthermore, we discuss monitoring techniques and evaluation parameters critical for assessing the performance of cardiac organoids, considering the complex interactions within multi-organ systems. This approach is vital for enhancing precision and efficiency in drug development, allowing for more effective therapeutic strategies. Ultimately, this review aims to provide a thorough and innovative perspective on both fundamental research and clinical treatment of cardiovascular diseases, offering insights that could pave the way for future advancements in understanding and addressing these prevalent health challenges.
Collapse
Affiliation(s)
- Ziteng Huang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Keran Jia
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yadan Tan
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yang Yu
- Department of Cardiology, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Wudian Xiao
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiangyu Zhou
- Department of Thyroid Surgery, The Affiliated Hospital, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
15
|
Nishimura A, Ogata S, Tang X, Hengphasatporn K, Umezawa K, Sanbo M, Hirabayashi M, Kato Y, Ibuki Y, Kumagai Y, Kobayashi K, Kanda Y, Urano Y, Shigeta Y, Akaike T, Nishida M. Polysulfur-based bulking of dynamin-related protein 1 prevents ischemic sulfide catabolism and heart failure in mice. Nat Commun 2025; 16:276. [PMID: 39747092 PMCID: PMC11695708 DOI: 10.1038/s41467-024-55661-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 12/18/2024] [Indexed: 01/04/2025] Open
Abstract
The presence of redox-active molecules containing catenated sulfur atoms (supersulfides) in living organisms has led to a review of the concepts of redox biology and its translational strategy. Glutathione (GSH) is the body's primary detoxifier and antioxidant, and its oxidized form (GSSG) has been considered as a marker of oxidative status. However, we report that GSSG, but not reduced GSH, prevents ischemic supersulfide catabolism-associated heart failure in male mice by electrophilic modification of dynamin-related protein (Drp1). In healthy exercised hearts, the redox-sensitive Cys644 of Drp1 is highly S-glutathionylated. Nearly 40% of Cys644 is normally polysulfidated, which is a preferential target for GSSG-mediated S-glutathionylation. Cys644 S-glutathionylation is resistant to Drp1 depolysulfidation-dependent mitochondrial hyperfission and myocardial dysfunction caused by hypoxic stress. MD simulation of Drp1 structure and site-directed mutagenetic analysis reveal a functional interaction between Cys644 and a critical phosphorylation site Ser637, through Glu640. Bulky modification at Cys644 via polysulfidation or S-glutathionylation reduces Drp1 activity by disrupting Ser637-Glu640-Cys644 interaction. Disruption of Cys644 S-glutathionylation nullifies the cardioprotective effect of GSSG against heart failure after myocardial infarction. Our findings suggest a therapeutic potential of supersulfide-based Cys bulking on Drp1 for ischemic heart disease.
Collapse
Grants
- 20348438 MEXT | JST | Core Research for Evolutional Science and Technology (CREST)
- 20348438 MEXT | JST | Core Research for Evolutional Science and Technology (CREST)
- 20348438 MEXT | JST | Core Research for Evolutional Science and Technology (CREST)
- 22H02772 MEXT | Japan Society for the Promotion of Science (JSPS)
- 22K19395 MEXT | Japan Society for the Promotion of Science (JSPS)
- 24K02869 MEXT | Japan Society for the Promotion of Science (JSPS)
- 23K28237 MEXT | Japan Society for the Promotion of Science (JSPS)
- 18H05277 MEXT | Japan Society for the Promotion of Science (JSPS)
- 22K19397 MEXT | Japan Society for the Promotion of Science (JSPS)
- 21H05269 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 21H05263 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 21H05258 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- 23K20040 Ministry of Education, Culture, Sports, Science and Technology (MEXT)
- JP15km0908001 Japan Agency for Medical Research and Development (AMED)
- JP15km0908001 Japan Agency for Medical Research and Development (AMED)
- Naito Foundation
- Smoking Research Foundation (SRF)
- Sumitomo Foundation
- MEXT | JST | Exploratory Research for Advanced Technology (ERATO)
Collapse
Affiliation(s)
- Akiyuki Nishimura
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, Japan
- Exploratory Research Center on Life and Living Systems, NINS, Okazaki, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan
| | - Seiryo Ogata
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Xiaokang Tang
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, Japan
- Exploratory Research Center on Life and Living Systems, NINS, Okazaki, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan
| | | | - Keitaro Umezawa
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Makoto Sanbo
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, Japan
| | - Masumi Hirabayashi
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, Japan
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuko Ibuki
- Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, Japan
| | - Yoshito Kumagai
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenta Kobayashi
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences (NIHS), Kanagawa, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
- Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuteru Shigeta
- Center for Computational Sciences, University of Tsukuba, Tsukuba, Japan
| | - Takaaki Akaike
- Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Motohiro Nishida
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, Japan.
- Exploratory Research Center on Life and Living Systems, NINS, Okazaki, Japan.
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki, Japan.
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
16
|
Yin J, Lees JG, Gong S, Nguyen JT, Phang RJ, Shi Q, Huang Y, Kong AM, Dyson JM, Lim SY, Cheng W. Real-time electro-mechanical profiling of dynamically beating human cardiac organoids by coupling resistive skins with microelectrode arrays. Biosens Bioelectron 2025; 267:116752. [PMID: 39276439 DOI: 10.1016/j.bios.2024.116752] [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: 06/05/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/17/2024]
Abstract
Cardiac organoids differentiated from induced pluripotent stem cells are emerging as a promising platform for pre-clinical drug screening, assessing cardiotoxicity, and disease modelling. However, it is challenging to simultaneously measure mechanical contractile forces and electrophysiological signals of cardiac organoids in real-time and in-situ with the existing methods. Here, we present a biting-inspired sensory system based on a resistive skin sensor and a microelectrode array. The bite-like contact can be established with a micromanipulator to precisely position the resistive skin sensor on the top of the cardiac organoid while the 3D microneedle electrode array probes from underneath. Such reliable contact is key to achieving simultaneous electro-mechanical measurements. We demonstrate the use of our system for modelling cardiotoxicity with the anti-cancer drug doxorubicin. The electro-mechanical parameters described here elucidate the acute cardiotoxic effects induced by doxorubicin. This integrated electro-mechanical system enables a suite of new diagnostic options for assessing cardiac organoids and tissues.
Collapse
Affiliation(s)
- Jialiang Yin
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Jarmon G Lees
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia; O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia
| | - Shu Gong
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - John Tan Nguyen
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Ren Jie Phang
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia
| | - Qianqian Shi
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Yifeng Huang
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia
| | - Anne M Kong
- O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia
| | - Jennifer M Dyson
- Department of Biochemistry & Molecular Biology, Biomedicine Discovery Institute, Clayton, Victoria, 3800, Australia; Faculty of Engineering, Monash Institute of Medical Engineering (MIME), Monash University, Clayton, Victoria, 3800, Australia
| | - Shiang Y Lim
- Department of Medicine and Surgery, University of Melbourne, VIC, Australia; O'Brien Institute Department, St. Vincent's Institute of Medical Research, VIC, Australia; Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Victoria, Monash University, Australia; National Heart Research Institute Singapore, National Heart Centre, Singapore
| | - Wenlong Cheng
- Department of Chemical & Biological Engineering, Monash University, Clayton, Victoria, 3800, Australia; The Melbourne Centre for Nanofabrication, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
17
|
Marimon X, Esquinas F, Ferrer M, Cerrolaza M, Portela A, Benítez R. A Novel non-invasive optical framework for simultaneous analysis of contractility and calcium in single-cell cardiomyocytes. J Mech Behav Biomed Mater 2025; 161:106812. [PMID: 39566161 DOI: 10.1016/j.jmbbm.2024.106812] [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: 08/05/2024] [Revised: 10/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
The use of a video method based on the Digital Image Correlation (DIC) algorithm from experimental mechanics to estimate the displacements, strain field, and sarcolemma length in a beating single-cell cardiomyocyte is proposed in this work. The obtained deformation is then correlated with the calcium signal, from calcium imaging where fluorescent dyes sensitive to calcium Ca2+ are used. Our proposed video-based method for simultaneous contraction and intracellular calcium analysis results in a low-cost, non-invasive, and label-free method. This technique has shown great advantages in long-term observations because this type of intervention-free measurement neutralizes the possible alteration in the beating cardiomyocyte introduced by other techniques for measuring cell contractility (e.g., Traction Force Microscopy, Atomic Force Microscopy, Microfabrication or Optical tweezers). Three tests were performed with synthetically augmented data from cardiomyocyte images to validate the robustness of the algorithm. First, a simulated rigid translation of a referenced image is applied, then a rotation, and finally a controlled longitudinal deformation of the referenced image, thus simulating a native realistic deformation. Finally, the proposed framework is evaluated with real experimental data. To validate contraction induced by intracellular calcium concentration, this signal is correlated with a new deformation measure proposed in this article, which is independent of cell orientation in the imaging setup. Finally, based on the displacements obtained by the DIC algorithm, the change in sarcolemma length in a contracting cardiomyocyte is calculated and its temporal correlation with the calcium signal is obtained.
Collapse
Affiliation(s)
- Xavier Marimon
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain; Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| | - Ferran Esquinas
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miquel Ferrer
- Department of Strength of Materials and Structural Engineering, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miguel Cerrolaza
- School of Engineering, Science and Technology, Valencian International University (VIU), Valencia, Spain
| | - Alejandro Portela
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Raúl Benítez
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain
| |
Collapse
|
18
|
Huang Y, Zhang Y, Wang Z, Miao L, Tan P, Guan Y, Ran Y, Feng X, Wang Y, Guo Y, Guo X. Modified mRNA-based gene editing reveals sarcomere-based regulation of gene expression in human induced-pluripotent stem cell-derived cardiomyocytes. Int Immunopharmacol 2024; 143:113378. [PMID: 39423657 DOI: 10.1016/j.intimp.2024.113378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024]
Abstract
Mutations in genes coding sarcomere components are the major causes of human inherited cardiomyopathy. Genome editing is widely applied to genetic modification of human pluripotent stem cells (hPSCs) before hPSCs were differentiated into cardiomyocytes to model cardiomyopathy. Whether genetic mutations influence the early hPSC differentiation process or solely the terminally differentiated cardiomyocytes during cardiac pathogenesis remains challenging to distinguish. To solve this problem, here we harnessed chemically modified mRNA (modRNA) and synthetic single-guide RNA to develop an efficient genome editing approach in hPSC-derived cardiomyocytes (hPSC-CMs). We showed that modRNA-based CRISPR/Cas9 mutagenesis of TNNT2, the coding gene for cardiac troponin T, results in sarcomere disassembly and contractile dysfunction in hPSC-CMs. These structural and functional phenotypes were associated with profound downregulation of oxidative phosphorylation genes and upregulation of cardiac stress markers NPPA and NPPB. These data confirmed that sarcomeres regulate gene expression in hPSC-CMs and highlighted the RNA technology as a powerful tool to achieve stage-specific genome editing during hPSC differentiation.
