1
|
Liu X, Yu L, Xiao A, Sun W, Wang H, Wang X, Zhou Y, Li C, Li J, Wang Y, Wang G. Analytical methods in studying cell force sensing: principles, current technologies and perspectives. Regen Biomater 2025; 12:rbaf007. [PMID: 40337625 PMCID: PMC12057814 DOI: 10.1093/rb/rbaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/16/2024] [Accepted: 02/10/2025] [Indexed: 05/09/2025] Open
Abstract
Mechanical stimulation plays a crucial role in numerous biological activities, including tissue development, regeneration and remodeling. Understanding how cells respond to their mechanical microenvironment is vital for investigating mechanotransduction with adequate spatial and temporal resolution. Cell force sensing-also known as mechanosensation or mechanotransduction-involves force transmission through the cytoskeleton and mechanochemical signaling. Insights into cell-extracellular matrix interactions and mechanotransduction are particularly relevant for guiding biomaterial design in tissue engineering. To establish a foundation for mechanical biomedicine, this review will provide a comprehensive overview of cell mechanotransduction mechanisms, including the structural components essential for effective mechanical responses, such as cytoskeletal elements, force-sensitive ion channels, membrane receptors and key signaling pathways. It will also discuss the clutch model in force transmission, the role of mechanotransduction in both physiology and pathological contexts, and biomechanics and biomaterial design. Additionally, we outline analytical approaches for characterizing forces at cellular and subcellular levels, discussing the advantages and limitations of each method to aid researchers in selecting appropriate techniques. Finally, we summarize recent advancements in cell force sensing and identify key challenges for future research. Overall, this review should contribute to biomedical engineering by supporting the design of biomaterials that integrate precise mechanical information.
Collapse
Affiliation(s)
- Xiaojun Liu
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao 266024, China
| | - Lei Yu
- Department of Traditional Chinese Medicine, Qingdao Special Service Sanatorium of PLA Navy, Qingdao 266071, China
| | - Adam Xiao
- Department of Chemistry, University of British Columbia, Vancouver, BC V6T 1Z1, Canada
| | - Wenxu Sun
- School of Sciences, Nantong University, Nantong 226019, China
| | - Han Wang
- State Key Laboratory of Precision Measuring Technology and Instruments, School of Precision Instrument and Optoelectronics Engineering, Tianjin University, Tianjin 300072, China
| | - Xiangxiu Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Yanghao Zhou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Chao Li
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao 266024, China
| | - Jiangtao Li
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
| | - Yongliang Wang
- College of Life Sciences and Health, University of Health and Rehabilitation Sciences, Qingdao 266113, China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao 266024, China
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
- Qindao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266044, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
- JinFeng Laboratory, Chongqing 401329, China
| |
Collapse
|
2
|
Sileo A, Montrone F, Rouchon A, Trueb D, Selvi J, Schmid M, Graef J, Züger F, Serino G, Massai D, Di Maggio N, Melo Rodriguez G, Köser J, Schoelkopf J, Banfi A, Marsano A, Gullo M. Toward Origami-Inspired In Vitro Cardiac Tissue Models. ACS Biomater Sci Eng 2025; 11:1583-1597. [PMID: 39978771 PMCID: PMC11897953 DOI: 10.1021/acsbiomaterials.4c01594] [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: 08/30/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/22/2025]
Abstract
The advancement of in vitro engineered cardiac tissue-based patches is paramount for providing viable solutions for restoring cardiac function through in vivo implantation. Numerous techniques described in the literature aim to provide diverse mechanical and topographical cues simultaneously, fostering enhanced in vitro cardiac maturation and functionality. Among these, cellulose paper-based scaffolds have gained attention owing to their inherent benefits, such as biocompatibility and ease of chemical and physical modification. This study introduces a novel approach of utilizing customized paper-based scaffolds as cell culture substrates, facilitating both the formation and manipulation of cell constructs while promoting mechanical contraction. Here, we investigated two methodologies to foster mechanical contractions of paper-based constructs: the incorporation of micropatterns on paper to dictate cell orientation and macropattern created by the origami-folded paper. Both approaches provide mechanical support and foster cardiac functionality. However, while micropatterning does not significantly improve the functional parameters, a macropattern created by origami folding proves to be essential in facilitating contraction of the paper-based cardiac constructs. Furthermore, we provide proof of principle for the combination with a layer of physiologically differentiated microvascular networks. This approach holds great promise for the development of structurally organized contractile cardiac tissues with the possibility of creating multistrata of cardiac and vascular layers to promote in vivo cell survival and function beyond what is typically achieved in conventional cell culture.
Collapse
Affiliation(s)
- Antonio Sileo
- Department
of Surgery, University Hospital Basel, Basel 4031, Switzerland
- Department
of Biomedicine, University Hospital Basel
and University of Basel, Basel 4031, Switzerland
| | - Federica Montrone
- Department
of Surgery, University Hospital Basel, Basel 4031, Switzerland
- Department
of Biomedicine, University Hospital Basel
and University of Basel, Basel 4031, Switzerland
| | - Adelin Rouchon
- Department
of Surgery, University Hospital Basel, Basel 4031, Switzerland
- Department
of Biomedicine, University Hospital Basel
and University of Basel, Basel 4031, Switzerland
| | - Donata Trueb
- Institute
for Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz 4132, Switzerland
| | - Jasmin Selvi
- Institute
for Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz 4132, Switzerland
| | - Moritz Schmid
- Institute
for Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz 4132, Switzerland
| | - Julian Graef
- Institute
for Chemistry and Bioanalytics, University
of Applied Sciences and Arts Northwestern Switzerland, Muttenz 4132, Switzerland
| | - Fabian Züger
- Institute
for Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz 4132, Switzerland
| | - Gianpaolo Serino
- Department
of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
| | - Diana Massai
- Department
of Mechanical and Aerospace Engineering and PolitoBIOMed Lab, Politecnico di Torino, Turin 10129, Italy
| | - Nunzia Di Maggio
- Department
of Surgery, University Hospital Basel, Basel 4031, Switzerland
- Department
of Biomedicine, University Hospital Basel
and University of Basel, Basel 4031, Switzerland
| | | | - Joachim Köser
- Institute
for Chemistry and Bioanalytics, University
of Applied Sciences and Arts Northwestern Switzerland, Muttenz 4132, Switzerland
| | | | - Andrea Banfi
- Department
of Surgery, University Hospital Basel, Basel 4031, Switzerland
- Department
of Biomedicine, University Hospital Basel
and University of Basel, Basel 4031, Switzerland
| | - Anna Marsano
- Department
of Surgery, University Hospital Basel, Basel 4031, Switzerland
- Department
of Biomedicine, University Hospital Basel
and University of Basel, Basel 4031, Switzerland
| | - Maurizio Gullo
- Institute
for Medical Engineering and Medical Informatics, University of Applied Sciences and Arts Northwestern Switzerland, Muttenz 4132, Switzerland
| |
Collapse
|
3
|
Chen S, Tan L, Serpooshan V, Chen H. A 3D bioprinted adhesive tissue engineering scaffold to repair ischemic heart injury. Biomater Sci 2025; 13:506-522. [PMID: 39639799 DOI: 10.1039/d4bm00988f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Adhesive tissue engineering scaffold (ATES) devices can be secured on tissues by relying on their intrinsic adhesive properties, hence, avoiding the complications such as host tissue/scaffold damage that are associated with conventional scaffold fixation methods like suturing or bioglue. This study introduces a new generation of three-dimensional (3D) bioprinted ATES systems for use as cardiac patches to regenerate the adult human heart. Tyramine-modified methacrylated hyaluronic acid (HAMA-tyr), gelatin methacrylate (GelMA), and gelatin were used to create the hybrid bioink formulation with self-adhesive properties. ATESs were bioprinted and further modified to improve the adhesion properties. In-depth characterization of printing fidelity, pore size, mechanical properties, swelling behavior, as well as biocompatibility was used to create ATESs with optimal biological function. Following in vitro testing, the ATESs were tested in a mouse model of myocardial infarction to study the scaffold adhesive strength in biological milieu. The method developed in this study can be used to manufacture off-the-shelf ATESs with complex cellular and extracellular architecture, with robust potential for clinical translation into a variety of personalized tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Lindan Tan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| | - Vahid Serpooshan
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, GA 30322, USA
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
- Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Haifeng Chen
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China.
| |
Collapse
|
4
|
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
|
5
|
Gonzalo A, Augustin CM, Bifulco SF, Telle Å, Chahine Y, Kassar A, Guerrero-Hurtado M, Durán E, Martínez-Legazpi P, Flores O, Bermejo J, Plank G, Akoum N, Boyle PM, Del Alamo JC. Multiphysics simulations reveal haemodynamic impacts of patient-derived fibrosis-related changes in left atrial tissue mechanics. J Physiol 2024; 602:6789-6812. [PMID: 39513553 DOI: 10.1113/jp287011] [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: 05/29/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Stroke is a leading cause of death and disability worldwide. Atrial myopathy, including fibrosis, is associated with an increased risk of ischaemic stroke, but the mechanisms underlying this association are poorly understood. Fibrosis modifies myocardial structure, impairing electrical propagation and tissue biomechanics, and creating stagnant flow regions where clots could form. Fibrosis can be mapped non-invasively using late gadolinium enhancement magnetic resonance imaging (LGE-MRI). However, fibrosis maps are not currently incorporated into stroke risk calculations or computational electro-mechano-fluidic models. We present multiphysics simulations of left atrial (LA) myocardial motion and haemodynamics using patient-specific anatomies and fibrotic maps from LGE-MRI. We modify tissue stiffness and active tension generation in fibrotic regions and investigate how these changes affect LA flow for different fibrotic burdens. We find that fibrotic regions and, to a lesser extent, non-fibrotic regions experience reduced myocardial strain, resulting in decreased LA emptying fraction consistent with clinical observations. Both fibrotic tissue stiffening and hypocontractility independently reduce LA function, but, together, these two alterations cause more pronounced effects than either one alone. Fibrosis significantly alters flow patterns throughout the atrial chamber, and particularly, the filling and emptying jets of the left atrial appendage (LAA). The effects of fibrosis in LA flow are largely captured by the concomitant changes in LA emptying fraction except inside the LAA, where a multifactorial behaviour is observed. This work illustrates how high-fidelity, multiphysics models can be used to study thrombogenesis mechanisms in patient-specific anatomies, shedding light onto the links between atrial fibrosis and ischaemic stroke. KEY POINTS: Left atrial (LA) fibrosis is associated with arrhythmogenesis and increased risk of ischaemic stroke; its extent and pattern can be quantified on a patient-specific basis using late gadolinium enhancement magnetic resonance imaging. Current stroke risk prediction tools have limited personalization, and their accuracy could be improved by incorporating patient-specific information such as fibrotic maps and haemodynamic patterns. We present the first electro-mechano-fluidic multiphysics computational simulations of LA flow, including fibrosis and anatomies from medical imaging. Mechanical changes in fibrotic tissue impair global LA motion, decreasing LA and left atrial appendage (LAA) emptying fractions, especially in subjects with higher fibrosis burdens. Fibrotic-mediated LA motion impairment alters LA and LAA flow near the endocardium and the whole cavity, ultimately leading to more stagnant blood regions in the LAA.
