1
|
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
|
2
|
Zhang B, Powers JD, McCulloch AD, Chi NC. Nuclear mechanosignaling in striated muscle diseases. Front Physiol 2023; 14:1126111. [PMID: 36960155 PMCID: PMC10027932 DOI: 10.3389/fphys.2023.1126111] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/22/2023] [Indexed: 03/09/2023] Open
Abstract
Mechanosignaling describes processes by which biomechanical stimuli are transduced into cellular responses. External biophysical forces can be transmitted via structural protein networks that span from the cellular membrane to the cytoskeleton and the nucleus, where they can regulate gene expression through a series of biomechanical and/or biochemical mechanosensitive mechanisms, including chromatin remodeling, translocation of transcriptional regulators, and epigenetic factors. Striated muscle cells, including cardiac and skeletal muscle myocytes, utilize these nuclear mechanosignaling mechanisms to respond to changes in their intracellular and extracellular mechanical environment and mediate gene expression and cell remodeling. In this brief review, we highlight and discuss recent experimental work focused on the pathway of biomechanical stimulus propagation at the nucleus-cytoskeleton interface of striated muscles, and the mechanisms by which these pathways regulate gene regulation, muscle structure, and function. Furthermore, we discuss nuclear protein mutations that affect mechanosignaling function in human and animal models of cardiomyopathy. Furthermore, current open questions and future challenges in investigating striated muscle nuclear mechanosignaling are further discussed.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Joseph D. Powers
- Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
| | - Andrew D. McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
- Institute for Engineering in Medicine, University of California San Diego, La Jolla, CA, United States
| | - Neil C. Chi
- Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
- Institute for Engineering in Medicine, University of California San Diego, La Jolla, CA, United States
- Department of Medicine, Division of Cardiovascular Medicine, University of California San Diego, La Jolla, CA, United States
- Institute of Genomic Medicine, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
3
|
Navaee F, Renaud P, Piacentini N, Durand M, Bayat DZ, Ledroit D, Heub S, Boder-Pasche S, Kleger A, Braschler T, Weder G. Toward a Physiologically Relevant 3D Helicoidal-Oriented Cardiac Model: Simultaneous Application of Mechanical Stimulation and Surface Topography. Bioengineering (Basel) 2023; 10:bioengineering10020266. [PMID: 36829760 PMCID: PMC9952807 DOI: 10.3390/bioengineering10020266] [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: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Myocardium consists of cardiac cells that interact with their environment through physical, biochemical, and electrical stimulations. The physiology, function, and metabolism of cardiac tissue are affected by this dynamic structure. Within the myocardium, cardiomyocytes' orientations are parallel, creating a dominant orientation. Additionally, local alignments of fibers, along with a helical organization, become evident at the macroscopic level. For the successful development of a reliable in vitro cardiac model, evaluation of cardiac cells' behavior in a dynamic microenvironment, as well as their spatial architecture, is mandatory. In this study, we hypothesize that complex interactions between long-term contraction boundary conditions and cyclic mechanical stimulation may provide a physiological mechanism to generate off-axis alignments in the preferred mechanical stretch direction. This off-axis alignment can be engineered in vitro and, most importantly, mirrors the helical arrangements observed in vivo. For this purpose, uniaxial mechanical stretching of dECM-fibrin hydrogels was performed on pre-aligned 3D cultures of cardiac cells. In view of the potential development of helical structures similar to those in native hearts, the possibility of generating oblique alignments ranging between 0° and 90° was explored. Indeed, our investigations of cell alignment in 3D, employing both mechanical stimulation and groove constraint, provide a reliable mechanism for the generation of helicoidal structures in the myocardium. By combining cyclic stretch and geometric alignment in grooves, an intermediate angle toward favored direction can be achieved experimentally: while cyclic stretch produces a perpendicular orientation, geometric alignment is associated with a parallel one. In our 2D and 3D culture conditions, nonlinear cellular addition of the strains and strain avoidance concept reliably predicted the preferred cellular alignment. The 3D dECM-fibrin model system in this study shows that cyclical stretching supports cell survival and development. Using mechanical stimulation of pre-aligned heart cells, maturation markers are augmented in neonatal cardiomyocytes, while the beating culture period is prolonged, indicating an improved model function. We propose a simplified theoretical model based on numerical simulation and nonlinear strain avoidance by cells to explain oblique alignment angles. Thus, this work lays a possible rational basis for understanding and engineering oblique cellular alignments, such as the helicoidal layout of the heart, using approaches that simultaneously enhance maturation and function.