Collapse
Affiliation(s)
- Yuqing Huang
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yueyang Zhang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Ze Wang
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China
| | - Lei Miao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Beijing Key Laboratory of Molecular Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Pingping Tan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Guan
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqing Ran
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xing Feng
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijia Wang
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxuan Guo
- School of Basic Medical Sciences, Institute of Cardiovascular Sciences, Peking University Health Science Center, Beijing, China; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China; Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China.
| | - Xiaoling Guo
- Basic Medical Research Center, the Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
19
|
Horiuchi S, Koda N, Ikeda Y, Tanaka Y, Masuo Y, Kato Y, Yamazaki D. Examination of common culture medium for human hepatocytes and engineered heart tissue: Towards an evaluation of cardiotoxicity associated with hepatic drug metabolism in vitro. PLoS One 2024; 19:e0315997. [PMID: 39715174 DOI: 10.1371/journal.pone.0315997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
Cardiotoxicity associated with hepatic metabolism and drug-drug interactions is a serious concern. Predicting drug toxicity using animals remains challenging due to species and ethical concerns, necessitating the need to develop alternative approaches. Drug cardiotoxicity associated with hepatic metabolism cannot be detected using a cardiomyocyte-only evaluation system. Therefore, we aimed to establish a system for evaluating cardiotoxicity via hepatic metabolism by co-culturing cryopreserved human hepatocytes (cryoheps) and human iPS cell-derived engineered heart tissues (hiPSC-EHTs) using a stirrer-based microphysiological system. We investigated candidate media to identify a medium that can be used commonly for hepatocytes and cardiomyocytes. We found that the contraction length was significantly greater in the HM Dex (-) medium, the medium used for cryohep culture without dexamethasone, than that in the EHT medium used for hiPSC-EHT culture. Additionally, the beating rate, contraction length, contraction speed, and relaxation speed of hiPSC-EHT cultured in the HM Dex (-) medium were stable throughout the culture period. Among the major CYPs, the expression of CYP3A4 alone was low in cryoheps cultured in the HM Dex (-) medium. However, improved oxygenation using the InnoCell plate increased CYP3A4 expression to levels comparable to those found in the human liver. In addition, CYP3A4 activity was also increased by the improved oxygenation. Furthermore, expression levels of hepatic function-related gene and nuclear receptors in cryoheps cultured in HM Dex (-) medium were comparable to those in the human liver. These results suggest that the HM Dex (-) medium can be applied to co-culture and may allow the evaluation of cardiotoxicity via hepatic metabolism. Moreover, CYP induction by typical inducers was confirmed in cryoheps cultured in the HM Dex (-) medium, suggesting that drug-drug interactions could also be evaluated using this medium. Our findings may facilitate the evaluation of cardiotoxicity via hepatic metabolism, potentially reducing animal testing, lowering costs, and expediting drug development.
Collapse
Affiliation(s)
- Shinichiro Horiuchi
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Nanae Koda
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Yui Ikeda
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuto Tanaka
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Daiju Yamazaki
- Division of Pharmacology, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| |
Collapse
|
20
|
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
|
21
|
Pavel MA, Chen H, Hill M, Sridhar A, Barney M, DeSantiago J, Owais A, Sandu S, Darbar FA, Ornelas-Loredo A, Al-Azzam B, Chalazan B, Rehman J, Darbar D. A Titin Missense Variant Causes Atrial Fibrillation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.06.24318402. [PMID: 39677424 PMCID: PMC11643245 DOI: 10.1101/2024.12.06.24318402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Rare and common genetic variants contribute to the risk of atrial fibrillation (AF). Although ion channels were among the first AF candidate genes identified, rare loss-of-function variants in structural genes such as TTN have also been implicated in AF pathogenesis partly by the development of an atrial myopathy, but the underlying mechanisms are poorly understood. While TTN truncating variants (TTNtvs) have been causally linked to arrhythmia and cardiomyopathy syndromes, the role of missense variants (mvs) remains unclear. We report that rare TTNmvs are associated with adverse clinical outcomes in AF patients and we have identified a mechanism by which a TTNmv (T32756I) causes AF. Modeling the TTN-T32756I variant using human induced pluripotent stem cell-derived atrial cardiomyocytes (iPSC-aCMs) revealed that the mutant cells display aberrant contractility, increased activity of a cardiac potassium channel (KCNQ1, Kv7.1), and dysregulated calcium homeostasis without compromising the sarcomeric integrity of the atrial cardiomyocytes. We also show that a titin-binding protein, the Four-and-a-Half Lim domains 2 (FHL2), has increased binding with KCNQ1 and its modulatory subunit KCNE1 in the TTN-T32756I-iPSC-aCMs, enhancing the slow delayed rectifier potassium current (I ks). Suppression of FHL2 in mutant iPSC-aCMs normalized the I ks, supporting FHL2 as an I ks modulator. Our findings demonstrate that a single amino acid change in titin not only affects function but also causes ion channel remodeling and AF. These findings emphasize the need for high-throughput screening to evaluate the pathogenicity of TTNmvs and establish a mechanistic link between titin, potassium ion channels, and sarcomeric proteins that may represent a novel therapeutic target.
Collapse
Affiliation(s)
- Mahmud Arif Pavel
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Hanna Chen
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Michael Hill
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Arvind Sridhar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Miles Barney
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Jaime DeSantiago
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Asia Owais
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Shashank Sandu
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Faisal A. Darbar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Aylin Ornelas-Loredo
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Bahaa Al-Azzam
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Brandon Chalazan
- Division of Genetics, Genomics, and Metabolism, Department of Pediatrics, Lurie Children’s Hospital of Chicago, Chicago, IL, USA
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, University of Illinois Chicago, Chicago, IL, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL, USA
- Department of Pharmacology, University of Illinois Chicago, Chicago, IL, USA
- Jesse Brown Veterans Administration Medical Center, Chicago, IL, USA
| |
Collapse
|
22
|
O'Hern C, Caywood S, Aminova S, Kiselev A, Volmert B, Wang F, Sewavi ML, Cao W, Dionise M, Muniyandi P, Popa M, Basrai H, Skoric M, Boulos G, Huang A, Nuñez-Regueiro I, Chalfoun N, Park S, Ashammakhi N, Zhou C, Contag C, Aguirre A. Human heart assembloids with autologous tissue-resident macrophages recreate physiological immuno-cardiac interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.13.623051. [PMID: 39677610 PMCID: PMC11642760 DOI: 10.1101/2024.11.13.623051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Interactions between the developing heart and the embryonic immune system are essential for proper cardiac development and maintaining homeostasis, with disruptions linked to various diseases. While human pluripotent stem cell (hPSC)-derived organoids are valuable models for studying human organ function, they often lack critical tissue-resident immune cells. Here, we introduce an advanced human heart assembloid model, termed hHMA (human heart-macrophage assembloid), which fully integrates autologous cardiac tissue- resident macrophages (MPs) with pre-existing human heart organoids (hHOs). Through multi-omic analyses, we confirmed that these MPs are phenotypically similar to embryonic cardiac tissue-resident MPs and remain viable in the assembloids over time. The inclusion of MPs significantly impacts hHMA development, influencing cardiac cellular composition, boosting cellular communication, remodeling the extracellular matrix, promoting ventricular morphogenesis, and enhancing sarcomeric maturation. Our findings indicate that MPs contribute to homeostasis via efferocytosis, integrate into the cardiomyocyte electrical system, and support catabolic metabolism. To demonstrate the versatility of this model, we developed a platform to study cardiac arrhythmias by chronic exposure to pro-inflammatory factors linked to arrhythmogenesis in clinical settings, successfully replicating key features of inflammasome-mediated atrial fibrillation. Overall, this work introduces a robust platform for examining the role of immune cells in cardiac development, disease mechanisms, and drug discovery, bridging the gap between in vitro models and human physiology. These findings offer insights into cardiogenesis and inflammation-driven heart disease, positioning the hHMA system as an invaluable tool for future cardiovascular research and therapeutic development.
Collapse
|
23
|
Ireland J, Kilian KA. The importance of matrix in cardiomyogenesis: Defined substrates for maturation and chamber specificity. Matrix Biol Plus 2024; 24:100160. [PMID: 39291079 PMCID: PMC11403269 DOI: 10.1016/j.mbplus.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CM) are a promising source of cardiac cells for disease modelling and regenerative medicine. However, current protocols invariably lead to mixed population of cardiac cell types and often generate cells that resemble embryonic phenotypes. Here we developed a combinatorial approach to assess the importance of extracellular matrix proteins (ECMP) in directing the differentiation of cardiomyocytes from human embryonic stem cells (hESC). We did this by focusing on combinations of ECMP commonly found in the developing heart with a broad goal of identifying combinations that promote maturation and influence chamber specific differentiation. We formulated 63 unique ECMP combinations fabricated from collagen 1, collagen 3, collagen 4, fibronectin, laminin, and vitronectin, presented alone and in combinations, leading to the identification of specific ECMP combinations that promote hESC proliferation, pluripotency, and germ layer specification. When hESC were subjected to a differentiation protocol on the ECMP combinations, it revealed precise protein combinations that enhance differentiation as determined by the expression of cardiac progenitor markers kinase insert domain receptor (KDR) and mesoderm posterior transcription factor 1 (MESP1). High expression of cardiac troponin (cTnT) and the relative expression of myosin light chain isoforms (MLC2a and MLC2v) led to the identification of three surfaces that promote a mature cardiomyocyte phenotype. Action potential morphology was used to assess chamber specificity, which led to the identification of matrices that promote chamber-specific cardiomyocytes. This study provides a matrix-based approach to improve control over cardiomyocyte phenotypes during differentiation, with the scope for translation to cardiac laboratory models and for the generation of functional chamber specific cardiomyocytes for regenerative therapies.