Collapse
Affiliation(s)
- Alejandro Gonzalo
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Christoph M Augustin
- Gottfried Schatz Research Center, Division of Biophysics, Medical University of Graz, Graz, Austria
| | - Savannah F Bifulco
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Åshild Telle
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Yaacoub Chahine
- School of Cardiology, University of Washington, Seattle, WA, USA
| | - Ahmad Kassar
- School of Cardiology, University of Washington, Seattle, WA, USA
| | - Manuel Guerrero-Hurtado
- Department of Aerospace and Biomedical Engineering, Universidad Carlos III de Madrid, Leganés, Spain
| | - Eduardo Durán
- Dept. Ing. Mecánica, Térmica y de Fluidos, Universidad de Málaga, Málaga, Spain
| | | | - Oscar Flores
- Department of Aerospace and Biomedical Engineering, Universidad Carlos III de Madrid, Leganés, Spain
| | - Javier Bermejo
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medical School, Complutense University of Madrid, Madrid, Spain
| | - Gernot Plank
- Gottfried Schatz Research Center, Division of Biophysics, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Nazem Akoum
- School of Cardiology, University of Washington, Seattle, WA, USA
| | - Patrick M Boyle
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Juan C Del Alamo
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Youssef AS, Petrescu A, Salaets T, Bézy S, Wouters L, Orlowska M, Caenen A, Duchenne J, Puvrez A, Cools B, Heying R, D'hooge J, Gewillig M, Voigt JU. Evolution of Natural Myocardial Shear Wave Behavior in Young Hearts: Determinant Factors and Reproducibility Analysis. J Am Soc Echocardiogr 2024; 37:1051-1061. [PMID: 39002706 DOI: 10.1016/j.echo.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 07/02/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Myocardial diastolic function assessment in children by conventional echocardiography is challenging. High-frame rate echocardiography facilitates the assessment of myocardial stiffness, a key factor in diastolic function, by measuring the propagation velocities of myocardial shear waves (SWs). However, normal values of natural SWs in children are currently lacking. The aim of this study was to explore the behavior of natural SWs among children and adolescents, their reproducibility, and the factors affecting SW velocities from childhood into adulthood. METHODS One hundred six healthy children (2-18 years of age) and 62 adults (19-80 years of age) were recruited. High-frame rate images were acquired using a modified commercial scanner. An anatomic M-mode line was drawn along the ventricular septum, and propagation velocities of natural SWs after mitral valve closure were measured in the tissue acceleration-coded M-mode display. RESULTS Throughout life, SW velocities after mitral valve closure exhibited pronounced age dependency (r = 0.73; P < .001). Among the pediatric population, SW velocities correlated significantly with measures of cardiac geometry (septal thickness and left ventricular end-diastolic dimension), local hemodynamics (systolic blood pressure), and echocardiographic parameters of systolic and diastolic function (global longitudinal strain, mitral E/e' ratio, isovolumic relaxation time, and mitral deceleration time) (P < .001). In a multivariate analysis including all these factors, the predictors of SW velocities were age, mitral E/e', and global longitudinal strain (r = 0.81). CONCLUSIONS Natural myocardial SW velocities in children can be detected and measured. SW velocities showed significant dependence on age and diastolic function. Natural SWs could be a promising additive tool for the assessment of diastolic function among children.
Collapse
Affiliation(s)
- Ahmed S Youssef
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Cardiovascular Medicine, Suez Canal University, Ismailia, Egypt
| | - Aniela Petrescu
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Cardiology, University Medical Center of the Johannes Gutenber-Universitat Mainz, Mainz, Germany
| | - Thomas Salaets
- Department of Pediatric Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Stéphanie Bézy
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Laurine Wouters
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marta Orlowska
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Annette Caenen
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Jürgen Duchenne
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Alexis Puvrez
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Bjorn Cools
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Pediatric Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Ruth Heying
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Pediatric Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Jan D'hooge
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Marc Gewillig
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Pediatric Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Jens-Uwe Voigt
- Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Romanelli G, Villarreal L, Espasandín C, Benech JC. Diabetes induces modifications in costameric proteins and increases cardiomyocyte stiffness. Am J Physiol Cell Physiol 2024; 327:C1263-C1273. [PMID: 39374079 DOI: 10.1152/ajpcell.00273.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 10/09/2024]
Abstract
Several studies have demonstrated that diabetes mellitus can increase the risk of cardiovascular disease and remains the principal cause of death in these patients. Costameres connect the sarcolemma with the cytoskeleton and extracellular matrix, facilitating the transmission of mechanical forces and cell signaling. They are related to cardiac physiology because individual cardiac cells are connected by intercalated discs that synchronize muscle contraction. Diabetes impacts the nanomechanical properties of cardiomyocytes, resulting in increased cellular and left ventricular stiffness, as evidenced in clinical studies of these patients. The question of whether costameric proteins are affected by diabetes in the heart has not been studied. This work analyzes whether type 1 diabetes mellitus (T1DM) modifies the costameric proteins and coincidentally changes the cellular mechanics in the same cardiomyocytes. The samples were analyzed by immunotechniques using laser confocal microscopy. Significant statistical differences were found in the spatial arrangement of the costameric proteins. However, these differences are not due to their expression. Atomic force microscopy was used to compare intrinsic cellular stiffness between diabetic and normal cardiomyocytes and obtain the first elasticity map sections of diabetic living cardiomyocytes. Data obtained demonstrated that diabetic cardiomyocytes had higher stiffness than control. The present work shows experimental evidence that intracellular changes related to cell-cell and cell-extracellular matrix communication occur, which could be related to cardiac pathogenic mechanisms. These changes could contribute to alterations in the mechanical and electrical properties of cardiomyocytes and, consequently, to diabetic cardiomyopathy.NEW & NOTEWORTHY The structural organization of cardiomyocyte proteins is critical for their efficient functioning as a contractile unit in the heart. This work shows that diabetes mellitus induces significant changes in the spatial organization of costamere proteins, t tubules, and intercalated discs. We obtained the first elasticity map sections of living diabetic cardiomyocytes. The results show statistical differences in the map sections of diabetic and control cardiomyocytes, with diabetic cardiomyocytes being stiffer than normal ones.
Collapse
Affiliation(s)
- Gerardo Romanelli
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Lihuén Villarreal
- Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Camila Espasandín
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Departamento de Biociencias Veterinarias, Facultad de Veterinaria, Universidad de la República, Montevideo, Uruguay
| | - Juan Claudio Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| |
Collapse
|
8
|
Gonzalo A, Augustin CM, Bifulco SF, Telle Å, Chahine Y, Kassar A, Guerrero-Hurtado M, Durán E, Martínez-Legazpi P, Flores O, Bermejo J, Plank G, Akoum N, Boyle PM, Del Alamo JC. Multi-physics simulations reveal hemodynamic impacts of patient-derived fibrosis-related changes in left atrial tissue mechanics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596526. [PMID: 38853952 PMCID: PMC11160719 DOI: 10.1101/2024.05.29.596526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Stroke is a leading cause of death and disability worldwide. Atrial myopathy, including fibrosis, is associated with an increased risk of ischemic stroke, but the mechanisms underlying this association are poorly understood. Fibrosis modifies myocardial structure, impairing electrical propagation and tissue biomechanics, and creating stagnant flow regions where clots could form. Fibrosis can be mapped non-invasively using late gadolinium enhancement magnetic resonance imaging (LGE-MRI). However, fibrosis maps are not currently incorporated into stroke risk calculations or computational electro-mechano-fluidic models. We present multi-physics simulations of left atrial (LA) myocardial motion and hemodynamics using patient-specific anatomies and fibrotic maps from LGE-MRI. We modify tissue stiffness and active tension generation in fibrotic regions and investigate how these changes affect LA flow for different fibrotic burdens. We find that fibrotic regions and, to a lesser extent, non-fibrotic regions experience reduced myocardial strain, resulting in decreased LA emptying fraction consistent with clinical observations. Both fibrotic tissue stiffening and hypocontractility independently reduce LA function, but together, these two alterations cause more pronounced effects than either one alone. Fibrosis significantly alters flow patterns throughout the atrial chamber, and particularly, the filling and emptying jets of the left atrial appendage (LAA). The effects of fibrosis in LA flow are largely captured by the concomitant changes in LA emptying fraction except inside the LAA, where a multi-factorial behavior is observed. This work illustrates how high-fidelity, multi-physics models can be used to study thrombogenesis mechanisms in patient-specific anatomies, shedding light onto the links between atrial fibrosis and ischemic stroke. Key points Left atrial (LA) fibrosis is associated with arrhythmogenesis and increased risk of ischemic stroke; its extent and pattern can be quantified on a patient-specific basis using late gadolinium enhancement magnetic resonance imaging.Current stroke risk prediction tools have limited personalization, and their accuracy could be improved by incorporating patient-specific information like fibrotic maps and hemodynamic patterns.We present the first electro-mechano-fluidic multi-physics computational simulations of LA flow, including fibrosis and anatomies from medical imaging. Mechanical changes in fibrotic tissue impair global LA motion, decreasing LA and left atrial appendage (LAA) emptying fractions, especially in subjects with higher fibrosis burdens. Fibrotic-mediated LA motion impairment alters LA and LAA flow near the endocardium and the whole cavity, ultimately leading to more stagnant blood regions in the LAA.
Collapse
|
9
|
Erhardt J, Ludwig S, Brock J, Hörning M. Native mechano-regulative matrix properties stabilize alternans dynamics and reduce spiral wave stabilization in cardiac tissue. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1443156. [PMID: 39381499 PMCID: PMC11458432 DOI: 10.3389/fnetp.2024.1443156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024]
Abstract
The stability of wave conduction in the heart is strongly related to the proper interplay between the electrophysiological activation and mechanical contraction of myocytes and extracellular matrix (ECM) properties. In this study, we statistically compare bioengineered cardiac tissues cultured on soft hydrogels ( E ≃ 12 kPa) and rigid glass substrates by focusing on the critical threshold of alternans, network-physiological tissue properties, and the formation of stable spiral waves that manifest after wave breakups. For the classification of wave dynamics, we use an improved signal oversampling technique and introduce simple probability maps to identify and visualize spatially concordant and discordant alternans as V- and X-shaped probability distributions. We found that cardiac tissues cultured on ECM-mimicking soft hydrogels show a lower variability of the calcium transient durations among cells in the tissue. This lowers the likelihood of forming stable spiral waves because of the larger dynamical range that tissues can be stably entrained with to form alternans and larger spatial spiral tip movement that increases the chance of self-termination on the tissue boundary. Conclusively, we show that a dysfunction in the excitation-contraction coupling dynamics facilitates life-threatening arrhythmic states such as spiral waves and, thus, highlights the importance of the network-physiological interplay between contractile myocytes and the ECM.
Collapse
Affiliation(s)
| | | | | | - Marcel Hörning
- Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
10
|
Hassan S, Rezaei Z, Luna E, Yilmaz-Aykut D, Lee MC, Perea AM, Jamaiyar A, Bassous N, Hirano M, Tourk FM, Choi C, Becker M, Yazdi I, Fan K, Avila-Ramirez A, Ge D, Abdi R, Fisch S, Leijten J, Feinberg MW, Mandal BB, Liao R, Shin SR. Injectable Self-Oxygenating Cardio-Protective and Tissue Adhesive Silk-Based Hydrogel for Alleviating Ischemia After Mi Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2312261. [PMID: 38733225 PMCID: PMC11309903 DOI: 10.1002/smll.202312261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Indexed: 05/13/2024]
Abstract
Myocardial infarction (MI) is a significant cardiovascular disease that restricts blood flow, resulting in massive cell death and leading to stiff and noncontractile fibrotic scar tissue formation. Recently, sustained oxygen release in the MI area has shown regeneration ability; however, improving its therapeutic efficiency for regenerative medicine remains challenging. Here, a combinatorial strategy for cardiac repair by developing cardioprotective and oxygenating hybrid hydrogels that locally sustain the release of stromal cell-derived factor-1 alpha (SDF) and oxygen for simultaneous activation of neovascularization at the infarct area is presented. A sustained release of oxygen and SDF from injectable, mechanically robust, and tissue-adhesive silk-based hybrid hydrogels is achieved. Enhanced endothelialization under normoxia and anoxia is observed. Furthermore, there is a marked improvement in vascularization that leads to an increment in cardiomyocyte survival by ≈30% and a reduction of the fibrotic scar formation in an MI animal rodent model. Improved left ventricular systolic and diastolic functions by ≈10% and 20%, respectively, with a ≈25% higher ejection fraction on day 7 are also observed. Therefore, local delivery of therapeutic oxygenating and cardioprotective hydrogels demonstrates beneficial effects on cardiac functional recovery for reparative therapy.