Collapse
Affiliation(s)
- Fatemeh Navaee
- Microsystems Laboratory-LMIS4, EPFL, 1015 Lausanne, Switzerland
- Department of Pathology and Immunology, Faculty of Medicine, CMU, 1206 Geneva, Switzerland
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, 89081 Ulm, Germany
| | - Philippe Renaud
- Microsystems Laboratory-LMIS4, EPFL, 1015 Lausanne, Switzerland
| | | | - Mathilde Durand
- Swiss Center for Electronics and Microtechnology (CSEM), 2002 Neuchatel, Switzerland
| | - Dara Zaman Bayat
- Swiss Center for Electronics and Microtechnology (CSEM), 2002 Neuchatel, Switzerland
| | - Diane Ledroit
- Swiss Center for Electronics and Microtechnology (CSEM), 2002 Neuchatel, Switzerland
| | - Sarah Heub
- Swiss Center for Electronics and Microtechnology (CSEM), 2002 Neuchatel, Switzerland
| | | | - Alexander Kleger
- Institute of Molecular Oncology and Stem Cell Biology, Ulm University Hospital, 89081 Ulm, Germany
- Interdisciplinary Pancreatology, Department of Internal Medicine 1, Ulm University Hospital, 89081 Ulm, Germany
- Organoid Core Facility, Medical Faculty, Ulm University Hospital, 89081 Ulm, Germany
| | - Thomas Braschler
- Department of Pathology and Immunology, Faculty of Medicine, CMU, 1206 Geneva, Switzerland
- Correspondence:
| | - Gilles Weder
- Swiss Center for Electronics and Microtechnology (CSEM), 2002 Neuchatel, Switzerland
| |
Collapse
|
4
|
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
|
5
|
Urdeitx P, Doweidar MH. Enhanced Piezoelectric Fibered Extracellular Matrix to Promote Cardiomyocyte Maturation and Tissue Formation: A 3D Computational Model. BIOLOGY 2021; 10:biology10020135. [PMID: 33572184 PMCID: PMC7914718 DOI: 10.3390/biology10020135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/26/2022]
Abstract
Mechanical and electrical stimuli play a key role in tissue formation, guiding cell processes such as cell migration, differentiation, maturation, and apoptosis. Monitoring and controlling these stimuli on in vitro experiments is not straightforward due to the coupling of these different stimuli. In addition, active and reciprocal cell-cell and cell-extracellular matrix interactions are essential to be considered during formation of complex tissue such as myocardial tissue. In this sense, computational models can offer new perspectives and key information on the cell microenvironment. Thus, we present a new computational 3D model, based on the Finite Element Method, where a complex extracellular matrix with piezoelectric properties interacts with cardiac muscle cells during the first steps of tissue formation. This model includes collective behavior and cell processes such as cell migration, maturation, differentiation, proliferation, and apoptosis. The model has employed to study the initial stages of in vitro cardiac aggregate formation, considering cell-cell junctions, under different extracellular matrix configurations. Three different cases have been purposed to evaluate cell behavior in fibered, mechanically stimulated fibered, and mechanically stimulated piezoelectric fibered extra-cellular matrix. In this last case, the cells are guided by the coupling of mechanical and electrical stimuli. Accordingly, the obtained results show the formation of more elongated groups and enhancement in cell proliferation.