Collapse
Affiliation(s)
- Jake Ireland
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kristopher A Kilian
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
- School of Materials Science and Engineering, UNSW Sydney, Sydney, New South Wales, Australia
- Australian Centre for NanoMedicine, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
24
|
Maione AS, Iengo L, Sala L, Massaiu I, Chiesa M, Lippi M, Ghilardi S, Florindi C, Lodola F, Zaza A, Tondo C, Schiavone M, Banfi C, Pompilio G, Poggio P, Sommariva E. Cardiomyocyte and stromal cell cross-talk influences the pathogenesis of arrhythmogenic cardiomyopathy: a multi-level analysis uncovers DLK1-NOTCH pathway role in fibro-adipose remodelling. Cell Death Discov 2024; 10:484. [PMID: 39609399 PMCID: PMC11604953 DOI: 10.1038/s41420-024-02232-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Arrhythmogenic Cardiomyopathy (ACM) is a life-threatening, genetically determined disease primarily caused by mutations in desmosomal genes, such as PKP2. Currently, there is no etiological therapy for ACM due to its complex and not fully elucidated pathogenesis. Various cardiac cell types affected by the genetic mutation, such as cardiomyocytes (CM) and cardiac mesenchymal stromal cells (cMSC), individually contribute to the ACM phenotype, driving functional abnormalities and fibro-fatty substitution, respectively. However, the relative importance of the CM and cMSC alterations, as well as their reciprocal influence in disease progression remain poorly understood. We hypothesised that ACM-dependent phenotypes are driven not only by alterations in individual cell types but also by the reciprocal interactions between CM and cMSC, which may further impact disease pathogenesis. We utilized a patient-specific, multicellular cardiac system composed of either control or PKP2-mutated CM and cMSC to assess the mutation's role in fibro-fatty phenotype by immunofluorescence, and contractile behaviour of co-cultures using cell motion detection software. Additionally, we investigated reciprocal interactions both in silico and via multi-targeted proteomics. We demonstrated that ACM CM can promote fibro-adipose differentiation of cMSC. Conversely, ACM cMSC contribute to increasing the rate of abnormal contractile events with likely arrhythmic significance. Furthermore, we showed that an ACM-causative mutation alters the CM-cMSC interaction pattern. We identified the CM-sourced DLK1 as a novel regulator of fibro-adipose remodelling in ACM. Our study challenges the paradigm of exclusive cell-specific mechanisms in ACM. A deeper understanding of the cell-cell influence is crucial for identifying novel therapeutic targets for ACM, and this concept is exploitable for other cardiomyopathies.
Collapse
Affiliation(s)
- Angela Serena Maione
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy.
| | - Lara Iengo
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Luca Sala
- Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Istituto Auxologico Italiano IRCCS, 20095, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, 20126, Italy
| | - Ilaria Massaiu
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Mattia Chiesa
- Bioinformatics and Artificial Intelligence Facility, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Electronics, Information and Biomedical Engineering, Politecnico di Milano, 20133, Milan, Italy
| | - Melania Lippi
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Stefania Ghilardi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Chiara Florindi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, 20126, Italy
| | - Francesco Lodola
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, 20126, Italy
| | - Antonio Zaza
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, 20126, Italy
| | - Claudio Tondo
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122, Milan, Italy
| | - Marco Schiavone
- Department of Clinical Electrophysiology and Cardiac Pacing, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics, and Network Analysis, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122, Milan, Italy
| | - Paolo Poggio
- Unit for the Study of Aortic, Valvular and Coronary Pathologies, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, 20122, Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| |
Collapse
|
25
|
Zhang T, Yang S, Ge Y, Yin L, Pu Y, Gu Z, Chen Z, Liang G. Unveiling the Heart's Hidden Enemy: Dynamic Insights into Polystyrene Nanoplastic-Induced Cardiotoxicity Based on Cardiac Organoid-on-a-Chip. ACS NANO 2024; 18:31569-31585. [PMID: 39482939 DOI: 10.1021/acsnano.4c13262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Exposure to micro- and nanoplastics (MNPs) has been implicated in potential cardiotoxicity. However, in vitro models based on cardiomyocyte cell lines lack crucial cardiac characteristics, while interspecies differences in animal models compromise the reliability of the conclusions. In addition, current research has predominantly focused on single-time point exposures to MNPs, neglecting comparative analyses of cardiac injury across early and late stages. Moreover, there remains a large gap in understanding the susceptibility to MNPs under pathological conditions. To address these limitations, this study integrated cardiac organoids (COs) and organ-on-a-chip (OoC) technology to develop the cardiac organoid-on-a-chip (COoC), which was validated for cardiotoxicity evaluation through multiple dimensions. Based on COoC, we conducted a dynamic observation of the cardiac damage caused by short- and long-term exposure to polystyrene nanoplastics (PS-NPs). Oxidative stress, inflammation, disruption of calcium ion homeostasis, and mitochondrial dysfunction were confirmed as the potential mechanisms of PS-NP-induced cardiotoxicity and the crucial events in the early stages, while cardiac fibrosis emerged as a prominent feature in late stages. Notably, low-dose exposure exacerbated myocardial infarction symptoms under pathological states, despite no significant cardiotoxicity shown in healthy models. In conclusion, these findings further deepened our understanding of PS-NP-induced cardiotoxic effects and introduced a promising in vitro platform for assessing cardiotoxicity.
Collapse
Affiliation(s)
- Tianyi Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Yiling Ge
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, China
| |
Collapse
|
26
|
Maihemuti W, Murata K, Abulaiti M, Minatoya K, Masumoto H. Simultaneous electro-dynamic stimulation accelerates maturation of engineered cardiac tissues generated by human iPS cells. Biochem Biophys Res Commun 2024; 733:150605. [PMID: 39197194 DOI: 10.1016/j.bbrc.2024.150605] [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: 08/05/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Electrical and dynamic stimulation are commonly employed to enhance the maturation of engineered cardiac tissue (ECT) derived from human induced pluripotent stem cells (iPSCs), reflecting the physiological environment of the heart. While electrical stimulation mimics natural bioelectrical signals and dynamic stimulation replicates mechanical forces, the combined effects of these stimuli on ECT maturation have not been thoroughly explored. We hypothesized that simultaneous electro-dynamic stimulation would enhance ECT maturation and function more effectively than either stimulus alone. Human iPSC-derived cardiovascular cells were co-cultured with Collagen I and Matrigel for 2 weeks, followed by a comparative analysis of four groups: no stimulation, dynamic stimulation, electrical stimulation, and simultaneous electro-dynamic stimulation. The functionality of ECTs was assessed by measuring contractile capacity and calcium indicators, and histological assessments examined structural maturation. Our results demonstrated that simultaneous electro-dynamic stimulation significantly increased the CM component, elevated TNNT2 mRNA expression levels, and enhanced calcium transient capacity. Additionally, ECTs subjected to simultaneous stimulation exhibited a positive force-frequency relationship in contractility and an elevation in peak calcium flux, indicative of advanced tissue maturation. Moreover, simultaneous stimulation promoted vascular network formation within the ECTs, suggesting improved structural organization. These findings underscore the importance of simultaneous stimulation for developing effective cardiac tissue engineering strategies.
Collapse
Affiliation(s)
- Wusiman Maihemuti
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kozue Murata
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mosha Abulaiti
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidetoshi Masumoto
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan; Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
27
|
Rivera ES, LeBrun ES, Breidenbach JD, Solomon E, Sanders CK, Harvey T, Tseng CY, Thornhill MG, Blackwell BR, McBride EM, Luchini KA, Alvarez M, Williams RF, Norris JL, Mach PM, Glaros TG. Feature-agnostic metabolomics for determining effective subcytotoxic doses of common pesticides in human cells. Toxicol Sci 2024; 202:85-95. [PMID: 39110521 DOI: 10.1093/toxsci/kfae101] [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: 10/29/2024] Open
Abstract
Although classical molecular biology assays can provide a measure of cellular response to chemical challenges, they rely on a single biological phenomenon to infer a broader measure of cellular metabolic response. These methods do not always afford the necessary sensitivity to answer questions of subcytotoxic effects, nor do they work for all cell types. Likewise, boutique assays such as cardiomyocyte beat rate may indirectly measure cellular metabolic response, but they too, are limited to measuring a specific biological phenomenon and are often limited to a single cell type. For these reasons, toxicological researchers need new approaches to determine metabolic changes across various doses in differing cell types, especially within the low-dose regime. The data collected herein demonstrate that LC-MS/MS-based untargeted metabolomics with a feature-agnostic view of the data, combined with a suite of statistical methods including an adapted environmental threshold analysis, provides a versatile, robust, and holistic approach to directly monitoring the overall cellular metabolomic response to pesticides. When employing this method in investigating two different cell types, human cardiomyocytes and neurons, this approach revealed separate subcytotoxic metabolomic responses at doses of 0.1 and 1 µM of chlorpyrifos and carbaryl. These findings suggest that this agnostic approach to untargeted metabolomics can provide a new tool for determining effective dose by metabolomics of chemical challenges, such as pesticides, in a direct measurement of metabolomic response that is not cell type-specific or observable using traditional assays.