Collapse
Affiliation(s)
- Shabir Hassan
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Zahra Rezaei
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Eder Luna
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Dilara Yilmaz-Aykut
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Chemical Engineering, Faculty of Engineering, Istanbul University-Cerrahpaşa, 34320 Istanbul, Turkey
| | - Myung Chul Lee
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ana Marie Perea
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Anurag Jamaiyar
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nicole Bassous
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Minoru Hirano
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Future Vehicle Research Department, Toyota Research Institute North America, Toyota Motor North America, Inc., 1555 Woodridge Ave., Ann Arbor, Michigan 48105, USA
| | - Fatima Mumtaza Tourk
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Cholong Choi
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Malin Becker
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Iman Yazdi
- School of Arts and Sciences, Regis College, 235 Wellesley Street, Weston, MA 02493, USA
| | - Kai Fan
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- BoYu Intelligent Health Innovation Laboratory, Hangzhou 311121, China
| | - Alan Avila-Ramirez
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
- Division of Biological and Environmental Science Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - David Ge
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital / Harvard Medical School, Boston, MA 02115, USA
| | - Sudeshna Fisch
- Cardiovascular Physiology Core, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jeroen Leijten
- Department of Developmental BioEngineering, Faculty of Science and Technology, Technical Medical Centre, University of Twente, Drienerlolaan 5, 7522 NB, Enschede, The Netherlands
| | - Mark W. Feinberg
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Ronglih Liao
- School of Medicine, Stanford University, California 94305-5101, USA
- Stanford Amyloid Center, Stanford University, California 94305-5101, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Brigham and Women’s Hospital, Department of Medicine, Harvard Medical School, Cambridge, MA 02139, USA
| |
Collapse
|
11
|
Chirikian O, Faynus MA, Merk M, Singh Z, Muray C, Pham J, Chialastri A, Vander Roest A, Goldstein A, Pyle T, Lane KV, Roberts B, Smith JE, Gunawardane RN, Sniadecki NJ, Mack DL, Davis J, Bernstein D, Streichan SJ, Clegg DO, Dey SS, Wilson MZ, Pruitt BL. YAP dysregulation triggers hypertrophy by CCN2 secretion and TGFβ uptake in human pluripotent stem cell-derived cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.03.597045. [PMID: 38895282 PMCID: PMC11185505 DOI: 10.1101/2024.06.03.597045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Hypertrophy Cardiomyopathy (HCM) is the most prevalent hereditary cardiovascular disease - affecting >1:500 individuals. Advanced forms of HCM clinically present with hypercontractility, hypertrophy and fibrosis. Several single-point mutations in b-myosin heavy chain (MYH7) have been associated with HCM and increased contractility at the organ level. Different MYH7 mutations have resulted in increased, decreased, or unchanged force production at the molecular level. Yet, how these molecular kinetics link to cell and tissue pathogenesis remains unclear. The Hippo Pathway, specifically its effector molecule YAP, has been demonstrated to be reactivated in pathological hypertrophic growth. We hypothesized that changes in force production (intrinsically or extrinsically) directly alter the homeostatic mechano-signaling of the Hippo pathway through changes in stresses on the nucleus. Using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), we asked whether homeostatic mechanical signaling through the canonical growth regulator, YAP, is altered 1) by changes in the biomechanics of HCM mutant cardiomyocytes and 2) by alterations in the mechanical environment. We use genetically edited hiPSC-CM with point mutations in MYH7 associated with HCM, and their matched controls, combined with micropatterned traction force microscopy substrates to confirm the hypercontractile phenotype in MYH7 mutants. We next modulate contractility in healthy and disease hiPSC-CMs by treatment with positive and negative inotropic drugs and demonstrate a correlative relationship between contractility and YAP activity. We further demonstrate the activation of YAP in both HCM mutants and healthy hiPSC-CMs treated with contractility modulators is through enhanced nuclear deformation. We conclude that the overactivation of YAP, possibly initiated and driven by hypercontractility, correlates with excessive CCN2 secretion (connective tissue growth factor), enhancing cardiac fibroblast/myofibroblast transition and production of known hypertrophic signaling molecule TGFβ. Our study suggests YAP being an indirect player in the initiation of hypertrophic growth and fibrosis in HCM. Our results provide new insights into HCM progression and bring forth a testbed for therapeutic options in treating HCM.
Collapse
|
12
|
Cordero J, Elsherbiny A, Wang Y, Jürgensen L, Constanty F, Günther S, Boerries M, Heineke J, Beisaw A, Leuschner F, Hassel D, Dobreva G. Leveraging chromatin state transitions for the identification of regulatory networks orchestrating heart regeneration. Nucleic Acids Res 2024; 52:4215-4233. [PMID: 38364861 PMCID: PMC11077086 DOI: 10.1093/nar/gkae085] [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: 05/26/2023] [Revised: 01/23/2024] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
The limited regenerative capacity of the human heart contributes to high morbidity and mortality worldwide. In contrast, zebrafish exhibit robust regenerative capacity, providing a powerful model for studying how to overcome intrinsic epigenetic barriers maintaining cardiac homeostasis and initiate regeneration. Here, we present a comprehensive analysis of the histone modifications H3K4me1, H3K4me3, H3K27me3 and H3K27ac during various stages of zebrafish heart regeneration. We found a vast gain of repressive chromatin marks one day after myocardial injury, followed by the acquisition of active chromatin characteristics on day four and a transition to a repressive state on day 14, and identified distinct transcription factor ensembles associated with these events. The rapid transcriptional response involves the engagement of super-enhancers at genes implicated in extracellular matrix reorganization and TOR signaling, while H3K4me3 breadth highly correlates with transcriptional activity and dynamic changes at genes involved in proteolysis, cell cycle activity, and cell differentiation. Using loss- and gain-of-function approaches, we identified transcription factors in cardiomyocytes and endothelial cells influencing cardiomyocyte dedifferentiation or proliferation. Finally, we detected significant evolutionary conservation between regulatory regions that drive zebrafish and neonatal mouse heart regeneration, suggesting that reactivating transcriptional and epigenetic networks converging on these regulatory elements might unlock the regenerative potential of adult human hearts.
Collapse
Affiliation(s)
- Julio Cordero
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Adel Elsherbiny
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yinuo Wang
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lonny Jürgensen
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Florian Constanty
- Institute of Experimental Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Stefan Günther
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
- German Cancer Consortium (DKTK), Partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, 69110 Heidelberg, Germany
| | - Joerg Heineke
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Arica Beisaw
- Institute of Experimental Cardiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Florian Leuschner
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - David Hassel
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| |
Collapse
|
13
|
Xu C, Zhang G, Wang X, Huang X, Zhang J, Han S, Wang J, Hall DD, Xu R, He F, Chang X, Wang F, Xie W, Wu Z, Song LS, Han P. Ptpn23 Controls Cardiac T-Tubule Patterning by Promoting the Assembly of Dystrophin-Glycoprotein Complex. Circulation 2024; 149:1375-1390. [PMID: 38214189 PMCID: PMC11039371 DOI: 10.1161/circulationaha.123.065767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/14/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Cardiac transverse tubules (T-tubules) are anchored to sarcomeric Z-discs by costameres to establish a regular spaced pattern. One of the major components of costameres is the dystrophin-glycoprotein complex (DGC). Nevertheless, how the assembly of the DGC coordinates with the formation and maintenance of T-tubules under physiological and pathological conditions remains unclear. METHODS Given the known role of Ptpn23 (protein tyrosine phosphatase, nonreceptor type 23) in regulating membrane deformation, its expression in patients with dilated cardiomyopathy was determined. Taking advantage of Cre/Loxp, CRISPR/Cas9, and adeno-associated virus 9 (AAV9)-mediated in vivo gene editing, we generated cardiomyocyte-specific Ptpn23 and Actn2 (α-actinin-2, a major component of Z-discs) knockout mice. We also perturbed the DGC by using dystrophin global knockout mice (DmdE4*). MM 4-64 and Di-8-ANEPPS staining, Cav3 immunofluorescence, and transmission electron microscopy were performed to determine T-tubule structure in isolated cells and intact hearts. In addition, the assembly of the DGC with Ptpn23 and dystrophin loss of function was determined by glycerol-gradient fractionation and SDS-PAGE analysis. RESULTS The expression level of Ptpn23 was reduced in failing hearts from dilated cardiomyopathy patients and mice. Genetic deletion of Ptpn23 resulted in disorganized T-tubules with enlarged diameters and progressive dilated cardiomyopathy without affecting sarcomere organization. AAV9-mediated mosaic somatic mutagenesis further indicated a cell-autonomous role of Ptpn23 in regulating T-tubule formation. Genetic and biochemical analyses showed that Ptpn23 was essential for the integrity of costameres, which anchor the T-tubule membrane to Z-discs, through interactions with α-actinin and dystrophin. Deletion of α-actinin altered the subcellular localization of Ptpn23 and DGCs. In addition, genetic inactivation of dystrophin caused similar T-tubule defects to Ptpn23 loss-of-function without affecting Ptpn23 localization at Z-discs. Last, inducible Ptpn23 knockout at 1 month of age showed Ptpn23 is also required for the maintenance of T-tubules in adult cardiomyocytes. CONCLUSIONS Ptpn23 is essential for cardiac T-tubule formation and maintenance along Z-discs. During postnatal heart development, Ptpn23 interacts with sarcomeric α-actinin and coordinates the assembly of the DGC at costameres to sculpt T-tubule spatial patterning and morphology.
Collapse
Affiliation(s)
- Chen Xu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Ge Zhang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Xinjian Wang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Xiaozhi Huang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Jiayin Zhang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Shuxian Han
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Jinxi Wang
- Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Duane D. Hall
- Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Ruoqing Xu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Feng He
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| | - Xing Chang
- Research Center for Industries of the Future, Westlake University, Hangzhou, China
| | - Fudi Wang
- The Fourth Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenjun Xie
- Department of Cardiology, First Affiliated Hospital of Xi'an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Zhichao Wu
- Department of Thoracic surgery, People’s hospital of Xinjiang Uyghur autonomous Region, Urumqi, Xinjiang, 830000
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510120
| | - Long-Sheng Song
- Department of Internal Medicine, Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Peidong Han
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Casarella S, Ferla F, Di Francesco D, Canciani E, Rizzi M, Boccafoschi F. Focal Adhesion's Role in Cardiomyocytes Function: From Cardiomyogenesis to Mechanotransduction. Cells 2024; 13:664. [PMID: 38667279 PMCID: PMC11049660 DOI: 10.3390/cells13080664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Mechanotransduction refers to the ability of cells to sense mechanical stimuli and convert them into biochemical signals. In this context, the key players are focal adhesions (FAs): multiprotein complexes that link intracellular actin bundles and the extracellular matrix (ECM). FAs are involved in cellular adhesion, growth, differentiation, gene expression, migration, communication, force transmission, and contractility. Focal adhesion signaling molecules, including Focal Adhesion Kinase (FAK), integrins, vinculin, and paxillin, also play pivotal roles in cardiomyogenesis, impacting cell proliferation and heart tube looping. In fact, cardiomyocytes sense ECM stiffness through integrins, modulating signaling pathways like PI3K/AKT and Wnt/β-catenin. Moreover, FAK/Src complex activation mediates cardiac hypertrophic growth and survival signaling in response to mechanical loads. This review provides an overview of the molecular and mechanical mechanisms underlying the crosstalk between FAs and cardiac differentiation, as well as the role of FA-mediated mechanotransduction in guiding cardiac muscle responses to mechanical stimuli.