Collapse
Affiliation(s)
- Pau Urdeitx
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, 50018 Zaragoza, Spain;
- Aragon Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 50018 Zaragoza, Spain
| | - Mohamed H. Doweidar
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, 50018 Zaragoza, Spain;
- Aragon Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 50018 Zaragoza, Spain
- Correspondence:
| |
Collapse
|
6
|
Liao H, Qi Y, Ye Y, Yue P, Zhang D, Li Y. Mechanotranduction Pathways in the Regulation of Mitochondrial Homeostasis in Cardiomyocytes. Front Cell Dev Biol 2021; 8:625089. [PMID: 33553165 PMCID: PMC7858659 DOI: 10.3389/fcell.2020.625089] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are one of the most important organelles in cardiomyocytes. Mitochondrial homeostasis is necessary for the maintenance of normal heart function. Mitochondria perform four major biological processes in cardiomyocytes: mitochondrial dynamics, metabolic regulation, Ca2+ handling, and redox generation. Additionally, the cardiovascular system is quite sensitive in responding to changes in mechanical stress from internal and external environments. Several mechanotransduction pathways are involved in regulating the physiological and pathophysiological status of cardiomyocytes. Typically, the extracellular matrix generates a stress-loading gradient, which can be sensed by sensors located in cellular membranes, including biophysical and biochemical sensors. In subsequent stages, stress stimulation would regulate the transcription of mitochondrial related genes through intracellular transduction pathways. Emerging evidence reveals that mechanotransduction pathways have greatly impacted the regulation of mitochondrial homeostasis. Excessive mechanical stress loading contributes to impairing mitochondrial function, leading to cardiac disorder. Therefore, the concept of restoring mitochondrial function by shutting down the excessive mechanotransduction pathways is a promising therapeutic strategy for cardiovascular diseases. Recently, viral and non-viral protocols have shown potentials in application of gene therapy. This review examines the biological process of mechanotransduction pathways in regulating mitochondrial function in response to mechanical stress during the development of cardiomyopathy and heart failure. We also summarize gene therapy delivery protocols to explore treatments based on mechanical stress-induced mitochondrial dysfunction, to provide new integrative insights into cardiovascular diseases.
Collapse
Affiliation(s)
- Hongyu Liao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yan Qi
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yida Ye
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Peng Yue
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, Wuhan, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Wei A, Wang Z, Rancu AL, Yang Z, Tan S, Borg TK, Gao BZ. In Vivo-Like Morphology of Intercalated Discs Achieved in a Neonatal Cardiomyocyte Culture Model. Tissue Eng Part A 2020; 26:1209-1221. [PMID: 32515285 PMCID: PMC7699015 DOI: 10.1089/ten.tea.2020.0068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
In vitro cultures to be used in various analytical investigations of cardiomyocyte (CM) growth and function for enhancing insight into physiological and pathological mechanisms should closely express in vivo morphology. The aim of the studies is to explore how to use microfabrication and physical-cue-addition techniques to establish a neonatal rat CM culture model that expresses an end-to-end connected rod shape with in vivo-like intercalated discs (ICDs). Freshly isolated neonatal rat CMs were cultured on microgrooved polydimethylsiloxane substrate. Cell alignment and ICD orientation were evaluated using confocal fluorescence and transmission electron microscopy under various combinations of different culture conditions. Cyclic stretch and blebbistatin tests were conducted to explore mechanical and electrical effects. Laboratory-made MATLAB software was developed to quantify cell alignment and ICD orientation. Our results demonstrate that the mechanical effect associated with the electrical stimulation may contribute to step-like ICD formation viewed from the top. In addition, our study reveals that a suspended elastic substrate that was slack with scattered folds, not taut, enabled CM contraction of equal strength on both apical and basal cell surfaces, allowing the cultured CMs to express a three-dimensional rod shape with disc-like ICDs viewed cross-sectionally. Impact statement In this article, we describe how the tugging forces generated by cardiomyocytes (CMs) facilitate the formation of the morphology of the intercalated discs (ICDs) to achieve mechanoelectrical coupling between CMs. Correspondingly, we report experimental techniques we developed to enable the in vivo-like behavior of the tugging forces to support the development of in vivo-like morphology in ICDs. These techniques will enhance insight into physiological and pathological mechanisms related to the development of tissue-engineered cardiac constructs in various analytical investigations of CM growth and function.