Collapse
Affiliation(s)
- Emilio S Rivera
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Erick S LeBrun
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Joshua D Breidenbach
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Emilia Solomon
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Claire K Sanders
- Microbial and Biome Sciences Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, United States
| | - Tara Harvey
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Chi Yen Tseng
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - M Grace Thornhill
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Brett R Blackwell
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Ethan M McBride
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Kes A Luchini
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Marc Alvarez
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Robert F Williams
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Jeremy L Norris
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37235, United States
| | - Phillip M Mach
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| | - Trevor G Glaros
- Biochemistry and Biotechnology Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 84545, United States
| |
Collapse
|
28
|
Dutton LC, Dudhia J, Guest DJ, Connolly DJ. CRISPR/Cas9 gene editing in induced pluripotent stem cells to investigate the feline hypertrophic cardiomyopathy causing MYBPC3/R820W mutation. PLoS One 2024; 19:e0311761. [PMID: 39388496 PMCID: PMC11466433 DOI: 10.1371/journal.pone.0311761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common heart disease in domestic cats, often leading to congestive heart failure and death, with current treatment strategies unable to reverse or prevent progression of the disease. The underlying pathological processes driving HCM remain unclear, which hinders novel drug discovery. The aim of this study was to generate a cellular model of the feline HCM-causing MYBPC3 mutation R820W. Using CRISPR/Cas9 gene editing we introduced the R820W mutation into a human induced pluripotent stem cell (iPSC) line. We differentiated both homozygous mutant clones and isogenic control clones to cardiomyocytes (iPSC-CMs). Protein quantification indicated that haploinsufficiency is not the disease mechanism of the mutation. Homozygous mutant iPSC-CMs had a larger cell area than isogenic controls, with the sarcomere structure and incorporation of cMyBP-C appearing similar between mutant and control iPSC-CMs. Contraction kinetic analysis indicated that homozygous iPSC-CMs have impaired relaxation and are hypocontractile compared to isogenic control iPSC-CMs. In summary, we demonstrate successful generation of an iPSC model of a feline MYBPC3 mutation, with the cellular model recapitulating aspects of HCM including cellular hypertrophy and impaired relaxation kinetics. We anticipate that further study of this model will lead to improved understanding of the disease-causing molecular mechanism, ultimately leading to novel drug discovery.
Collapse
Affiliation(s)
- Luke C. Dutton
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, London, United Kingdom
| | - Jayesh Dudhia
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, London, United Kingdom
| | - Deborah J. Guest
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, London, United Kingdom
| | - David J. Connolly
- Department of Clinical Science and Services, Royal Veterinary College, Hatfield, London, United Kingdom
| |
Collapse
|
29
|
Vesga-Castro C, Mosqueira-Martín L, Ubiria-Urkola P, Marco-Moreno P, González-Imaz K, Rendon-Hinestroza J, Vallejo-Illarramendi A, Paredes J. Development of an in vitro platform for the analysis of contractile and calcium dynamics in single human myotubes. LAB ON A CHIP 2024; 24:4741-4754. [PMID: 39264341 DOI: 10.1039/d3lc00442b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
In vitro myotube cultures are widely used as models for studying muscle pathophysiology, but their limited maturation and heterogeneity pose significant challenges for functional analyses. While they remain the gold standard for studying muscle function in vitro, myotube cultures do not fully recapitulate the complexity and native features of muscle fibers, which may compromise their ability to predict in vivo outcomes. To promote maturation and decrease heterogeneity, we have incorporated engineered structures into myotube cultures, based on a PDMS thin layer with micrometer-sized grooves (μGrooves) placed over a glass substrate. Different sizes and shapes of μGrooves were tested for their ability to promote alignment and fusion of myoblasts and enhance their differentiation into myotubes. A 24 hour electrical field stimulation protocol (4 V, 6 ms, 0.1 Hz) was used to further promote myotube maturation, after which several myotube features were assessed, including myotube alignment, width, fusion index, contractile function, and calcium handling. Our results indicate superior calcium and contractile performance in μGrooved myotubes, particularly with the 100 μm-width 700 μm-long geometry (7 : 1). This platform generated homogeneous and isolated myotubes that reproduced native muscle features, such as excitation-contraction coupling and force-frequency responses. Overall, our 2D muscle platform enables robust high-content assays of calcium dynamics and contractile readouts with increased sensitivity and reproducibility compared to traditional myotube cultures, making it particularly suitable for screening therapeutic candidates for different muscle pathologies.
Collapse
Affiliation(s)
- Camila Vesga-Castro
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| | - Laura Mosqueira-Martín
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Paul Ubiria-Urkola
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| | - Pablo Marco-Moreno
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Klaudia González-Imaz
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Jorge Rendon-Hinestroza
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Jacobo Paredes
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| |
Collapse
|
30
|
Shead KD, Huethorst E, Burton F, Lang NN, Myles RC, Smith GL. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes for Preclinical Cardiotoxicity Screening in Cardio-Oncology. JACC CardioOncol 2024; 6:678-683. [PMID: 39479327 PMCID: PMC11520213 DOI: 10.1016/j.jaccao.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/27/2024] [Accepted: 07/25/2024] [Indexed: 11/02/2024] Open
Abstract
•hiPSC-CM offer an alternative to in vivo models for predicting cardiotoxicity. •hiPSC-CM monolayers detect pro-arrhythmic effects; inotropic detection is less established. •Cardiac spheroids and engineered tissue may suit chronic cardiotoxicity studies (>2 weeks). •Cardiac assays with non-myocyte cells may be key to identifying some cardiotoxicity forms. •hiPSC-CM technologies are well placed to develop patient-specific assays in the future.
Collapse
Affiliation(s)
- Kyle D. Shead
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Eline Huethorst
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Francis Burton
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Ninian N. Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Rachel C. Myles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Godfrey L. Smith
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
31
|
Salmenov R, Mummery C, ter Huurne M. Cell cycle visualization tools to study cardiomyocyte proliferation in real-time. Open Biol 2024; 14:240167. [PMID: 39378987 PMCID: PMC11461051 DOI: 10.1098/rsob.240167] [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: 06/19/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/10/2024] Open
Abstract
Cardiomyocytes in the adult human heart are quiescent and those lost following heart injury are not replaced by proliferating survivors. Considerable effort has been made to understand the mechanisms underlying cardiomyocyte cell cycle exit and re-entry, with view to discovering therapeutics that could stimulate cardiomyocyte proliferation and heart regeneration. The advent of large compound libraries and robotic liquid handling platforms has enabled the screening of thousands of conditions in a single experiment but success of these screens depends on the appropriateness and quality of the model used. Quantification of (human) cardiomyocyte proliferation in high throughput has remained problematic because conventional antibody-based staining is costly, technically challenging and does not discriminate between cardiomyocyte division and failure in karyokinesis or cytokinesis. Live cell imaging has provided alternatives that facilitate high-throughput screening but these have other limitations. Here, we (i) review the cell cycle features of cardiomyocytes, (ii) discuss various cell cycle fluorescent reporter systems, and (iii) speculate on what could improve their predictive value in the context of cardiomyocyte proliferation. Finally, we consider how these new methods can be used in combination with state-of-the-art three-dimensional human cardiac organoid platforms to identify pro-proliferative signalling pathways that could stimulate regeneration of the human heart.
Collapse
Affiliation(s)
- Rustem Salmenov
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden2300RC, The Netherlands
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden2300RC, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden2300RC, The Netherlands
| | - Menno ter Huurne
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden2300RC, The Netherlands
| |
Collapse
|
32
|
Iwanski JB, Pappas CT, Mayfield RM, Farman GP, Ahrens-Nicklas R, Churko JM, Gregorio CC. Leiomodin 2 neonatal dilated cardiomyopathy mutation results in altered actin gene signatures and cardiomyocyte dysfunction. NPJ Regen Med 2024; 9:21. [PMID: 39285234 PMCID: PMC11405699 DOI: 10.1038/s41536-024-00366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Neonatal dilated cardiomyopathy (DCM) is a poorly understood muscular disease of the heart. Several homozygous biallelic variants in LMOD2, the gene encoding the actin-binding protein Leiomodin 2, have been identified to result in severe DCM. Collectively, LMOD2-related cardiomyopathies present with cardiac dilation and decreased heart contractility, often resulting in neonatal death. Thus, it is evident that Lmod2 is essential to normal human cardiac muscle function. This study aimed to understand the underlying pathophysiology and signaling pathways related to the first reported LMOD2 variant (c.1193 G > A, p.Trp398*). Using patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and a mouse model harboring the homologous mutation to the patient, we discovered dysregulated actin-thin filament lengths, altered contractility and calcium handling properties, as well as alterations in the serum response factor (SRF)-dependent signaling pathway. These findings reveal that LMOD2 may be regulating SRF activity in an actin-dependent manner and provide a potential new strategy for the development of biologically active molecules to target LMOD2-related cardiomyopathies.
Collapse
Grants
- R01HL123078 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R00 HL128906 NHLBI NIH HHS
- R01 HL164644 NHLBI NIH HHS
- R01 GM120137 NIGMS NIH HHS
- F30HL151139 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32HL007249 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32 HL007249 NHLBI NIH HHS
- R01 HL123078 NHLBI NIH HHS
- R01HL164644 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F30 HL151139 NHLBI NIH HHS
- R01GM120137 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Jessika B Iwanski
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Christopher T Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Rachel M Mayfield
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Gerrie P Farman
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Rebecca Ahrens-Nicklas
- Department of Pediatrics and Division of Human Genetics and Metabolism, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jared M Churko
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA.
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA.
- Department of Medicine and Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
33
|
Finsberg H, Charwat V, Healy KE, Wall ST. Automatic motion estimation with applications to hiPSC-CMs. Biomed Phys Eng Express 2024; 10:065004. [PMID: 39173648 DOI: 10.1088/2057-1976/ad7268] [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: 04/25/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are an effective tool for studying cardiac function and disease, and hold promise for screening drug effects on human tissue. Understanding alterations in motion patterns within these cells is crucial for comprehending how the administration of a drug or the onset of a disease can impact the rhythm of the human heart. However, quantifying motion accurately and efficiently from optical measurements using microscopy is currently time consuming. In this work, we present a unified framework for performing motion analysis on a sequence of microscopically obtained images of tissues consisting of hiPSC-CMs. We provide validation of our developed software using a synthetic test case and show how it can be used to extract displacements and velocities in hiPSC-CM microtissues. Finally, we show how to apply the framework to quantify the effect of an inotropic compound. The described software system is distributed as a python package that is easy to install, well tested and can be integrated into any python workflow.