Collapse
Affiliation(s)
- Simona Casarella
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Federica Ferla
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Dalila Di Francesco
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering, University Hospital Research Center, Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Elena Canciani
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Manuela Rizzi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (S.C.); (D.D.F.); (E.C.); (M.R.)
| |
Collapse
|
15
|
Bouhrira N, Vite A, Margulies KB. Distinct cytoskeletal regulators of mechanical memory in cardiac fibroblasts and cardiomyocytes. Basic Res Cardiol 2024; 119:277-289. [PMID: 38349539 DOI: 10.1007/s00395-023-01030-0] [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: 08/16/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 04/12/2024]
Abstract
Recognizing that cells "feel" and respond to their mechanical environment, recent studies demonstrate that many cells exhibit a phenomenon of "mechanical memory" in which features induced by prior mechanical cues persist after the mechanical stimulus has ceased. While there is a general recognition that different cell types exhibit different responses to changes in extracellular matrix stiffening, the phenomenon of mechanical memory within myocardial cell types has received little attention to date. To probe the dynamics of mechanical memory in cardiac fibroblasts (CFs) and cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CMs), we employed a magnetorheological elastomer (MRE) cell culture substrate with tunable and reversible stiffness spanning the range from normal to diseased myocardium. In CFs, using increased cell area and increases in α-smooth muscle actin as markers of cellular responses to matrix stiffening, we found that induction of mechanical memory required seven days of stiff priming. Both induction and maintenance of persistent CF activation were blocked with the F-actin inhibitor cytochalasin D, while inhibitors of microtubule detyrosination had no impact on CFs. In iPSC-CMs, mechanical memory was invoked after only 24 h of stiff priming. Moreover, mechanical memory induction and maintenance were microtubule-dependent in CMs with no dependence on F-actin. Overall, these results identify the distinct temporal dynamics of mechanical memory in CFs and iPSC-CMs with different cytoskeletal mediators responsible for inducing and maintaining the stiffness-activated phenotype. Due to its flexibility, this model is broadly applicable to future studies interrogating mechanotransduction and mechanical memory in the heart and might inform strategies for attenuating the impact of load-induced pathology and excess myocardial stiffness.
Collapse
Affiliation(s)
- Nesrine Bouhrira
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA
| | - Alexia Vite
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA
| | - Kenneth B Margulies
- Perelman School of Medicine, Cardiovascular Institute, University of Pennsylvania, 3400 Civic Center Boulevard, Smilow TRC 11-101, Philadelphia, PA, 19104, USA.
| |
Collapse
|
16
|
Allwood MA, Edgett BA, Platt MJ, Marrow JP, Coyle-Asbil B, Holjak EJB, Nelson VL, Bangali S, Alshamali R, Jacyniak K, Klein JM, Farquharson L, Romanova N, Northrup V, Ogilvie LM, Ayoub A, Ask K, Vickaryous MK, Hare GMT, Brunt KR, Simpson JA. Novel roles of cardiac-derived erythropoietin in cardiac development and function. J Mol Cell Cardiol 2024; 188:90-104. [PMID: 38382296 DOI: 10.1016/j.yjmcc.2024.01.006] [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: 08/28/2023] [Revised: 01/15/2024] [Accepted: 01/22/2024] [Indexed: 02/23/2024]
Abstract
The role of erythropoietin (EPO) has extended beyond hematopoiesis to include cytoprotection, inotropy, and neurogenesis. Extra-renal EPO has been reported for multiple tissue/cell types, but the physiological relevance remains unknown. Although the EPO receptor is expressed by multiple cardiac cell types and human recombinant EPO increases contractility and confers cytoprotection against injury, whether the heart produces physiologically meaningful amounts of EPO in vivo is unclear. We show a distinct circadian rhythm of cardiac EPO mRNA expression in adult mice and increased mRNA expression during embryogenesis, suggesting physiological relevance to cardiac EPO production throughout life. We then generated constitutive, cardiomyocyte-specific EPO knockout mice driven by the Mlc2v promoter (EPOfl/fl:Mlc2v-cre+/-; EPOΔ/Δ-CM). During cardiogenesis, cardiac EPO mRNA expression and cellular proliferation were reduced in EPOΔ/Δ-CM hearts. However, in adult EPOΔ/Δ- CM mice, total heart weight was preserved through increased cardiomyocyte cross-sectional area, indicating the reduced cellular proliferation was compensated for by cellular hypertrophy. Echocardiography revealed no changes in cardiac dimensions, with modest reductions in ejection fraction, stroke volume, and tachycardia, whereas invasive hemodynamics showed increased cardiac contractility and lusitropy. Paradoxically, EPO mRNA expression in the heart was elevated in adult EPOΔ/Δ-CM, along with increased serum EPO protein content and hematocrit. Using RNA fluorescent in situ hybridization, we found that Epo RNA colocalized with endothelial cells in the hearts of adult EPOΔ/Δ-CM mice, identifying the endothelial cells as a cell responsible for the EPO hyper-expression. Collectively, these data identify the first physiological roles for cardiomyocyte-derived EPO. We have established cardiac EPO mRNA expression is a complex interplay of multiple cell types, where loss of embryonic cardiomyocyte EPO production results in hyper-expression from other cells within the adult heart.
Collapse
Affiliation(s)
- Melissa A Allwood
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada
| | - Brittany A Edgett
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; Department of Pharmacology, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John, New Brunswick E2L 4L5, Canada; IMPART investigator Team, Canada; Department of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Mathew J Platt
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada
| | - Jade P Marrow
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada
| | - Bridget Coyle-Asbil
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada
| | - Emma J B Holjak
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada
| | - Victoria L Nelson
- Department of Pharmacology, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John, New Brunswick E2L 4L5, Canada; IMPART investigator Team, Canada
| | - Swara Bangali
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Razan Alshamali
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada
| | - Kathy Jacyniak
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Jorden M Klein
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Laura Farquharson
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Nadya Romanova
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Victoria Northrup
- Department of Pharmacology, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John, New Brunswick E2L 4L5, Canada; IMPART investigator Team, Canada
| | - Leslie M Ogilvie
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada
| | - Anmar Ayoub
- Firestone Institute for Respiratory Health, McMaster University, Hamilton, Ontario, Canada
| | - Kjetil Ask
- Firestone Institute for Respiratory Health, McMaster University, Hamilton, Ontario, Canada
| | - Matthew K Vickaryous
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada
| | - Gregory M T Hare
- Departments of Anesthesia & Physiology, St. Michel's Hospital, University of Toronto, Toronto, Ontario, Canada; IMPART investigator Team, Canada
| | - Keith R Brunt
- Department of Pharmacology, Dalhousie Medicine New Brunswick, 100 Tucker Park Road, Saint John, New Brunswick E2L 4L5, Canada; IMPART investigator Team, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, 50 Stone Road East, Guelph, ON N1G 2W1, Canada; IMPART investigator Team, Canada.
| |
Collapse
|
17
|
Hong MH, Jang YJ, Yoon JJ, Lee HS, Kim HY, Kang DG. Dohongsamul-tang inhibits cardiac remodeling and fibrosis through calcineurin/NFAT and TGF-β/Smad2 signaling in cardiac hypertrophy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116844. [PMID: 37453625 DOI: 10.1016/j.jep.2023.116844] [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: 03/26/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dohongsammul-tang (DH) is a Korean traditional herbal medicine used to alleviate symptoms caused by extravasated blood. It is known to protect against cardiovascular diseases and promote blood circulation by activating blood circulation to dispel blood stasis. The DH based on the characteristics of its medicinal properties has discovered the potential of alleviating cardiac hypertrophy. Therefore, this study was performed to verify the pharmacological effect of DH on improving cardiovascular disorders and to demonstrate its mutual improvement effect on renal function. Furthermore, aim of this study is founding the new potential beyond the traditional medicinal efficacy of DH, a traditional medicine. AIM OF THE STUDY In cardiovascular disease, cardiac hypertrophy refers to a change in the shape of the heart's structure due to pressure overload. It is known that an increase in myofibrils causes thickening of the heart, resulting in high blood pressure. Therefore, suppressing cardiac hypertrophy may be a major factor in lowering the morbidity, mortality, and heart failure associated with cardiovascular disease. Therefore, the study was performed to investigate whether DH, traditionally used, has effects on improving and alleviating cardiac injury and fibrosis caused by cardiac hypertrophy. MATERIALS AND METHODS Dohongsamul-tang was composed of 6 herbal medicine and each material were boiled with 4 L distilled water for 2 h. The mixture for dohongsamul-tang centrifuged at 3000 rpm for 10 min and concentrated. The concentrated dohongsamul-tang extraction freeze-dried and sotred at 70 °C. The powder of dohongsamul-tang was diluted with distilled water and administered orally. In this study, pressure overload was induced by tying the transverse aortic arch, which is connected to the left ventricle, to the thickness of a 27G needle by performing a surgical operation. The resulting cardiac hypertrophy and heart remodeling was induced and maintained for 8 weeks. RESULTS The study administered propranolol and dohongsamul-tang orally for 10 weeks to investigate their effects on cardiac hypertrophy induced by transverse aortic contraction (TAC) surgery. Results showed that TAC group increased the left ventricle weight and decreased cardiac function, but dohongsamul-tang treatment attenuated these effects. The pressure-volume curve experiment revealed that dohongsamul-tang improved cardiovascular function, which was worsened by TAC group. Dohongsamul-tang treatment also downregulated collagen I and III through the TGF-β/Smad2 signaling pathway and improved hematological biomarkers of cardiac hypertrophy. In addition, dohongsamul-tang treatment improved renal function-related biomarkers, such as blood creatinine, blood urea nitrogen, and neutrophil gelatinase-associated lipocalin, which were increased by TAC-induced cardiac hypertrophy. CONCLUSIONS Taken together, dohongsamul-tang treatment inhibited cardiac remodeling due to pressure overload in the TAC-induced cardiac hypertrophy model, and this effect is thought to be manifested by improving the functional and morphological changes through the calcineurin/NFATc4 and reducing the cardiac fibrosis by suppressing TGF-β/Smad2 signaling pathways.
Collapse
Affiliation(s)
- Mi Hyeon Hong
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Youn Jae Jang
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Jung Joo Yoon
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Ho Sub Lee
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Hye Yoom Kim
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| | - Dae Gill Kang
- Hanbang Cardio-renal Research Center & Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea; College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, South Korea.
| |
Collapse
|
18
|
Osten F, Weber N, Wendland M, Holler T, Piep B, Kröhn S, Teske J, Bodenschatz AK, Devadas SB, Menge KS, Chatterjee S, Schwanke K, Kosanke M, Montag J, Thum T, Zweigerdt R, Kraft T, Iorga B, Meissner JD. Myosin expression and contractile function are altered by replating stem cell-derived cardiomyocytes. J Gen Physiol 2023; 155:e202313377. [PMID: 37656049 PMCID: PMC10473967 DOI: 10.1085/jgp.202313377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 09/02/2023] Open
Abstract
Myosin heavy chain (MyHC) is the main determinant of contractile function. Human ventricular cardiomyocytes (CMs) predominantly express the β-isoform. We previously demonstrated that ∼80% of human embryonic stem cell-derived cardiomyocytes (hESC-CMs) express exclusively β-MyHC after long-term culture on laminin-coated glass coverslips. Here, we investigated the impact of enzymatically detaching hESC-CMs after long-term culture and subsequently replating them for characterization of cellular function. We observed that force-related kinetic parameters, as measured in a micromechanical setup, resembled α- rather than β-MyHC-expressing myofibrils, as well as changes in calcium transients. Single-cell immunofluorescence analysis revealed that replating hESC-CMs led to rapid upregulation of α-MyHC, as indicated by increases in exclusively α-MyHC- and in mixed α/β-MyHC-expressing hESC-CMs. A comparable increase in heterogeneity of MyHC isoform expression was also found among individual human induced pluripotent stem cell (hiPSC)-derived CMs after replating. Changes in MyHC isoform expression and cardiomyocyte function induced by replating were reversible in the course of the second week after replating. Gene enrichment analysis based on RNA-sequencing data revealed changes in the expression profile of mechanosensation/-transduction-related genes and pathways, especially integrin-associated signaling. Accordingly, the integrin downstream mediator focal adhesion kinase (FAK) promoted β-MyHC expression on a stiff matrix, further validating gene enrichment analysis. To conclude, detachment and replating induced substantial changes in gene expression, MyHC isoform composition, and function of long-term cultivated human stem cell-derived CMs, thus inducing alterations in mechanosensation/-transduction, that need to be considered, particularly for downstream in vitro assays.