Collapse
Affiliation(s)
- Ailin Wei
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Zhonghai Wang
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | | | - Zongming Yang
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Shenghao Tan
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| | - Thomas Keith Borg
- Department of Regenerative Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Bruce Zhi Gao
- Department of Bioengineering, Clemson University, Clemson, South Carolina, USA
| |
Collapse
|
8
|
A Computational Model for Cardiomyocytes Mechano-Electric Stimulation to Enhance Cardiac Tissue Regeneration. MATHEMATICS 2020. [DOI: 10.3390/math8111875] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Electrical and mechanical stimulations play a key role in cell biological processes, being essential in processes such as cardiac cell maturation, proliferation, migration, alignment, attachment, and organization of the contractile machinery. However, the mechanisms that trigger these processes are still elusive. The coupling of mechanical and electrical stimuli makes it difficult to abstract conclusions. In this sense, computational models can establish parametric assays with a low economic and time cost to determine the optimal conditions of in-vitro experiments. Here, a computational model has been developed, using the finite element method, to study cardiac cell maturation, proliferation, migration, alignment, and organization in 3D matrices, under mechano-electric stimulation. Different types of electric fields (continuous, pulsating, and alternating) in an intensity range of 50–350 Vm−1, and extracellular matrix with stiffnesses in the range of 10–40 kPa, are studied. In these experiments, the group’s morphology and cell orientation are compared to define the best conditions for cell culture. The obtained results are qualitatively consistent with the bibliography. The electric field orientates the cells and stimulates the formation of elongated groups. Group lengthening is observed when applying higher electric fields in lower stiffness extracellular matrix. Groups with higher aspect ratios can be obtained by electrical stimulation, with better results for alternating electric fields.
Collapse
|
9
|
Nielsen NR, Rangarajan KV, Mao L, Rockman HA, Caron KM. A murine model of increased coronary sinus pressure induces myocardial edema with cardiac lymphatic dilation and fibrosis. Am J Physiol Heart Circ Physiol 2020; 318:H895-H907. [PMID: 32142379 DOI: 10.1152/ajpheart.00436.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Myocardial edema is a consequence of many cardiovascular stressors, including myocardial infarction, cardiac bypass surgery, and hypertension. The aim of this study was to establish a murine model of myocardial edema and elucidate the response of cardiac lymphatics and the myocardium. Myocardial edema without infarction was induced in mice by cauterizing the coronary sinus, increasing pressure in the coronary venous system, and inducing myocardial edema. In male mice, there was rapid development of edema 3 h following coronary sinus cauterization (CSC), with associated dilation of cardiac lymphatics. By 24 h, males displayed significant cardiovascular contractile dysfunction. In contrast, female mice exhibited a temporal delay in the formation of myocardial edema, with onset of cardiovascular dysfunction by 24 h. Furthermore, myocardial edema induced a ring of fibrosis around the epicardial surface of the left ventricle in both sexes that included fibroblasts, immune cells, and increased lymphatics. Interestingly, the pattern of fibrosis and the cells that make up the fibrotic epicardial ring differ between sexes. We conclude that a novel surgical model of myocardial edema without infarct was established in mice. Cardiac lymphatics compensated by exhibiting both an acute dilatory and chronic growth response. Transient myocardial edema was sufficient to induce a robust epicardial fibrotic and inflammatory response, with distinct sex differences, which underscores the sex-dependent differences that exist in cardiac vascular physiology.NEW & NOTEWORTHY Myocardial edema is a consequence of many cardiovascular stressors, including myocardial infarction, cardiac bypass surgery, and high blood pressure. Cardiac lymphatics regulate interstitial fluid balance and, in a myocardial infarction model, have been shown to be therapeutically targetable by increasing heart function. Cardiac lymphatics have only rarely been studied in a noninfarct setting in the heart, and so we characterized the first murine model of increased coronary sinus pressure to induce myocardial edema, demonstrating distinct sex differences in the response to myocardial edema. The temporal pattern of myocardial edema induction and resolution is different between males and females, underscoring sex-dependent differences in the response to myocardial edema. This model provides an important platform for future research in cardiovascular and lymphatic fields with the potential to develop therapeutic interventions for many common cardiovascular diseases.