Collapse
Affiliation(s)
| | | | - Kevin E Healy
- Department of Material Science and Engineering, University of California, Berkeley, CA, United States of America
- Department of Bioengineering, University of California, Berkeley, CA, United States of America
| | - Samuel T Wall
- Simula Research Laboratory, Norway
- Organos, Inc, Berkeley, CA, United States of America
| |
Collapse
|
34
|
Zhang X, Burattini M, Duru J, Chala N, Wyssen N, Cofiño-Fabres C, Rivera-Arbeláez JM, Passier R, Poulikakos D, Ferrari A, Tringides C, Vörös J, Luciani GB, Miragoli M, Zambelli T. Multimodal Mapping of Electrical and Mechanical Latency of Human-Induced Pluripotent Stem Cell-Derived Cardiomyocyte Layers. ACS NANO 2024; 18:24060-24075. [PMID: 39172696 DOI: 10.1021/acsnano.4c03896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
The synchronization of the electrical and mechanical coupling assures the physiological pump function of the heart, but life-threatening pathologies may jeopardize this equilibrium. Recently, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have emerged as a model for personalized investigation because they can recapitulate human diseased traits, such as compromised electrical capacity or mechanical circuit disruption. This research avails the model of hiPSC-CMs and showcases innovative techniques to study the electrical and mechanical properties as well as their modulation due to inherited cardiomyopathies. In this work, hiPSC-CMs carrying either Brugada syndrome (BRU) or dilated cardiomyopathy (DCM), were organized in a bilayer configuration to first validate the experimental methods and second mimic the physiological environment. High-density CMOS-based microelectrode arrays (HD-MEA) have been employed to study the electrical activity. Furthermore, mechanical function was investigated via quantitative video-based evaluation, upon stimulation with a β-adrenergic agonist. This study introduces two experimental methods. First, high-throughput mechanical measurements in the hiPSC-CM layers (xy-inspection) are obtained using both a recently developed optical tracker (OPT) and confocal reference-free traction force microscopy (cTFM) aimed to quantify cardiac kinematics. Second, atomic force microscopy (AFM) with FluidFM probes, combined with the xy-inspection methods, supplemented a three-dimensional understanding of cell-cell mechanical coupling (xyz-inspection). This particular combination represents a multi-technique approach to detecting electrical and mechanical latency among the cell layers, examining differences and possible implications following inherited cardiomyopathies. It can not only detect disease characteristics in the proposed in vitro model but also quantitatively assess its response to drugs, thereby demonstrating its feasibility as a scalable tool for clinical and pharmacological studies.
Collapse
Affiliation(s)
- Xinyu Zhang
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - Margherita Burattini
- Laboratory of Experimental and Applied Medical Technologies, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Department of Maternity, Surgery and Dentistry, University of Verona, 37134 Verona, Italy
| | - Jens Duru
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - Nafsika Chala
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zurich,Switzerland
| | - Nino Wyssen
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - Carla Cofiño-Fabres
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherland
| | - José Manuel Rivera-Arbeláez
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherland
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed Centre, University of Twente, 7500 AE Enschede, The Netherland
| | - Dimos Poulikakos
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zurich,Switzerland
| | - Aldo Ferrari
- Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zürich, 8092 Zurich,Switzerland
- Experimental Continuum Mechanics, EMPA, Swiss Federal Laboratories for Material Science and Technologies, 8600 Dübendorf, Switzerland
| | - Christina Tringides
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | - János Vörös
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| | | | - Michele Miragoli
- Laboratory of Experimental and Applied Medical Technologies, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Humanitas Research Hospital ─ IRCCS, 20089 Rozzano, Italy
| | - Tomaso Zambelli
- Laboratory of Biosensors and Bioelectronics, Institute for Biomedical Engineering, ETH Zürich, 8092 Zürich,Switzerland
| |
Collapse
|
35
|
Patel L, Roy A, Barlow J, O'Shea C, Nieves D, Azad AJ, Hall C, Davies B, Rath P, Pavlovic D, Chikermane A, Geberhiwot T, Steeds RP, Gehmlich K. Characterisation of infantile cardiomyopathy in Alström syndrome using ALMS1 knockout induced pluripotent stem cell derived cardiomyocyte model. Mol Genet Metab 2024; 143:108575. [PMID: 39243575 DOI: 10.1016/j.ymgme.2024.108575] [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: 07/17/2024] [Revised: 08/27/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alström syndrome (AS) is an inherited rare ciliopathy characterised by multi-organ dysfunction and premature cardiovascular disease. This may manifest as an infantile-onset dilated cardiomyopathy with significant associated mortality. An adult-onset restrictive cardiomyopathy may also feature later in life. Loss of function pathogenic variants in ALMS1 have been identified in AS patients, leading to a lack of ALMS1 protein. The biological role of ALMS1 is unknown, particularly in a cardiovascular context. To understand the role of ALMS1 in infantile cardiomyopathy, the reduction of ALMS1 protein seen in AS patients was modelled using human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), in which ALMS1 was knocked out. MuscleMotion analysis and calcium optical mapping experiments suggest that ALMS1 knockout (KO) cells have increased contractility, with altered calcium extrusion and impaired calcium handling dynamics compared to wildtype (WT) counterparts. Seahorse metabolic assays showed ALMS1 knockout iPSC-CMs had increased glycolytic and mitochondrial respiration rates, with ALMS1 knockout cells portraying increased energetic demand and respiratory capacity than WT counterparts. Using senescence associated β-galactosidase (SA-β gal) staining assay, we identified increased senescence of ALMS1 knockout iPSC-CMs. Overall, this study provides insights into the molecular mechanisms in AS, particularly the role of ALMS1 in infantile cardiomyopathy in AS, using iPSC-CMs as a 'disease in a dish' model to provide insights into multiple aspects of this complex disease.
Collapse
Affiliation(s)
- Leena Patel
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK.
| | - Ashwin Roy
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK; Department of Cardiology, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Jonathan Barlow
- Cellular Health and Metabolism Facility, School of Sport, Exercise and Rehabilitation, University of Birmingham, Birmingham, UK
| | - Christopher O'Shea
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Daniel Nieves
- Institute of Immunology and Immunotherapy, University of Birmingham, UK
| | - Amar J Azad
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK; Center of Biological Design, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Caitlin Hall
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Ben Davies
- Genetic Modification Service, The Francis Crick Institute, London, UK; Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Phalguni Rath
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Ashish Chikermane
- Paediatric Cardiology, Birmingham Children's Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Tarekegn Geberhiwot
- Department of Inherited Metabolic Diseases, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK; Institute of Metabolism and System Research, University of Birmingham, Birmingham, UK
| | - Richard P Steeds
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK; Department of Cardiology, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham, Birmingham, UK
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK; Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK.
| |
Collapse
|
36
|
Pachter N, Allen K, Hookway TA. Exogenous ECM in an environmentally-mediated in vitro model for cardiac fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608840. [PMID: 39229021 PMCID: PMC11370619 DOI: 10.1101/2024.08.20.608840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Few clinical solutions exist for cardiac fibrosis, creating the need for a tunable in vitro model to better understand fibrotic disease mechanisms and screen potential therapeutic compounds. Here, we combined cardiomyocytes, cardiac fibroblasts, and exogenous extracellular matrix (ECM) proteins to create an environmentally-mediated in vitro cardiac fibrosis model. Cells and ECM were combined into 2 types of cardiac tissues- aggregates and tissue rings. The addition of collagen I had a drastic negative impact on aggregate formation, but ring formation was not as drastically affected. In both tissue types, collagen and other ECM did not severely affect contractile function. Histological analysis showed direct incorporation of collagen into tissues, indicating that we can directly modulate the cells' ECM environment. This modulation affects tissue formation and distribution of cells, indicating that this model provides a useful platform for understanding how cells respond to changes in their extracellular environment and for potential therapeutic screening.
Collapse
Affiliation(s)
- Natalie Pachter
- Department of Biomedical Engineering, Binghamton University, the State University of New York, Binghamton, NY 13902, United States
| | - Kristen Allen
- Department of Biomedical Engineering, Binghamton University, the State University of New York, Binghamton, NY 13902, United States
| | - Tracy A Hookway
- Department of Biomedical Engineering, Binghamton University, the State University of New York, Binghamton, NY 13902, United States
| |
Collapse
|
37
|
Arslan U, van den Hil FE, Mummery CL, Orlova V. Generation and Characterization of hiPSC-Derived Vascularized-, Perfusable Cardiac Microtissues-on-Chip. Curr Protoc 2024; 4:e1097. [PMID: 39036931 DOI: 10.1002/cpz1.1097] [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] [Indexed: 07/23/2024]
Abstract
In the heart in vivo, vasculature forms a semi-permeable endothelial barrier for selective nutrient and (immune) cell delivery to the myocardium and removal of waste products. Crosstalk between the vasculature and the heart cells regulates homeostasis in health and disease. To model heart development and disease in vitro it is important that essential features of this crosstalk are captured. Cardiac organoid and microtissue models often integrate endothelial cells (ECs) to form microvascular networks inside the 3D structure. However, in static culture without perfusion, these networks may fail to show essential functionality. Here, we describe a protocol to generate an in vitro model of human induced pluripotent stem cell (hiPSC)-derived vascularized cardiac microtissues on a microfluidic organ-on-chip platform (VMToC) in which the blood vessels are perfusable. First, prevascularized cardiac microtissues (MT) are formed by combining hiPSC-derived cardiomyocytes, ECs, and cardiac fibroblasts in a pre-defined ratio. Next, these prevascularized MTs are integrated in the chips in a fibrin hydrogel containing additional vascular cells, which self-organize into tubular structures. The MTs become vascularized through anastomosis between the pre-existing microvasculature in the MT and the external vascular network. The VMToCs are then ready for downstream structural and functional assays and basic characterization. Using this protocol, cardiac MTs can be efficiently and robustly vascularized and perfused within 7 days. In vitro vascularized organoid and MT models have the potential to transition current 3D cardiac models to more physiologically relevant organ models that allow the role of the endothelial barrier in drug and inflammatory response to be investigated. © 2024 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Generation of VMToC Support Protocol 1: Functional Characterization of VMToC Support Protocol 2: Structural Characterization of VMToC.