Collapse
Affiliation(s)
- Felix Osten
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Natalie Weber
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Meike Wendland
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Tim Holler
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Birgit Piep
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Simon Kröhn
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Jana Teske
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Alea K. Bodenschatz
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Santoshi Biswanath Devadas
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Kaja S. Menge
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Shambhabi Chatterjee
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Kristin Schwanke
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Maike Kosanke
- Research Core Unit Genomics, Hannover Medical School, Hannover, Germany
| | - Judith Montag
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- REBIRTH Center for Translational Regenerative Therapies, Hannover Medical School, Hannover, Germany
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Robert Zweigerdt
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany
| | - Theresia Kraft
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| | - Bogdan Iorga
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
- Department of Analytical Chemistry and Physical Chemistry, Faculty of Chemistry, University of Bucharest, Bucharest, Romania
| | - Joachim D. Meissner
- Institute of Molecular and Cell Physiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
19
|
Narayanan G, Halim A, Hu A, Avin KG, Lu T, Zehnder D, Hato T, Chen NX, Moe SM, Lim K. Molecular Phenotyping and Mechanisms of Myocardial Fibrosis in Advanced Chronic Kidney Disease. KIDNEY360 2023; 4:1562-1579. [PMID: 37858297 PMCID: PMC10695648 DOI: 10.34067/kid.0000000000000276] [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: 02/25/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
Key Points Myocardial fibrosis in hearts from patients with CKD is characterized by increased trimeric tensile collagen type I and decreased elastic collagen type III compared with hearts from hypertensive or healthy donors, suggesting a unique fibrotic phenotype. Myocardial fibrosis in CKD is driven by alterations in extracellular matrix proteostasis, including dysregulation of metalloproteinases and cross-linking enzymes. CKD-associated mineral stressors uniquely induce a fibronectin-independent mechanism of fibrillogenesis characterized by formation of trimeric collagen compared with proinflammatory/fibrotic cytokines. Background Myocardial fibrosis is a major life-limiting problem in CKD. Despite this, the molecular phenotype and metabolism of collagen fibrillogenesis in fibrotic hearts of patients with advanced CKD have been largely unstudied. Methods We analyzed explanted human left ventricular (LV) heart tissues in a three-arm cross-sectional cohort study of deceased donor patients on hemodialysis (HD, n =18), hypertension with preserved renal function (HTN, n =8), and healthy controls (CON, n =17), ex vivo . RNA-seq and protein analysis was performed on human donor hearts and cardiac fibroblasts treated with mineral stressors (high phosphate and high calcium). Further mechanistic studies were performed using primary cardiac fibroblasts, in vitro treated with mineral stressors, proinflammatory and profibrotic cytokines. Results Of the 43 donor participants, there was no difference in age (P > 0.2), sex (P > 0.8), or body mass index (P > 0.1) between the groups. Hearts from the HD group had extensive fibrosis (P < 0.01). All LV tissues expressed only the trimeric form of collagen type I. HD hearts expressed increased collagen type I (P < 0.03), elevated collagen type I:III ratio (P < 0.05), and decreased MMP1 (P < 0.05) and MMP2 (P < 0.05). RNA-seq revealed no significant differential gene expression of extracellular matrix proteins of interest in HD hearts, but there was significant upregulation of LH2, periostin, α -SMA, and TGF-β 1 gene expression in mineral stressor–treated cardiac fibroblasts. Both mineral stressors (P < 0.009) and cytokines (P < 0.03) increased collagen type I:III ratio. Mineral stressors induced trimeric collagen type I, but cytokine treatment induced only dimeric collagen type I in cardiac fibroblasts. Mineral stressors downregulated fibronectin (P < 0.03) and MMP2 zymogen (P < 0.01) but did not significantly affect expression of periostin, MMP1, or cross-linking enzymes. TGF-β upregulated fibronectin (P < 0.01) and periostin (P < 0.02) only. Conclusions Myocardial fibrosis in advanced CKD hearts is characterized by increased trimeric collagen type I and dysregulated collagen metabolism, and is differentially regulated by components of uremia.
Collapse
Affiliation(s)
- Gayatri Narayanan
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
| | - Arvin Halim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
| | - Alvin Hu
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Medicine, Indiana University Health Ball Memorial Hospital, Indianapolis, Indiana
| | - Keith G. Avin
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Physical Therapy, Indiana University School of Health and Human Sciences, Indiana University, Indianapolis, Indiana
| | - Tzongshi Lu
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel Zehnder
- Department of Nephrology and Department of Acute Medicine, North Cumbria University Hospital NHS Trust, Carlisle, United Kingdom
| | - Takashi Hato
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
| | - Neal X. Chen
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
| | - Sharon M. Moe
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
| | - Kenneth Lim
- Division of Nephrology and Hypertension, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
20
|
Lamberto F, Shashikadze B, Elkhateib R, Lombardo SD, Horánszky A, Balogh A, Kistamás K, Zana M, Menche J, Fröhlich T, Dinnyés A. Low-dose Bisphenol A exposure alters the functionality and cellular environment in a human cardiomyocyte model. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 335:122359. [PMID: 37567409 DOI: 10.1016/j.envpol.2023.122359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/26/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023]
Abstract
Early embryonic development represents a sensitive time-window during which the foetus might be vulnerable to the exposure of environmental contaminants, potentially leading to heart diseases also later in life. Bisphenol A (BPA), a synthetic chemical widely used in plastics manufacturing, has been associated with heart developmental defects, even in low concentrations. This study aims to investigate the effects of environmentally relevant doses of BPA on developing cardiomyocytes using a human induced pluripotent stem cell (hiPSC)-derived model. Firstly, a 2D in vitro differentiation system to obtain cardiomyocytes from hiPSCs (hiPSC-CMs) have been established and characterised to provide a suitable model for the early stages of cardiac development. Then, the effects of a repeated BPA exposure, starting from the undifferentiated stage throughout the differentiation process, were evaluated. The chemical significantly decreased the beat rate of hiPSC-CMs, extending the contraction and relaxation time in a dose-dependent manner. Quantitative proteomics analysis revealed a high abundance of basement membrane (BM) components (e.g., COL4A1, COL4A2, LAMC1, NID2) and a significant increase in TNNC1 and SERBP1 proteins in hiPSC-CMs treated with BPA. Network analysis of proteomics data supported altered extracellular matrix remodelling and provided a disease-gene association with well-known pathological conditions of the heart. Furthermore, upon hypoxia-reoxygenation challenge, hiPSC-CMs treated with BPA showed higher rate of apoptotic events. Taken together, our results revealed that a long-term treatment, even with low doses of BPA, interferes with hiPSC-CMs functionality and alters the surrounding cellular environment, providing new insights about diseases that might arise upon the toxin exposure. Our study contributes to the current understanding of BPA effects on developing human foetal cardiomyocytes, in correlation with human clinical observations and animal studies, and it provides a suitable model for New Approach Methodologies (NAMs) for environmental chemical hazard and risk assessment.
Collapse
Affiliation(s)
- Federica Lamberto
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary; Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, H-2100, Gödöllő, Hungary
| | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377, Munich, Germany
| | - Radwa Elkhateib
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377, Munich, Germany
| | - Salvo Danilo Lombardo
- Max Perutz Labs, Vienna Biocenter Campus (VBC), 1030, Vienna, Austria; Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, 1030, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria
| | - Alex Horánszky
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary; Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, H-2100, Gödöllő, Hungary
| | - Andrea Balogh
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary
| | - Kornél Kistamás
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary
| | - Melinda Zana
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary
| | - Jörg Menche
- Max Perutz Labs, Vienna Biocenter Campus (VBC), 1030, Vienna, Austria; Department of Structural and Computational Biology, Center for Molecular Biology, University of Vienna, 1030, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090, Vienna, Austria; Faculty of Mathematics, University of Vienna, 1090, Vienna, Austria
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377, Munich, Germany
| | - András Dinnyés
- BioTalentum Ltd., Aulich Lajos Str. 26, Gödöllő, H-2100, Hungary; Department of Physiology and Animal Health, Institute of Physiology and Animal Nutrition, Hungarian University of Agriculture and Life Sciences, Páter Károly Str. 1, H-2100, Gödöllő, Hungary; Department of Cell Biology and Molecular Medicine, University of Szeged, H-6720, Szeged, Hungary.
| |
Collapse
|
21
|
Brown SJ, Šoltić D, Synowsky SA, Shirran SL, Chilcott E, Shorrock HK, Gillingwater TH, Yáñez-Muñoz RJ, Schneider B, Bowerman M, Fuller HR. AAV9-mediated SMN gene therapy rescues cardiac desmin but not lamin A/C and elastin dysregulation in Smn2B/- spinal muscular atrophy mice. Hum Mol Genet 2023; 32:2950-2965. [PMID: 37498175 PMCID: PMC10549791 DOI: 10.1093/hmg/ddad121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/27/2023] [Accepted: 07/25/2023] [Indexed: 07/28/2023] Open
Abstract
Structural, functional and molecular cardiac defects have been reported in spinal muscular atrophy (SMA) patients and mouse models. Previous quantitative proteomics analyses demonstrated widespread molecular defects in the severe Taiwanese SMA mouse model. Whether such changes are conserved across different mouse models, including less severe forms of the disease, has yet to be established. Here, using the same high-resolution proteomics approach in the less-severe Smn2B/- SMA mouse model, 277 proteins were found to be differentially abundant at a symptomatic timepoint (post-natal day (P) 18), 50 of which were similarly dysregulated in severe Taiwanese SMA mice. Bioinformatics analysis linked many of the differentially abundant proteins to cardiovascular development and function, with intermediate filaments highlighted as an enriched cellular compartment in both datasets. Lamin A/C was increased in the cardiac tissue, whereas another intermediate filament protein, desmin, was reduced. The extracellular matrix (ECM) protein, elastin, was also robustly decreased in the heart of Smn2B/- mice. AAV9-SMN1-mediated gene therapy rectified low levels of survival motor neuron protein and restored desmin levels in heart tissues of Smn2B/- mice. In contrast, AAV9-SMN1 therapy failed to correct lamin A/C or elastin levels. Intermediate filament proteins and the ECM have key roles in cardiac function and their dysregulation may explain cardiac impairment in SMA, especially since mutations in genes encoding these proteins cause other diseases with cardiac aberration. Cardiac pathology may need to be considered in the long-term care of SMA patients, as it is unclear whether currently available treatments can fully rescue peripheral pathology in SMA.