Collapse
Affiliation(s)
- Natalie R Nielsen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill Chapel Hill, North Carolina
| | - Krsna V Rangarajan
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill Chapel Hill, North Carolina
| | - Lan Mao
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Howard A Rockman
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Kathleen M Caron
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill Chapel Hill, North Carolina
| |
Collapse
|
10
|
Najafian B, Tøndel C, Svarstad E, Gubler MC, Oliveira JP, Mauer M. Accumulation of Globotriaosylceramide in Podocytes in Fabry Nephropathy Is Associated with Progressive Podocyte Loss. J Am Soc Nephrol 2020; 31:865-875. [PMID: 32127409 DOI: 10.1681/asn.2019050497] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 01/06/2020] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND In males with classic Fabry disease, the processes leading to the frequent outcome of ESKD are poorly understood. Defects in the gene encoding α-galactosidase A lead to accumulation of globotriaosylceramide (GL3) in various cell types. In the glomerular podocytes, accumulation of GL3 progresses with age. Of concern, podocytes are relatively resistant to enzyme replacement therapy and are poorly replicating, with little ability to compensate for cell loss. METHODS In this study of 55 males (mean age 27 years) with classic Fabry disease genotype and/or phenotype, we performed unbiased quantitative morphometric electron microscopic studies of biopsied kidney samples from patients and seven living transplant donors (to serve as controls). We extracted clinical information from medical records and clinical trial databases. RESULTS Podocyte GL3 volume fraction (proportion of podocyte cytoplasm occupied by GL3) increased with age up to about age 27, suggesting that increasing podocyte GL3 volume fraction beyond a threshold may compromise survival of these cells. GL3 accumulation was associated with podocyte injury and loss, as evidenced by increased foot process width (a generally accepted structural marker of podocyte stress and injury) and with decreased podocyte number density per glomerular volume. Worsening podocyte structural parameters (increasing podocyte GL3 volume fraction and foot process width) was also associated with increasing urinary protein excretion-a strong prognosticator of adverse renal outcomes in Fabry disease-as well as with decreasing GFR. CONCLUSIONS Given the known association between podocyte loss and irreversible FSGS and global glomerulosclerosis, this study points to an important role for podocyte injury and loss in the progression of Fabry nephropathy and indicates a need for therapeutic intervention before critical podocyte loss occurs.