Collapse
Affiliation(s)
- Ulgu Arslan
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Valeria Orlova
- Department of Anatomy and Embryology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
38
|
Roshanbinfar K, Schiffer M, Carls E, Angeloni M, Koleśnik-Gray M, Schruefer S, Schubert DW, Ferrazzi F, Krstić V, Fleischmann BK, Roell W, Engel FB. Electrically Conductive Collagen-PEDOT:PSS Hydrogel Prevents Post-Infarct Cardiac Arrhythmia and Supports hiPSC-Cardiomyocyte Function. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403642. [PMID: 38653478 DOI: 10.1002/adma.202403642] [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: 03/11/2024] [Indexed: 04/25/2024]
Abstract
Myocardial infarction (MI) causes cell death, disrupts electrical activity, triggers arrhythmia, and results in heart failure, whereby 50-60% of MI-associated deaths manifest as sudden cardiac deaths (SCD). The most effective therapy for SCD prevention is implantable cardioverter defibrillators (ICDs). However, ICDs contribute to adverse remodeling and disease progression and do not prevent arrhythmia. This work develops an injectable collagen-PEDOT:PSS (poly(3,4-ethylenedioxythiophene) polystyrene sulfonate) hydrogel that protects infarcted hearts against ventricular tachycardia (VT) and can be combined with human induced pluripotent stem cell (hiPSC)-cardiomyocytes to promote partial cardiac remuscularization. PEDOT:PSS improves collagen gel formation, micromorphology, and conductivity. hiPSC-cardiomyocytes in collagen-PEDOT:PSS hydrogels exhibit near-adult sarcomeric length, improved contractility, enhanced calcium handling, and conduction velocity. RNA-sequencing data indicate enhanced maturation and improved cell-matrix interactions. Injecting collagen-PEDOT:PSS hydrogels in infarcted mouse hearts decreases VT to the levels of healthy hearts. Collectively, collagen-PEDOT:PSS hydrogels offer a versatile platform for treating cardiac injuries.
Collapse
Affiliation(s)
- Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Miriam Schiffer
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany
| | - Esther Carls
- Department of Cardiac Surgery, UKB, University of Bonn, Germany
| | - Miriam Angeloni
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Maria Koleśnik-Gray
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany
| | - Stefan Schruefer
- Institute of Polymer Materials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Martensstr. 7, 91058, Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Martensstr. 7, 91058, Erlangen, Germany
| | - Fulvia Ferrazzi
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Vojislav Krstić
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany
| | - Wilhelm Roell
- Department of Cardiac Surgery, UKB, University of Bonn, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| |
Collapse
|
39
|
Lickiss B, Hunker J, Bhagwan J, Linder P, Thomas U, Lotay H, Broadbent S, Dragicevic E, Stoelzle-Feix S, Turner J, Gossmann M. Chamber-specific contractile responses of atrial and ventricular hiPSC-cardiomyocytes to GPCR and ion channel targeting compounds: A microphysiological system for cardiac drug development. J Pharmacol Toxicol Methods 2024; 128:107529. [PMID: 38857637 DOI: 10.1016/j.vascn.2024.107529] [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: 02/26/2024] [Revised: 05/15/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) have found utility for conducting in vitro drug screening and disease modelling to gain crucial insights into pharmacology or disease phenotype. However, diseases such as atrial fibrillation, affecting >33 M people worldwide, demonstrate the need for cardiac subtype-specific cells. Here, we sought to investigate the base characteristics and pharmacological differences between commercially available chamber-specific atrial or ventricular hiPSC-CMs seeded onto ultra-thin, flexible PDMS membranes to simultaneously measure contractility in a 96 multi-well format. We investigated the effects of GPCR agonists (acetylcholine and carbachol), a Ca2+ channel agonist (S-Bay K8644), an HCN channel antagonist (ivabradine) and K+ channel antagonists (4-AP and vernakalant). We observed differential effects between atrial and ventricular hiPSC-CMs on contractile properties including beat rate, beat duration, contractile force and evidence of arrhythmias at a range of concentrations. As an excerpt of the compound analysis, S-Bay K8644 treatment showed an induced concentration-dependent transient increase in beat duration of atrial hiPSC-CMs, whereas ventricular cells showed a physiological increase in beat rate over time. Carbachol treatment produced marked effects on atrial cells, such as increased beat duration alongside a decrease in beat rate over time, but only minimal effects on ventricular cardiomyocytes. In the context of this chamber-specific pharmacology, we not only add to contractile characterization of hiPSC-CMs but propose a multi-well platform for medium-throughput early compound screening. Overall, these insights illustrate the key pharmacological differences between chamber-specific cardiomyocytes and their application on a multi-well contractility platform to gain insights for in vitro cardiac liability studies and disease modelling.
Collapse
Affiliation(s)
| | - Jan Hunker
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Jamie Bhagwan
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Peter Linder
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Ulrich Thomas
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Hardeep Lotay
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Steven Broadbent
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Elena Dragicevic
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | | | - Jan Turner
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | |
Collapse
|
40
|
Groen E, Mummery CL, Yiangou L, Davis RP. Three-dimensional cardiac models: a pre-clinical testing platform. Biochem Soc Trans 2024; 52:1045-1059. [PMID: 38778769 PMCID: PMC11346450 DOI: 10.1042/bst20230444] [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: 01/31/2024] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Major advancements in human pluripotent stem cell (hPSC) technology over recent years have yielded valuable tools for cardiovascular research. Multi-cell type 3-dimensional (3D) cardiac models in particular, are providing complementary approaches to animal studies that are better representatives than simple 2-dimensional (2D) cultures of differentiated hPSCs. These human 3D cardiac models can be broadly divided into two categories; namely those generated through aggregating pre-differentiated cells and those that form self-organizing structures during their in vitro differentiation from hPSCs. These models can either replicate aspects of cardiac development or enable the examination of interactions among constituent cell types, with some of these models showing increased maturity compared with 2D systems. Both groups have already emerged as physiologically relevant pre-clinical platforms for studying heart disease mechanisms, exhibiting key functional attributes of the human heart. In this review, we describe the different cardiac organoid models derived from hPSCs, their generation methods, applications in cardiovascular disease research and use in drug screening. We also address their current limitations and challenges as pre-clinical testing platforms and propose potential improvements to enhance their efficacy in cardiac drug discovery.
Collapse
Affiliation(s)
- Eline Groen
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300RC Leiden, The Netherlands
| | - Loukia Yiangou
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Richard P. Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, 2300RC Leiden, The Netherlands
| |
Collapse
|
41
|
Pardon G, Vander Roest AS, Chirikian O, Birnbaum F, Lewis H, Castillo EA, Wilson R, Denisin AK, Blair CA, Holbrook C, Koleckar K, Chang ACY, Blau HM, Pruitt BL. Tracking single hiPSC-derived cardiomyocyte contractile function using CONTRAX an efficient pipeline for traction force measurement. Nat Commun 2024; 15:5427. [PMID: 38926342 PMCID: PMC11208611 DOI: 10.1038/s41467-024-49755-3] [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: 03/17/2021] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) are powerful in vitro models to study the mechanisms underlying cardiomyopathies and cardiotoxicity. Quantification of the contractile function in single hiPSC-CMs at high-throughput and over time is essential to disentangle how cellular mechanisms affect heart function. Here, we present CONTRAX, an open-access, versatile, and streamlined pipeline for quantitative tracking of the contractile dynamics of single hiPSC-CMs over time. Three software modules enable: parameter-based identification of single hiPSC-CMs; automated video acquisition of >200 cells/hour; and contractility measurements via traction force microscopy. We analyze >4,500 hiPSC-CMs over time in the same cells under orthogonal conditions of culture media and substrate stiffnesses; +/- drug treatment; +/- cardiac mutations. Using undirected clustering, we reveal converging maturation patterns, quantifiable drug response to Mavacamten and significant deficiencies in hiPSC-CMs with disease mutations. CONTRAX empowers researchers with a potent quantitative approach to develop cardiac therapies.
Collapse
Grants
- R00 HL153679 NHLBI NIH HHS
- K99 HL153679 NHLBI NIH HHS
- RM1 GM131981 NIGMS NIH HHS
- 20POST35211011 American Heart Association (American Heart Association, Inc.)
- 17CSA33590101 American Heart Association (American Heart Association, Inc.)
- 18CDA34110411 American Heart Association (American Heart Association, Inc.)
- 1R21HL13099301 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 18POST34080160 American Heart Association (American Heart Association, Inc.)
- 1F31HL158227 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F31 HL158227 NHLBI NIH HHS
- 201411MFE-338745-169197 Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de Recherche en Santé du Canada)
- P2SKP2_164954 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- 13POST14480004 American Heart Association (American Heart Association, Inc.)
- RM1GM131981 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 82070248 National Natural Science Foundation of China (National Science Foundation of China)
- P400PM_180825 Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung (Swiss National Science Foundation)
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Shanghai Pujiang Program 19PJ1407000 Program for Professor of Special Appointment (Eastern Scholar) at Shanghai Institutions of Higher Learning 0900000024 to A.C.Y.C. Innovative Research Team of High-Level Local Universities in Shanghai (A.C.Y.C.)
- the Baxter Foundation, Li Ka Shing Foundation and The Stanford Cardiovascular Institute
Collapse
Affiliation(s)
- Gaspard Pardon
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
- School of Life Sciences, EPFL École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alison S Vander Roest
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Engineering, Michigan Engineering, University of Michigan Ann Arbor, MI, USA
| | - Orlando Chirikian
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA
| | - Foster Birnbaum
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Henry Lewis
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Erica A Castillo
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
| | - Robin Wilson
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Aleksandra K Denisin
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
| | - Cheavar A Blair
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Colin Holbrook
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kassie Koleckar
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Alex C Y Chang
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Shanghai Institute of Precision Medicine and Department of Cardiology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200125, China
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Beth L Pruitt
- Departments of Mechanical Engineering and of Bioengineering, Stanford University, School of Engineering and School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Departments of Bioengineering and Mechanical Engineering, University of California, Santa Barbara, CA, USA.
- Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA, USA.
| |
Collapse
|
42
|
Baheux Blin M, Loreau V, Schnorrer F, Mangeol P. PatternJ: an ImageJ toolset for the automated and quantitative analysis of regular spatial patterns found in sarcomeres, axons, somites, and more. Biol Open 2024; 13:bio060548. [PMID: 38887972 PMCID: PMC11212633 DOI: 10.1242/bio.060548] [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: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 06/20/2024] Open
Abstract
Regular spatial patterns are ubiquitous forms of organization in nature. In animals, regular patterns can be found from the cellular scale to the tissue scale, and from early stages of development to adulthood. To understand the formation of these patterns, how they assemble and mature, and how they are affected by perturbations, a precise quantitative description of the patterns is essential. However, accessible tools that offer in-depth analysis without the need for computational skills are lacking for biologists. Here, we present PatternJ, a novel toolset to analyze regular one-dimensional patterns precisely and automatically. This toolset, to be used with the popular imaging processing program ImageJ/Fiji, facilitates the extraction of key geometric features within and between pattern repeats in static images and time-lapse series. We validate PatternJ with simulated data and test it on images of sarcomeres from insect muscles and contracting cardiomyocytes, actin rings in neurons, and somites from zebrafish embryos obtained using confocal fluorescence microscopy, STORM, electron microscopy, and brightfield imaging. We show that the toolset delivers subpixel feature extraction reliably even with images of low signal-to-noise ratio. PatternJ's straightforward use and functionalities make it valuable for various scientific fields requiring quantitative one-dimensional pattern analysis, including the sarcomere biology of muscles or the patterning of mammalian axons, speeding up discoveries with the bonus of high reproducibility.
Collapse
Affiliation(s)
- Mélina Baheux Blin
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| | - Vincent Loreau
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| | - Frank Schnorrer
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| | - Pierre Mangeol
- Aix Marseille Université, CNRS, Institut de Biologie du Développement de Marseille, IBDM-UMR7288, Marseille 13009, France
| |
Collapse
|
43
|
Yuan X, Yu T, Zhang Z, Li S. Non-invasive assessment of proarrhythmic risks associated with isoprenaline and the dietary supplement ingredient synephrine using human induced pluripotent stem cell-derived cardiomyocytes. Front Cardiovasc Med 2024; 11:1407138. [PMID: 38911513 PMCID: PMC11190318 DOI: 10.3389/fcvm.2024.1407138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/23/2024] [Indexed: 06/25/2024] Open
Abstract
Background There have been conflicting reports about the proarrhythmic risk of p-synephrine (SYN). To address this, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) combined with the microelectrode array (MEA) system have been utilized to assess arrhythmia risks, particularly in the context of adrenomimetic drugs. Aim This study aims to determine whether MEA recordings from hiPSC-CMs could predict the proarrhythmic risk of adrenomimetic drugs and to investigate the cardiovascular effects and mechanisms of SYN. Materials and methods We employed MEA recordings to assess the electrophysiological properties of hiPSC-CMs and conducted concentration-response analyses to evaluate the effects of SYN and Isoprenaline (ISO) on beating rate and contractility. A risk scoring system for proarrhythmic risks was established based on hiPSC-CMs in this study. ISO, a classic beta-adrenergic drug, was also evaluated. Furthermore, the study evaluated the risk of SYN and recorded the concentration-response of beating rate, contractility and the change in the presence or absence of selective β1, β2 and β3 adrenergic blockers. Results Our results suggested that ISO carries a high risk of inducing arrhythmias, aligning with existing literature. SYN caused a 30% prolongation of the field potential duration (FPD) at a concentration of 206.326 μM, a change significantly different from baseline measurements and control treatments. The half maximal effective concentration (EC50) of SYN (3.31 μM) to affect hiPSC-CM beating rate is much higher than that of ISO (18.00 nM). The effect of SYN at an EC50 of 3.31 μM is about ten times more potent in hiPSC-CMs compared to neonatal rat cardiomyocytes (34.12 μM). SYN increased the contractility of cardiomyocytes by 29.97 ± 11.65%, compared to ISO's increase of 50.56 ± 24.15%. β1 receptor blockers almost eliminated the beating rate increase induced by both ISO and SYN, while neither β2 nor β3 blockers had a complete inhibitory effect. Conclusion The MEA and hiPSC-CM system could effectively predict the risk of adrenomimetic drugs. The study concludes that the proarrhythmia risk of SYN at conventional doses is low. SYN is more sensitive in increasing beating rate and contractility in human cardiomyocytes compared to rats, primarily activating β1 receptor.
Collapse
Affiliation(s)
| | | | | | - Sen Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
44
|
Gokhan I, Blum TS, Campbell SG. Engineered heart tissue: Design considerations and the state of the art. BIOPHYSICS REVIEWS 2024; 5:021308. [PMID: 38912258 PMCID: PMC11192576 DOI: 10.1063/5.0202724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/29/2024] [Indexed: 06/25/2024]
Abstract
Originally developed more than 20 years ago, engineered heart tissue (EHT) has become an important tool in cardiovascular research for applications such as disease modeling and drug screening. Innovations in biomaterials, stem cell biology, and bioengineering, among other fields, have enabled EHT technologies to recapitulate many aspects of cardiac physiology and pathophysiology. While initial EHT designs were inspired by the isolated-trabecula culture system, current designs encompass a variety of formats, each of which have unique strengths and limitations. In this review, we describe the most common EHT formats, and then systematically evaluate each aspect of their design, emphasizing the rational selection of components for each application.
Collapse
Affiliation(s)
| | - Thomas S. Blum
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | | |
Collapse
|
45
|
Zhang B, Luo Y, Zhou X, Gao L, Yin X, Yang H. GelMA micropattern enhances cardiomyocyte organization, maturation, and contraction via contact guidance. APL Bioeng 2024; 8:026108. [PMID: 38699629 PMCID: PMC11065454 DOI: 10.1063/5.0182585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Cardiac tissue engineering has emerged as a promising approach for restoring the functionality of damaged cardiac tissues following myocardial infarction. To effectively replicate the native anisotropic structure of cardiac tissues in vitro, this study focused on the fabrication of micropatterned gelatin methacryloyl hydrogels with varying geometric parameters. These substrates were evaluated for their ability to guide induced pluripotent stem cell-derived cardiomyocytes (CMs). The findings demonstrate that the mechanical properties of this hydrogel closely resemble those of native cardiac tissues, and it exhibits high fidelity in micropattern fabrication. Micropatterned hydrogel substrates lead to enhanced organization, maturation, and contraction of CMs. A microgroove with 20-μm-width and 20-μm-spacing was identified as the optimal configuration for maximizing the contact guidance effect, supported by analyses of nuclear orientation and F-actin organization. Furthermore, this specific micropattern design was found to promote CMs' maturation, as evidenced by increased expression of connexin 43 and vinculin, along with extended sarcomere length. It also enhanced CMs' contraction, resulting in larger contractile amplitudes and greater contractile motion anisotropy. In conclusion, these results underscore the significant benefits of optimizing micropatterned gelatin methacryloyl for improving CMs' organization, maturation, and contraction. This valuable insight paves the way for the development of highly organized and functionally mature cardiac tissues in vitro.
Collapse
Affiliation(s)
- Bin Zhang
- Author to whom correspondence should be addressed:
| | | | | | | | | | | |
Collapse
|
46
|
Butler K, Ahmed S, Jablonski J, Hookway TA. Engineered Cardiac Microtissue Biomanufacturing Using Human Induced Pluripotent Stem Cell Derived Epicardial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593960. [PMID: 38798424 PMCID: PMC11118268 DOI: 10.1101/2024.05.13.593960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Epicardial cells are a crucial component in constructing in vitro 3D tissue models of the human heart, contributing to the ECM environment and the resident mesenchymal cell population. Studying the human epicardium and its development from the proepicardial organ is difficult, but induced pluripotent stem cells can provide a source of human epicardial cells for developmental modeling and for biomanufacturing heterotypic cardiac tissues. This study shows that a robust population of epicardial cells (approx. 87.7% WT1+) can be obtained by small molecule modulation of the Wnt signaling pathway. The population maintains WT1 expression and characteristic epithelial morphology over successive passaging, but increases in size and decreases in cell number, suggesting a limit to their expandability in vitro. Further, low passage number epicardial cells formed into more robust 3D microtissues compared to their higher passage counterparts, suggesting that the ideal time frame for use of these epicardial cells for tissue engineering and modeling purposes is early on in their differentiated state. Additionally, the differentiated epicardial cells displayed two distinct morphologic sub populations with a subset of larger, more migratory cells which led expansion of the epicardial cells across various extracellular matrix environments. When incorporated into a mixed 3D co-culture with cardiomyocytes, epicardial cells promoted greater remodeling and migration without impairing cardiomyocyte function. This study provides an important characterization of stem cell-derived epicardial cells, identifying key characteristics that influence their ability to fabricate consistent engineered cardiac tissues.