Collapse
Affiliation(s)
- Sharon J Brown
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Darija Šoltić
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| | - Silvia A Synowsky
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Sally L Shirran
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, St Andrews KY16 9ST, UK
| | - Ellie Chilcott
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Hannah K Shorrock
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Rafael J Yáñez-Muñoz
- AGCTlab.org, Centre of Gene and Cell Therapy, Centre for Biomedical Sciences, Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham Hill, Egham, Surrey TW20 0EX, UK
| | - Bernard Schneider
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Melissa Bowerman
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
- School of Medicine, Keele University, Keele ST5 5BG, UK
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Keele ST5 5BG, UK
- Wolfson Centre for Inherited Neuromuscular Disease, TORCH Building, RJAH Orthopaedic Hospital, Oswestry SY10 7AG, UK
| |
Collapse
|
22
|
Farrugia BL, Melrose J. The Glycosaminoglycan Side Chains and Modular Core Proteins of Heparan Sulphate Proteoglycans and the Varied Ways They Provide Tissue Protection by Regulating Physiological Processes and Cellular Behaviour. Int J Mol Sci 2023; 24:14101. [PMID: 37762403 PMCID: PMC10531531 DOI: 10.3390/ijms241814101] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/03/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
This review examines the roles of HS-proteoglycans (HS-PGs) in general, and, in particular, perlecan and syndecan as representative examples and their interactive ligands, which regulate physiological processes and cellular behavior in health and disease. HS-PGs are essential for the functional properties of tissues both in development and in the extracellular matrix (ECM) remodeling that occurs in response to trauma or disease. HS-PGs interact with a biodiverse range of chemokines, chemokine receptors, protease inhibitors, and growth factors in immune regulation, inflammation, ECM stabilization, and tissue protection. Some cell regulatory proteoglycan receptors are dually modified hybrid HS/CS proteoglycans (betaglycan, CD47). Neurexins provide synaptic stabilization, plasticity, and specificity of interaction, promoting neurotransduction, neurogenesis, and differentiation. Ternary complexes of glypican-1 and Robbo-Slit neuroregulatory proteins direct axonogenesis and neural network formation. Specific neurexin-neuroligin complexes stabilize synaptic interactions and neural activity. Disruption in these interactions leads to neurological deficits in disorders of functional cognitive decline. Interactions with HS-PGs also promote or inhibit tumor development. Thus, HS-PGs have complex and diverse regulatory roles in the physiological processes that regulate cellular behavior and the functional properties of normal and pathological tissues. Specialized HS-PGs, such as the neurexins, pikachurin, and Eyes-shut, provide synaptic stabilization and specificity of neural transduction and also stabilize the axenome primary cilium of phototoreceptors and ribbon synapse interactions with bipolar neurons of retinal neural networks, which are essential in ocular vision. Pikachurin and Eyes-Shut interactions with an α-dystroglycan stabilize the photoreceptor synapse. Novel regulatory roles for HS-PGs controlling cell behavior and tissue function are expected to continue to be uncovered in this fascinating class of proteoglycan.
Collapse
Affiliation(s)
- Brooke L. Farrugia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, VIC 3010, Australia;
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Raymond Purves Laboratory of Bone and Joint Research, Kolling Institute of Medical Research, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
- Sydney Medical School (Northern), University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
23
|
Moazzen H, Bolaji MD, Leube RE. Desmosomes in Cell Fate Determination: From Cardiogenesis to Cardiomyopathy. Cells 2023; 12:2122. [PMID: 37681854 PMCID: PMC10487268 DOI: 10.3390/cells12172122] [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: 07/18/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Desmosomes play a vital role in providing structural integrity to tissues that experience significant mechanical tension, including the heart. Deficiencies in desmosomal proteins lead to the development of arrhythmogenic cardiomyopathy (AC). The limited availability of preventative measures in clinical settings underscores the pressing need to gain a comprehensive understanding of desmosomal proteins not only in cardiomyocytes but also in non-myocyte residents of the heart, as they actively contribute to the progression of cardiomyopathy. This review focuses specifically on the impact of desmosome deficiency on epi- and endocardial cells. We highlight the intricate cross-talk between desmosomal proteins mutations and signaling pathways involved in the regulation of epicardial cell fate transition. We further emphasize that the consequences of desmosome deficiency differ between the embryonic and adult heart leading to enhanced erythropoiesis during heart development and enhanced fibrogenesis in the mature heart. We suggest that triggering epi-/endocardial cells and fibroblasts that are in different "states" involve the same pathways but lead to different pathological outcomes. Understanding the details of the different responses must be considered when developing interventions and therapeutic strategies.
Collapse
Affiliation(s)
- Hoda Moazzen
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany; (M.D.B.); (R.E.L.)
| | | | | |
Collapse
|
24
|
Glavaški M, Velicki L, Vučinić N. Hypertrophic Cardiomyopathy: Genetic Foundations, Outcomes, Interconnections, and Their Modifiers. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1424. [PMID: 37629714 PMCID: PMC10456451 DOI: 10.3390/medicina59081424] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent heritable cardiomyopathy. HCM is considered to be caused by mutations in cardiac sarcomeric protein genes. Recent research suggests that the genetic foundation of HCM is much more complex than originally postulated. The clinical presentations of HCM are very variable. Some mutation carriers remain asymptomatic, while others develop severe HCM, terminal heart failure, or sudden cardiac death. Heterogeneity regarding both genetic mutations and the clinical course of HCM hinders the establishment of universal genotype-phenotype correlations. However, some trends have been identified. The presence of a mutation in some genes encoding sarcomeric proteins is associated with earlier HCM onset, more severe left ventricular hypertrophy, and worse clinical outcomes. There is a diversity in the mechanisms implicated in the pathogenesis of HCM. They may be classified into groups, but they are interrelated. The lack of known supplementary elements that control the progression of HCM indicates that molecular mechanisms that exist between genotype and clinical presentations may be crucial. Secondary molecular changes in pathways implicated in HCM pathogenesis, post-translational protein modifications, and epigenetic factors affect HCM phenotypes. Cardiac loading conditions, exercise, hypertension, diet, alcohol consumption, microbial infection, obstructive sleep apnea, obesity, and environmental factors are non-molecular aspects that change the HCM phenotype. Many mechanisms are implicated in the course of HCM. They are mostly interconnected and contribute to some extent to final outcomes.
Collapse
Affiliation(s)
- Mila Glavaški
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| | - Lazar Velicki
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
- Institute of Cardiovascular Diseases Vojvodina, Put Doktora Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Nataša Vučinić
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| |
Collapse
|
25
|
Johnson CD, Aranda-Espinoza H, Fisher JP. A Case for Material Stiffness as a Design Parameter in Encapsulated Islet Transplantation. TISSUE ENGINEERING. PART B, REVIEWS 2023; 29:334-346. [PMID: 36475851 PMCID: PMC10442690 DOI: 10.1089/ten.teb.2022.0157] [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: 08/26/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Diabetes is a disease that plagues over 463 million people globally. Approximately 40 million of these patients have type 1 diabetes mellitus (T1DM), and the global incidence is increasing by up to 5% per year. T1DM is where the body's immune system attacks the pancreas, specifically the pancreatic beta cells, with antibodies to prevent insulin production. Although current treatments such as exogenous insulin injections have been successful, exorbitant insulin costs and meticulous administration present the need for alternative long-term solutions to glucose dysregulation caused by diabetes. Encapsulated islet transplantation (EIT) is a tissue-engineered solution to diabetes. Donor islets are encapsulated in a semipermeable hydrogel, allowing the diffusion of oxygen, glucose, and insulin but preventing leukocyte infiltration and antibody access to the transplanted cells. Although successful in small animal models, EIT is still far from commercial use owing to necessary long-term systemic immunosuppressants and consistent immune rejection. Most published research has focused on tailoring the characteristics of the capsule material to promote clinical viability. However, most studies have been limited in scope to biochemical changes. Current mechanobiology studies on the effect of substrate stiffness on the function of leukocytes, especially macrophages-primary foreign body response (FBR) orchestrators, show promise in tailoring a favorable response to tissue-engineered therapies such as EIT. In this review, we explore strategies to improve the clinical viability of EIT. A brief overview of the immune system, the FBR, and current biochemical approaches will be elucidated throughout this exploration. Furthermore, an argument for using substrate stiffness as a capsule design parameter to increase EIT efficacy and clinical viability will be posed.
Collapse
Affiliation(s)
- Courtney D. Johnson
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| | - Helim Aranda-Espinoza
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
26
|
Young KM, Reinhart-King CA. Cellular mechanosignaling for sensing and transducing matrix rigidity. Curr Opin Cell Biol 2023; 83:102208. [PMID: 37473514 PMCID: PMC10527818 DOI: 10.1016/j.ceb.2023.102208] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
The mechanisms by which cells sense their mechanical environment and transduce the signal through focal adhesions and signaling pathways to the nucleus is an area of key focus for the field of mechanobiology. In the past two years, there has been expansion of our knowledge of commonly studied pathways, such as YAP/TAZ, FAK/Src, RhoA/ROCK, and Piezo1 signaling, as well as the discovery of new interactions, such as the effect of matrix rigidity of cell mitochondrial function and metabolism, which represent a new and exciting direction for the field as a whole. This review covers the most recent advances in the field of substrate stiffness sensing as well as perspective on future directions.
Collapse
Affiliation(s)
- Katherine M Young
- Vanderbilt University Department of Biomedical Engineering 2414 Highland Ave, Nashville, TN 37212, USA
| | - Cynthia A Reinhart-King
- Vanderbilt University Department of Biomedical Engineering 2414 Highland Ave, Nashville, TN 37212, USA.
| |
Collapse
|
27
|
Joshua J, Caswell JL, Monné Rodriguez JM, Kipar A, O'Sullivan ML, Wood G, Fonfara S. MicroRNA profiling of the feline left heart identifies chamber-specific expression signatures in health and in advanced hypertrophic cardiomyopathy. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2023; 4:100037. [PMID: 39801693 PMCID: PMC11708362 DOI: 10.1016/j.jmccpl.2023.100037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 01/16/2025]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common heart disease in humans and cats, nonetheless, the disease pathogenesis is still poorly understood. MicroRNAs are suspected to be involved in the disease process but the myocardial microRNA expression pattern in cats has not been identified. We hypothesized that microRNA profiles differ between healthy cats and cats with HCM. Small RNA sequencing on left ventricle (LV) and left atria (LA) samples from healthy cats (8 LV, 8 LA) and cats with HCM (7 LV, 5 LA) was performed. We identified 1039 differentially expressed microRNAs (False Discovery Rate <0.01, fold change >2). Cats with HCM were found to have a distinct microRNA expression profile with apparent regional heterogeneity. Comparing the HCM and control hearts, we detected 80 differentially expressed microRNAs for the HCM LV, and 37 for the LA. These included LV and LA enriched miR-21, miR-146b, and reduced miR-122-5p, which were recently suggested as key microRNAs for the HCM pathogenesis, and miR-132, which might be of therapeutic interest. Several top enriched microRNAs: miR-3958, miR-382-5p, miR-487a-5p (HCM LV); miR-chrD4_30107-3p (HCM LA); miR-3548 (HCM LV and LA) have either not been reported in the heart or only little is known. We identified potentially relevant microRNAs and further investigations into their role are required. Genes known to be targeted by the differentially expressed microRNAs were associated with inflammation and growth pathways in the HCM LV and LA, cardioprotective pathways in the LV, and fibrosis and structural changes in the LA when compared to healthy hearts.