Collapse
Affiliation(s)
- Behzad Najafian
- Department of Pathology, University of Washington, Seattle, Washington;
| | - Camilla Tøndel
- Department of Pediatrics, Haukland University Hospital, Bergen, Norway.,Institute of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Einar Svarstad
- Institute of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Marie-Claire Gubler
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Imagine Institute, Necker-Enfants Malades Hospital, Paris, France
| | - João-Paulo Oliveira
- Service of Medical Genetics, São João University Hospital, Porto, Portugal.,Department of Medical Genetics, Faculty of Medicine, i3S (Institute for Research and Innovation in Health), University of Porto, Porto, Portugal
| | - Michael Mauer
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota; and.,Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
11
|
Li Z, Liu T, Yang J, Lin J, Xin SX. Characterization of adhesion properties of the cardiomyocyte integrins and extracellular matrix proteins using atomic force microscopy. J Mol Recognit 2019; 33:e2823. [PMID: 31709699 DOI: 10.1002/jmr.2823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 10/08/2019] [Accepted: 10/19/2019] [Indexed: 11/10/2022]
Abstract
Integrins are transmembrane adhesion receptors that play important roles in the cardiovascular system by interacting with the extracellular matrix (ECM). However, direct quantitative measurements of the adhesion properties of the integrins on cardiomyocyte (CM) and their ECM ligands are lacking. In this study, we used atomic force microscopy (AFM) to quantify the adhesion force (peak force and mean force) and binding probability between CM integrins and three main heart tissue ECM proteins, ie, collagen (CN), fibronectin (FN), and laminin (LN). Functionalizing the AFM probes with ECM proteins, we found that the peak force (mean force) was 61.69 ± 5.5 pN (76.54 ± 4.0 pN), 39.26 ± 4.4 pN (59.84 ± 3.6 pN), and 108.31 ± 4.2 pN (129.63 ± 6.0 pN), respectively, for the bond of CN-integrin, FN-integrin, and LN-integrin. The binding specificity between CM integrins and ECM proteins was verified by using monoclonal antibodies, where α10 - and α11 -integrin bind to CN, α3 - and α5 -integrin bind to FN, and α3 - and α7 -integrin bind to LN. Furthermore, adhesion properties of CM integrins under physiologically high concentrations of extracellular Ca2+ and Mg2+ were tested. Additional Ca2+ reduced the adhesion mean force to 68.81 ± 4.0 pN, 49.84 ± 3.3 pN, and 119.21 ± 5.8 pN and binding probability to 0.31, 0.34, 0.40 for CN, FN, and LN, respectively, whereas Mg2+ caused very minor changes to adhesion properties of CM integrins. Thus, adhesion properties between adult murine CM integrins and its main ECM proteins were characterized, paving the way for an improved understanding of CM mechanobiology.
Collapse
Affiliation(s)
- Zecheng Li
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Tianqi Liu
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Junxian Yang
- Institute of Biomechanics, School of Biosciences and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jiangguo Lin
- Institute of Biomechanics, School of Biosciences and Bioengineering, South China University of Technology, Guangzhou, China
| | - Sherman Xuegang Xin
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
12
|
X Cai L, Tanada Y, D Bello G, C Fleming J, F Alkassis F, Ladd T, Golde T, Koh J, Chen S, Kasahara H. Cardiac MLC2 kinase is localized to the Z-disc and interacts with α-actinin2. Sci Rep 2019; 9:12580. [PMID: 31467300 PMCID: PMC6715661 DOI: 10.1038/s41598-019-48884-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac contractility is enhanced by phosphorylation of myosin light chain 2 (MLC2) by cardiac-specific MLC kinase (cMLCK), located at the neck region of myosin heavy chain. In normal mouse and human hearts, the level of phosphorylation is maintained relatively constant, at around 30-40% of total MLC2, likely by well-balanced phosphorylation and phosphatase-dependent dephosphorylation. Overexpression of cMLCK promotes sarcomere organization, while the loss of cMLCK leads to cardiac atrophy in vitro and in vivo. In this study, we showed that cMLCK is predominantly expressed at the Z-disc with additional diffuse cytosolic expression in normal adult mouse and human hearts. cMLCK interacts with the Z-disc protein, α-actinin2, with a high-affinity kinetic value of 13.4 ± 0.1 nM through the N-terminus region of cMLCK unique to cardiac-isoform. cMLCK mutant deficient for interacting with α-actinin2 did not promote sarcomeric organization and reduced cardiomyocyte cell size. In contrast, a cMLCK kinase-deficient mutant showed effects similar to wild-type cMLCK on sarcomeric organization and cardiomyocyte cell size. Our results suggest that cMLCK plays a role in sarcomere organization, likely distinct from its role in phosphorylating MLC2, both of which will contribute to the enhancement of cardiac contractility.