Collapse
Affiliation(s)
- Kirk Butler
- Biomedical Engineering Department, Binghamton University, the State University of New York, Binghamton NY 13902
| | - Saif Ahmed
- Biomedical Engineering Department, Binghamton University, the State University of New York, Binghamton NY 13902
| | - Justin Jablonski
- Biomedical Engineering Department, University of Rochester, Rochester, NY14627
| | - Tracy A. Hookway
- Biomedical Engineering Department, Binghamton University, the State University of New York, Binghamton NY 13902
| |
Collapse
|
47
|
Feaster TK, Ewoldt JK, Avila A, Casciola M, Narkar A, Chen CS, Blinova K. Nonclinical evaluation of chronic cardiac contractility modulation on 3D human engineered cardiac tissues. J Cardiovasc Electrophysiol 2024; 35:895-905. [PMID: 38433304 DOI: 10.1111/jce.16222] [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: 12/27/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Cardiac contractility modulation (CCM) is a medical device-based therapy delivering non-excitatory electrical stimulations to the heart to enhance cardiac function in heart failure (HF) patients. The lack of human in vitro tools to assess CCM hinders our understanding of CCM mechanisms of action. Here, we introduce a novel chronic (i.e., 2-day) in vitro CCM assay to evaluate the effects of CCM in a human 3D microphysiological system consisting of engineered cardiac tissues (ECTs). METHODS Cryopreserved human induced pluripotent stem cell-derived cardiomyocytes were used to generate 3D ECTs. The ECTs were cultured, incorporating human primary ventricular cardiac fibroblasts and a fibrin-based gel. Electrical stimulation was applied using two separate pulse generators for the CCM group and control group. Contractile properties and intracellular calcium were measured, and a cardiac gene quantitative PCR screen was conducted. RESULTS Chronic CCM increased contraction amplitude and duration, enhanced intracellular calcium transient amplitude, and altered gene expression related to HF (i.e., natriuretic peptide B, NPPB) and excitation-contraction coupling (i.e., sodium-calcium exchanger, SLC8). CONCLUSION These data represent the first study of chronic CCM in a 3D ECT model, providing a nonclinical tool to assess the effects of cardiac electrophysiology medical device signals complementing in vivo animal studies. The methodology established a standardized 3D ECT-based in vitro testbed for chronic CCM, allowing evaluation of physiological and molecular effects on human cardiac tissues.
Collapse
Affiliation(s)
- Tromondae K Feaster
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jourdan K Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| | - Anna Avila
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Maura Casciola
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Akshay Narkar
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
| | - Ksenia Blinova
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
48
|
Kim Y, Wang K, Lock RI, Nash TR, Fleischer S, Wang BZ, Fine BM, Vunjak-Novakovic G. BeatProfiler: Multimodal In Vitro Analysis of Cardiac Function Enables Machine Learning Classification of Diseases and Drugs. IEEE OPEN JOURNAL OF ENGINEERING IN MEDICINE AND BIOLOGY 2024; 5:238-249. [PMID: 38606403 PMCID: PMC11008807 DOI: 10.1109/ojemb.2024.3377461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/13/2024] [Accepted: 03/10/2024] [Indexed: 04/13/2024] Open
Abstract
Goal: Contractile response and calcium handling are central to understanding cardiac function and physiology, yet existing methods of analysis to quantify these metrics are often time-consuming, prone to mistakes, or require specialized equipment/license. We developed BeatProfiler, a suite of cardiac analysis tools designed to quantify contractile function, calcium handling, and force generation for multiple in vitro cardiac models and apply downstream machine learning methods for deep phenotyping and classification. Methods: We first validate BeatProfiler's accuracy, robustness, and speed by benchmarking against existing tools with a fixed dataset. We further confirm its ability to robustly characterize disease and dose-dependent drug response. We then demonstrate that the data acquired by our automatic acquisition pipeline can be further harnessed for machine learning (ML) analysis to phenotype a disease model of restrictive cardiomyopathy and profile cardioactive drug functional response. To accurately classify between these biological signals, we apply feature-based ML and deep learning models (temporal convolutional-bidirectional long short-term memory model or TCN-BiLSTM). Results: Benchmarking against existing tools revealed that BeatProfiler detected and analyzed contraction and calcium signals better than existing tools through improved sensitivity in low signal data, reduction in false positives, and analysis speed increase by 7 to 50-fold. Of signals accurately detected by published methods (PMs), BeatProfiler's extracted features showed high correlations to PMs, confirming that it is reliable and consistent with PMs. The features extracted by BeatProfiler classified restrictive cardiomyopathy cardiomyocytes from isogenic healthy controls with 98% accuracy and identified relax90 as a top distinguishing feature in congruence with previous findings. We also show that our TCN-BiLSTM model was able to classify drug-free control and 4 cardiac drugs with different mechanisms of action at 96% accuracy. We further apply Grad-CAM on our convolution-based models to identify signature regions of perturbations by these drugs in calcium signals. Conclusions: We anticipate that the capabilities of BeatProfiler will help advance in vitro studies in cardiac biology through rapid phenotyping, revealing mechanisms underlying cardiac health and disease, and enabling objective classification of cardiac disease and responses to drugs.
Collapse
Affiliation(s)
- Youngbin Kim
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Kunlun Wang
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Roberta I. Lock
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Trevor R. Nash
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Sharon Fleischer
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Bryan Z. Wang
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
| | - Barry M. Fine
- Department of MedicineDivision of CardiologyColumbia University Medical CenterNew YorkNY10032USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10032USA
- Department of MedicineDivision of CardiologyColumbia University Medical CenterNew YorkNY10032USA
| |
Collapse
|
49
|
Ma J, Ross L, Grube C, Wang HS. Toxicity of low dose bisphenols in human iPSC-derived cardiomyocytes and human cardiac organoids - Impact on contractile function and hypertrophy. CHEMOSPHERE 2024; 353:141567. [PMID: 38417488 DOI: 10.1016/j.chemosphere.2024.141567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 02/23/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
Bisphenol A (BPA) and its analogs are common environmental chemicals with various adverse health impacts, including cardiac toxicity. In this study, we examined the long term effect of low dose BPA and three common BPA analogs, bisphenol S (BPS), bisphenol F (BPF), and bisphenol AF (BPAF), in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) based models. HiPSC-CMs and human cardiac organoids were exposed to these chemicals for 4-5 or 20 days. 1 nM BPA, BPS, and BPAF, but not BPF, resulted in suppressed myocyte contractility, retarded contraction kinetics, and aberrant Ca2+ transients in hiPSC-CMs. In cardiac organoids, BPAF and BPA, but not the other bisphenols, resulted in suppressed contraction and Ca2+ transients, and aberrant contraction kinetics. The order of toxicities was BPAF > BPA>∼BPS > BPF and the toxicities of BPAF and BPA were more pronounced under longer exposure. The impact of BPAF on myocyte contraction and Ca2+ handling was mediated by reduction of sarcoplasmic reticulum Ca2+ load and inhibition of L-type Ca2+ channel involving alternation of Ca2+ handling proteins. Impaired myocyte Ca2+ handling plays a key role in cardiac pathophysiology and is a characteristic of cardiac hypertrophy; therefore we examined the potential pro-hypertrophic cardiotoxicity of these bisphenols. Four to five day exposure to BPAF did not cause hypertrophy in normal hiPSC-CMs, but significantly exacerbated the hypertrophic phenotype in myocytes with existing hypertrophy induced by endothelin-1, characterized by increased cell size and elevated expression of the hypertrophic marker proBNP. This pro-hypertrophic cardiotoxicity was also occurred in cardiac organoids, with BPAF having the strongest toxicity, followed by BPA. Our findings demonstrate that long term exposures to BPA and some of its analogs cause contractile dysfunction and abnormal Ca2+ handling, and have potential pro-hypertrophic cardiotoxicity in human heart cells/tissues, and suggest that some bisphenol chemicals may be a risk factor for cardiac hypertrophy in human hearts.
Collapse
Affiliation(s)
- Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA.
| | - Leah Ross
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Christian Grube
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267 USA
| |
Collapse
|
50
|
Kobeissi H, Jilberto J, Karakan MÇ, Gao X, DePalma SJ, Das SL, Quach L, Urquia J, Baker BM, Chen CS, Nordsletten D, Lejeune E. MicroBundleCompute: Automated segmentation, tracking, and analysis of subdomain deformation in cardiac microbundles. PLoS One 2024; 19:e0298863. [PMID: 38530829 PMCID: PMC10965069 DOI: 10.1371/journal.pone.0298863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/01/2024] [Indexed: 03/28/2024] Open
Abstract
Advancing human induced pluripotent stem cell derived cardiomyocyte (hiPSC-CM) technology will lead to significant progress ranging from disease modeling, to drug discovery, to regenerative tissue engineering. Yet, alongside these potential opportunities comes a critical challenge: attaining mature hiPSC-CM tissues. At present, there are multiple techniques to promote maturity of hiPSC-CMs including physical platforms and cell culture protocols. However, when it comes to making quantitative comparisons of functional behavior, there are limited options for reliably and reproducibly computing functional metrics that are suitable for direct cross-system comparison. In addition, the current standard functional metrics obtained from time-lapse images of cardiac microbundle contraction reported in the field (i.e., post forces, average tissue stress) do not take full advantage of the available information present in these data (i.e., full-field tissue displacements and strains). Thus, we present "MicroBundleCompute," a computational framework for automatic quantification of morphology-based mechanical metrics from movies of cardiac microbundles. Briefly, this computational framework offers tools for automatic tissue segmentation, tracking, and analysis of brightfield and phase contrast movies of beating cardiac microbundles. It is straightforward to implement, runs without user intervention, requires minimal input parameter setting selection, and is computationally inexpensive. In this paper, we describe the methods underlying this computational framework, show the results of our extensive validation studies, and demonstrate the utility of exploring heterogeneous tissue deformations and strains as functional metrics. With this manuscript, we disseminate "MicroBundleCompute" as an open-source computational tool with the aim of making automated quantitative analysis of beating cardiac microbundles more accessible to the community.
Collapse
Affiliation(s)
- Hiba Kobeissi
- Department of Mechanical Engineering, Boston University, Boston, MA, United States of America
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States of America
| | - Javiera Jilberto
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - M. Çağatay Karakan
- Department of Mechanical Engineering, Boston University, Boston, MA, United States of America
- Photonics Center, Boston University, Boston, MA, United States of America
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
| | - Xining Gao
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Shoshana L. Das
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - Lani Quach
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Jonathan Urquia
- Department of Electrical and Computer Engineering, New York Institute of Technology, New York, NY, United States of America
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Christopher S. Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, United States of America
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, United States of America
| | - David Nordsletten
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI, United States of America
- Department of Biomedical Engineering, School of Imaging Sciences and Biomedical Engineering, King’s Health Partners, King’s College London, King’s Health Partners, London, United Kingdom
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, United States of America
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States of America
| |
Collapse
|