Collapse
Affiliation(s)
- Jessica Joshua
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph N1G 2W1, Ontario, Canada
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph N1G 2W1, Ontario, Canada
| | - Jeff L. Caswell
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph N1G 2W1, Ontario, Canada
| | - Josep M. Monné Rodriguez
- University of Zurich, Vetsuisse Faculty, Institute of Veterinary Pathology, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - Anja Kipar
- University of Zurich, Vetsuisse Faculty, Institute of Veterinary Pathology, Winterthurerstrasse 268, CH-8057 Zurich, Switzerland
| | - M. Lynne O'Sullivan
- University of Prince Edward Island, Department of Companion Animals, Charlottetown C1A 4P3, Prince Edward Island, Canada
| | - Geoffrey Wood
- University of Guelph, Ontario Veterinary College, Department of Pathobiology, Guelph N1G 2W1, Ontario, Canada
| | - Sonja Fonfara
- University of Guelph, Ontario Veterinary College, Department of Clinical Studies, Guelph N1G 2W1, Ontario, Canada
| |
Collapse
|
28
|
Nattasit P, Niibe K, Yamada M, Ohori-Morita Y, Limraksasin P, Tiskratok W, Yamamoto M, Egusa H. Stiffness-Tunable Hydrogel-Sandwich Culture Modulates the YAP-Mediated Mechanoresponse in Induced-Pluripotent Stem Cell Embryoid Bodies and Augments Cardiomyocyte Differentiation. Macromol Biosci 2023:e2300021. [PMID: 36871184 DOI: 10.1002/mabi.202300021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Indexed: 03/06/2023]
Abstract
Microenvironmental factors, including substrate stiffness, regulate stem cell behavior and differentiation. However, the effects of substrate stiffness on the behavior of induced pluripotent stem cell (iPSC)- derived embryoid bodies (EB) remain unclear. To investigate the effects of mechanical cues on iPSC-EB differentiation, a 3D hydrogel-sandwich culture (HGSC) system is developed that controls the microenvironment surrounding iPSC-EBs using a stiffness-tunable polyacrylamide hydrogel assembly. Mouse iPSC-EBs are seeded between upper and lower polyacrylamide hydrogels of differing stiffness (Young's modulus [E'] = 54.3 ± 7.1 kPa [hard], 28.1 ± 2.3 kPa [moderate], and 5.1 ± 0.1 kPa [soft]) and cultured for 2 days. HGSC induces stiffness-dependent activation of the yes-associated protein (YAP) mechanotransducer and actin cytoskeleton rearrangement in the iPSC-EBs. Moreover, moderate-stiffness HGSC specifically upregulates the mRNA and protein expression of ectoderm and mesoderm lineage differentiation markers in iPSC-EBs via YAP-mediated mechanotransduction. Pretreatment of mouse iPSC-EBs with moderate-stiffness HGSC promotes cardiomyocyte (CM) differentiation and structural maturation of myofibrils. The proposed HGSC system provides a viable platform for investigating the role of mechanical cues on the pluripotency and differentiation of iPSCs that can be beneficial for research into tissue regeneration and engineering.
Collapse
Affiliation(s)
- Praphawi Nattasit
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Kunimichi Niibe
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Masahiro Yamada
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Yumi Ohori-Morita
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| | - Phoonsuk Limraksasin
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
- Dental Stem Cell Biology Research Unit, Center of Excellence for Regenerative Dentistry, and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Watcharaphol Tiskratok
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima, 30000, Thailand
| | - Masaya Yamamoto
- Department of Material Processing, Tohoku University Graduate School of Engineering, Sendai, Miyagi, 980-8579, Japan
| | - Hiroshi Egusa
- Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
- Center for Advanced Stem Cell and Regenerative Research, Tohoku University Graduate School of Dentistry, Sendai, Miyagi, 980-8575, Japan
| |
Collapse
|
29
|
Wang C, Ramahdita G, Genin G, Huebsch N, Ma Z. Dynamic mechanobiology of cardiac cells and tissues: Current status and future perspective. BIOPHYSICS REVIEWS 2023; 4:011314. [PMID: 37008887 PMCID: PMC10062054 DOI: 10.1063/5.0141269] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023]
Abstract
Mechanical forces impact cardiac cells and tissues over their entire lifespan, from development to growth and eventually to pathophysiology. However, the mechanobiological pathways that drive cell and tissue responses to mechanical forces are only now beginning to be understood, due in part to the challenges in replicating the evolving dynamic microenvironments of cardiac cells and tissues in a laboratory setting. Although many in vitro cardiac models have been established to provide specific stiffness, topography, or viscoelasticity to cardiac cells and tissues via biomaterial scaffolds or external stimuli, technologies for presenting time-evolving mechanical microenvironments have only recently been developed. In this review, we summarize the range of in vitro platforms that have been used for cardiac mechanobiological studies. We provide a comprehensive review on phenotypic and molecular changes of cardiomyocytes in response to these environments, with a focus on how dynamic mechanical cues are transduced and deciphered. We conclude with our vision of how these findings will help to define the baseline of heart pathology and of how these in vitro systems will potentially serve to improve the development of therapies for heart diseases.
Collapse
Affiliation(s)
| | - Ghiska Ramahdita
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, USA
| | | | | | - Zhen Ma
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
30
|
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment. Int J Mol Sci 2023; 24:ijms24032444. [PMID: 36768766 PMCID: PMC9916732 DOI: 10.3390/ijms24032444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.
Collapse
|
31
|
Garcia-Padilla C, Lozano-Velasco E, Garcia-Lopez V, Aranega A, Franco D, Garcia-Martinez V, Lopez-Sanchez C. Comparative Analysis of Non-Coding RNA Transcriptomics in Heart Failure. Biomedicines 2022; 10:3076. [PMID: 36551832 PMCID: PMC9775550 DOI: 10.3390/biomedicines10123076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Heart failure constitutes a clinical complex syndrome with different symptomatic characteristics depending on age, sex, race and ethnicity, among others, which has become a major public health issue with an increasing prevalence. One of the most interesting tools seeking to improve prevention, diagnosis, treatment and prognosis of this pathology has focused on finding new molecular biomarkers since heart failure relies on deficient cardiac homeostasis, which is regulated by a strict gene expression. Therefore, currently, analyses of non-coding RNA transcriptomics have been oriented towards human samples. The present review develops a comparative study emphasizing the relevance of microRNAs, long non-coding RNAs and circular RNAs as potential biomarkers in heart failure. Significantly, further studies in this field of research are fundamental to supporting their widespread clinical use. In this sense, the various methodologies used by the authors should be standardized, including larger cohorts, homogeneity of the samples and uniformity of the bioinformatic pipelines used to reach stratification and statistical significance of the results. These basic adjustments could provide promising steps to designing novel strategies for clinical management of patients with heart failure.
Collapse
Affiliation(s)
- Carlos Garcia-Padilla
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
| | - Estefanía Lozano-Velasco
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Virginio Garcia-Lopez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Amelia Aranega
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Diego Franco
- Department of Experimental Biology, University of Jaen, 23071 Jaen, Spain
- Medina Foundation, 18016 Granada, Spain
| | - Virginio Garcia-Martinez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| | - Carmen Lopez-Sanchez
- Department of Human Anatomy and Embryology, Faculty of Medicine, Institute of Molecular Pathology Biomarkers, University of Extremadura, 06006 Badajoz, Spain
| |
Collapse
|
32
|
Mohammadi MH, Okhovatian S, Savoji H, Campbell SB, Lai BFL, Wu J, Pascual-Gil S, Bannerman D, Rafatian N, Li RK, Radisic M. Toward Hierarchical Assembly of Aligned Cell Sheets into a Conical Cardiac Ventricle Using Microfabricated Elastomers. Adv Biol (Weinh) 2022; 6:e2101165. [PMID: 35798316 PMCID: PMC9691564 DOI: 10.1002/adbi.202101165] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 05/31/2022] [Indexed: 01/28/2023]
Abstract
Despite current efforts in organ-on-chip engineering to construct miniature cardiac models, they often lack some physiological aspects of the heart, including fiber orientation. This motivates the development of bioartificial left ventricle models that mimic the myofiber orientation of the native ventricle. Herein, an approach relying on microfabricated elastomers that enables hierarchical assembly of 2D aligned cell sheets into a functional conical cardiac ventricle is described. Soft lithography and injection molding techniques are used to fabricate micro-grooves on an elastomeric polymer scaffold with three different orientations ranging from -60° to +60°, each on a separate trapezoidal construct. The width of the micro-grooves is optimized to direct the majority of cells along the groove direction and while periodic breaks are used to promote cell-cell contact. The scaffold is wrapped around a central mandrel to obtain a conical-shaped left ventricle model inspired by the size of a human left ventricle 19 weeks post-gestation. Rectangular micro-scale holes are incorporated to alleviate oxygen diffusional limitations within the 3D scaffold. Cardiomyocytes within the 3D left ventricle constructs showed high viability in all layers after 7 days of cultivation. The hierarchically assembled left ventricle also provided functional readouts such as calcium transients and ejection fraction.
Collapse
Affiliation(s)
| | - Sargol Okhovatian
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Sainte Justine University Hospital Research Center, Montreal TransMedTech Institute, Montreal, Quebec, Canada
| | - Scott B. Campbell
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Benjamin Fook Lun Lai
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Simon Pascual-Gil
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Dawn Bannerman
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Naimeh Rafatian
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Ren-Ke Li
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Surgery, Division of Cardiovascular Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Menasché P. Immunothérapie par CAR-T cells : du traitement des hémopathies malignes à celui des maladies cardiaques ? BULLETIN DE L'ACADÉMIE NATIONALE DE MÉDECINE 2022. [DOI: 10.1016/j.banm.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
34
|
The role of the dystrophin glycoprotein complex in muscle cell mechanotransduction. Commun Biol 2022; 5:1022. [PMID: 36168044 PMCID: PMC9515174 DOI: 10.1038/s42003-022-03980-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Dystrophin is the central protein of the dystrophin-glycoprotein complex (DGC) in skeletal and heart muscle cells. Dystrophin connects the actin cytoskeleton to the extracellular matrix (ECM). Severing the link between the ECM and the intracellular cytoskeleton has a devastating impact on the homeostasis of skeletal muscle cells, leading to a range of muscular dystrophies. In addition, the loss of a functional DGC leads to progressive dilated cardiomyopathy and premature death. Dystrophin functions as a molecular spring and the DGC plays a critical role in maintaining the integrity of the sarcolemma. Additionally, evidence is accumulating, linking the DGC to mechanosignalling, albeit this role is still less understood. This review article aims at providing an up-to-date perspective on the DGC and its role in mechanotransduction. We first discuss the intricate relationship between muscle cell mechanics and function, before examining the recent research for a role of the dystrophin glycoprotein complex in mechanotransduction and maintaining the biomechanical integrity of muscle cells. Finally, we review the current literature to map out how DGC signalling intersects with mechanical signalling pathways to highlight potential future points of intervention, especially with a focus on cardiomyopathies. A review of the function of the Dystrophic Glycoprotein Complex (DGC) in mechanosignaling provides an overview of the various components of DGC and potential mechanopathogenic mechanisms, particularly as they relate to muscular dystrophy.
Collapse
|
35
|
Hamledari H, Asghari P, Jayousi F, Aguirre A, Maaref Y, Barszczewski T, Ser T, Moore E, Wasserman W, Klein Geltink R, Teves S, Tibbits GF. Using human induced pluripotent stem cell-derived cardiomyocytes to understand the mechanisms driving cardiomyocyte maturation. Front Cardiovasc Med 2022; 9:967659. [PMID: 36061558 PMCID: PMC9429949 DOI: 10.3389/fcvm.2022.967659] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality and reduced quality of life globally. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide a personalized platform to study inherited heart diseases, drug-induced cardiac toxicity, and cardiac regenerative therapy. However, the immaturity of CMs obtained by current strategies is a major hurdle in utilizing hiPSC-CMs at their fullest potential. Here, the major findings and limitations of current maturation methodologies to enhance the utility of hiPSC-CMs in the battle against a major source of morbidity and mortality are reviewed. The most recent knowledge of the potential signaling pathways involved in the transition of fetal to adult CMs are assimilated. In particular, we take a deeper look on role of nutrient sensing signaling pathways and the potential role of cap-independent translation mediated by the modulation of mTOR pathway in the regulation of cardiac gap junctions and other yet to be identified aspects of CM maturation. Moreover, a relatively unexplored perspective on how our knowledge on the effects of preterm birth on cardiovascular development can be actually utilized to enhance the current understanding of CM maturation is examined. Furthermore, the interaction between the evolving neonatal human heart and brown adipose tissue as the major source of neonatal thermogenesis and its endocrine function on CM development is another discussed topic which is worthy of future investigation. Finally, the current knowledge regarding transcriptional mediators of CM maturation is still limited. The recent studies have produced the groundwork to better understand CM maturation in terms of providing some of the key factors involved in maturation and development of metrics for assessment of maturation which proves essential for future studies on in vitro PSC-CMs maturation.
Collapse
Affiliation(s)
- Homa Hamledari
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Parisa Asghari
- Department of Cellular and Physiological Sciences, University of British Colombia, Vancouver, BC, Canada
| | - Farah Jayousi
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Alejandro Aguirre
- Department of Medical Genetics, University of British Colombia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Yasaman Maaref
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Tiffany Barszczewski
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Terri Ser
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Colombia, Vancouver, BC, Canada
| | - Edwin Moore
- Department of Cellular and Physiological Sciences, University of British Colombia, Vancouver, BC, Canada
| | - Wyeth Wasserman
- Department of Medical Genetics, University of British Colombia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Ramon Klein Geltink
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Colombia, Vancouver, BC, Canada
| | - Sheila Teves
- Department of Biochemistry and Molecular Biology, University of British Colombia, Vancouver, BC, Canada
| | - Glen F. Tibbits
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
36
|
Dutta A, Das M, Ghosh A, Rana S. Molecular and cellular pathophysiology of circulating cardiomyocyte-specific cell free DNA (cfDNA): Biomarkers of heart failure and potential therapeutic targets. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
|
37
|
Abstract
Heart disease remains the leading cause of morbidity and mortality worldwide. With the advancement of modern technology, the role(s) of microtubules in the pathogenesis of heart disease has become increasingly apparent, though currently there are limited treatments targeting microtubule-relevant mechanisms. Here, we review the functions of microtubules in the cardiovascular system and their specific adaptive and pathological phenotypes in cardiac disorders. We further explore the use of microtubule-targeting drugs and highlight promising druggable therapeutic targets for the future treatment of heart diseases.
Collapse
Affiliation(s)
- Emily F Warner
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, United Kingdom (E.F.W., X.L.)
| | - Yang Li
- Department of Cardiovascular Surgery, Zhongnan Hospital, Wuhan University School of Medicine, People's Republic of China (Y.L.)
| | - Xuan Li
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, United Kingdom (E.F.W., X.L.)
| |
Collapse
|
38
|
Mehrotra S, de Melo BAG, Miscuglio M, Kiaee K, Shin SR, Mandal BB. Mimicking Native Heart Tissue Physiology and Pathology in Silk Fibroin Constructs through a Perfusion-Based Dynamic Mechanical Stimulation Microdevice. Adv Healthc Mater 2022; 11:e2101678. [PMID: 34971210 PMCID: PMC11041525 DOI: 10.1002/adhm.202101678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 12/23/2021] [Indexed: 11/07/2022]
Abstract
In vitro cardiomyocyte (CM) maturation is an imperative step to replicate native heart tissue-like structures as cardiac tissue grafts or as drug screening platforms. CMs are known to interpret biophysical cues such as stiffness, topography, external mechanical stimulation or dynamic perfusion load through mechanotransduction and change their behavior, organization, and maturation. In this regard, a silk-based cardiac tissue (CT) coupled with a dynamic perfusion-based mechanical stimulation platform (DMM) for achieving maturation and functionality in vitro is tried to be delivered. Silk fibroin (SF) is used to fabricate lamellar scaffolds to provide native tissue-like anisotropic architecture and is found to be nonimmunogenic and biocompatible allowing cardiomyocyte attachment and growth in vitro. Further, the scaffolds display excellent mechanical properties by their ability to undergo cyclic compressions without any deformation when places in the DMM. Gradient compression strains (5% to 20%), mimicking the native physiological and pathological conditions, are applied to the cardiomyocyte culture seeded on lamellar silk scaffolds in the DMM. A strain-dependent difference in cardiomyocyte maturation, gene expression, sarcomere elongation, and extracellular matrix formation is observed. These silk-based CTs matured in the DMM can open up several avenues toward the development of host-specific grafts and in vitro models for drug screening.
Collapse
Affiliation(s)
- Shreya Mehrotra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Bruna Alice Gomes de Melo
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, 04039-002, Brazil
| | - Mario Miscuglio
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
- Department of Electrical and Computer Engineering, George Washington University, Washington, DC, 20052, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, and Brigham and Women's Hospital, Cambridge, MA, 02139, USA
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| |
Collapse
|
39
|
OUP accepted manuscript. Eur Heart J 2022; 43:1531-1532. [DOI: 10.1093/eurheartj/ehac090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
40
|
Vertii A. Stress as a Chromatin Landscape Architect. Front Cell Dev Biol 2021; 9:790138. [PMID: 34970548 PMCID: PMC8712864 DOI: 10.3389/fcell.2021.790138] [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: 10/06/2021] [Accepted: 11/19/2021] [Indexed: 11/16/2022] Open
Abstract
The exponential development of methods investigating different levels of spatial genome organization leads to the appreciation of the chromatin landscape's contribution to gene regulation and cell fate. Multiple levels of 3D chromatin organization include chromatin loops and topologically associated domains, followed by euchromatin and heterochromatin compartments, chromatin domains associated with nuclear bodies, and culminate with the chromosome territories. 3D chromatin architecture is exposed to multiple factors such as cell division and stress, including but not limited to mechanical, inflammatory, and environmental challenges. How exactly the stress exposure shapes the chromatin landscape is a new and intriguing area of research. In this mini-review, the developments that motivate the exploration of this field are discussed.
Collapse
Affiliation(s)
- Anastassiia Vertii
- Department of Molecular, Cellular and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
41
|
Loescher CM, Hobbach AJ, Linke WA. Titin (TTN): from molecule to modifications, mechanics and medical significance. Cardiovasc Res 2021; 118:2903-2918. [PMID: 34662387 PMCID: PMC9648829 DOI: 10.1093/cvr/cvab328] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022] Open
Abstract
The giant sarcomere protein titin is a major determinant of cardiomyocyte stiffness and contributor to cardiac strain sensing. Titin-based forces are highly regulated in health and disease, which aids in the regulation of myocardial function, including cardiac filling and output. Due to the enormous size, complexity, and malleability of the titin molecule, titin properties are also vulnerable to dysregulation, as observed in various cardiac disorders. This review provides an overview of how cardiac titin properties can be changed at a molecular level, including the role isoform diversity and post-translational modifications (acetylation, oxidation, and phosphorylation) play in regulating myocardial stiffness and contractility. We then consider how this regulation becomes unbalanced in heart disease, with an emphasis on changes in titin stiffness and protein quality control. In this context, new insights into the key pathomechanisms of human cardiomyopathy due to a truncation in the titin gene (TTN) are discussed. Along the way, we touch on the potential for titin to be therapeutically targeted to treat acquired or inherited cardiac conditions, such as HFpEF or TTN-truncation cardiomyopathy.
Collapse
Affiliation(s)
- Christine M Loescher
- Institute of Physiology II, University Hospital Münster, Robert-Koch-Str. 27B, Münster, 48149 Germany
| | - Anastasia J Hobbach
- Department of Cardiology I, Coronary, Peripheral Vascular Disease and Heart Failure, University Hospital Münster, Münster, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University Hospital Münster, Robert-Koch-Str. 27B, Münster, 48149 Germany
| |
Collapse
|
42
|
Emig R, Zgierski-Johnston CM, Timmermann V, Taberner AJ, Nash MP, Kohl P, Peyronnet R. Passive myocardial mechanical properties: meaning, measurement, models. Biophys Rev 2021; 13:587-610. [PMID: 34765043 PMCID: PMC8555034 DOI: 10.1007/s12551-021-00838-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Passive mechanical tissue properties are major determinants of myocardial contraction and relaxation and, thus, shape cardiac function. Tightly regulated, dynamically adapting throughout life, and affecting a host of cellular functions, passive tissue mechanics also contribute to cardiac dysfunction. Development of treatments and early identification of diseases requires better spatio-temporal characterisation of tissue mechanical properties and their underlying mechanisms. With this understanding, key regulators may be identified, providing pathways with potential to control and limit pathological development. Methodologies and models used to assess and mimic tissue mechanical properties are diverse, and available data are in part mutually contradictory. In this review, we define important concepts useful for characterising passive mechanical tissue properties, and compare a variety of in vitro and in vivo techniques that allow one to assess tissue mechanics. We give definitions of key terms, and summarise insight into determinants of myocardial stiffness in situ. We then provide an overview of common experimental models utilised to assess the role of environmental stiffness and composition, and its effects on cardiac cell and tissue function. Finally, promising future directions are outlined.
Collapse
Affiliation(s)
- Ramona Emig
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Callum M. Zgierski-Johnston
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Viviane Timmermann
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andrew J. Taberner
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - Martyn P. Nash
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - Peter Kohl
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Faculty of Engineering, University of Freiburg, Freiburg, Germany
| | - Rémi Peyronnet
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg, Bad Krozingen, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
43
|
The role of osteoprotegerin (OPG) in fibrosis: its potential as a biomarker and/or biological target for the treatment of fibrotic diseases. Pharmacol Ther 2021; 228:107941. [PMID: 34171336 DOI: 10.1016/j.pharmthera.2021.107941] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined by excessive formation and accumulation of extracellular matrix proteins, produced by myofibroblasts, that supersedes normal wound healing responses to injury and results in progressive architectural remodelling. Fibrosis is often detected in advanced disease stages when an organ is already severely damaged and can no longer function properly. Therefore, there is an urgent need for reliable and easily detectable markers to identify and monitor fibrosis onset and progression as early as possible; this will greatly facilitate the development of novel therapeutic strategies. Osteoprotegerin (OPG), a well-known regulator of bone extracellular matrix and most studied for its role in regulating bone mass, is expressed in various organs and functions as a decoy for receptor activator of nuclear factor kappa-B ligand (RANKL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Recently, OPG has been linked to fibrosis and fibrogenesis, and has been included in a panel of markers to diagnose liver fibrosis. Multiple studies now suggest that OPG may be a general biomarker suitable for detection of fibrosis and/or monitoring the impact of fibrosis treatment. This review summarizes our current understanding of the role of OPG in fibrosis and will discuss its potential as a biomarker and/or novel therapeutic target for fibrosis.
Collapse
|
44
|
ERK1/2: An Integrator of Signals That Alters Cardiac Homeostasis and Growth. BIOLOGY 2021; 10:biology10040346. [PMID: 33923899 PMCID: PMC8072600 DOI: 10.3390/biology10040346] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 12/24/2022]
Abstract
Integration of cellular responses to extracellular cues is essential for cell survival and adaptation to stress. Extracellular signal-regulated kinase (ERK) 1 and 2 serve an evolutionarily conserved role for intracellular signal transduction that proved critical for cardiomyocyte homeostasis and cardiac stress responses. Considering the importance of ERK1/2 in the heart, understanding how these kinases operate in both normal and disease states is critical. Here, we review the complexity of upstream and downstream signals that govern ERK1/2-dependent regulation of cardiac structure and function. Particular emphasis is given to cardiomyocyte hypertrophy as an outcome of ERK1/2 activation regulation in the heart.
Collapse
|