Collapse
Affiliation(s)
- Lawrence X Cai
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Yohei Tanada
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Gregory D Bello
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - James C Fleming
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Fariz F Alkassis
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Thomas Ladd
- Department of Neuroscience, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Todd Golde
- Department of Neuroscience, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Jin Koh
- Proteomics and Mass Spectrometry, Interdisciplinary Center for Biotechnology Research (ICBR), University of Florida, Gainesville, FL, 32610, USA
| | - Sixue Chen
- Proteomics and Mass Spectrometry, Interdisciplinary Center for Biotechnology Research (ICBR), University of Florida, Gainesville, FL, 32610, USA.,Department of Biology, Genetics Institute, Plant Molecular and Cellular Biology Program, University of Florida, Gainesville, FL, 32610, USA
| | - Hideko Kasahara
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA.
| |
Collapse
|
13
|
González A, López B, Ravassa S, San José G, Díez J. Reprint of "The complex dynamics of myocardial interstitial fibrosis in heart failure. Focus on collagen cross-linking". BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118521. [PMID: 31394074 DOI: 10.1016/j.bbamcr.2019.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 12/12/2022]
Abstract
Myocardial interstitial fibrosis (MIF) is a common finding in heart failure (HF) patients, both with preserved and reduced ejection fraction, as well as in HF animal models. MIF is associated with impaired cardiac function and worse clinical outcome. The impact of MIF is influenced not only by the quantity but also by changes in the quality of collagen fibers and in the extracellular matrix components, such as a shift in collagen types proportion, increased fibronectin polymerization and increased degree of collagen cross-linking (CCL). In particular, CCL, a process that renders collagen fibers stiffer and more resistant to degradation, is increased both in patients and animal models of HF. Importantly, in HF patients increased cardiac CCL is directly associated with increased left ventricular stiffness and a higher risk of hospitalization for HF. The aim of this review is to address the complexity of MIF in HF, focusing on CCL.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| | - Begoña López
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Susana Ravassa
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Gorka San José
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain; Departments of Cardiology and Cardiac Surgery and of Nephrology, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
14
|
González A, López B, Ravassa S, San José G, Díez J. The complex dynamics of myocardial interstitial fibrosis in heart failure. Focus on collagen cross-linking. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1421-1432. [PMID: 31181222 DOI: 10.1016/j.bbamcr.2019.06.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 05/21/2019] [Accepted: 06/03/2019] [Indexed: 12/11/2022]
Abstract
Myocardial interstitial fibrosis (MIF) is a common finding in heart failure (HF) patients, both with preserved and reduced ejection fraction, as well as in HF animal models. MIF is associated with impaired cardiac function and worse clinical outcome. The impact of MIF is influenced not only by the quantity but also by changes in the quality of collagen fibers and in the extracellular matrix components, such as a shift in collagen types proportion, increased fibronectin polymerization and increased degree of collagen cross-linking (CCL). In particular, CCL, a process that renders collagen fibers stiffer and more resistant to degradation, is increased both in patients and animal models of HF. Importantly, in HF patients increased cardiac CCL is directly associated with increased left ventricular stiffness and a higher risk of hospitalization for HF. The aim of this review is to address the complexity of MIF in HF, focusing on CCL.
Collapse
Affiliation(s)
- Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain.
| | - Begoña López
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Susana Ravassa
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Gorka San José
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Javier Díez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra and IdiSNA, Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain; Departments of Cardiology and Cardiac Surgery and of Nephrology, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
15
|
Patel VB, Zhabyeyev P, Chen X, Wang F, Paul M, Fan D, McLean BA, Basu R, Zhang P, Shah S, Dawson JF, Pyle WG, Hazra M, Kassiri Z, Hazra S, Vanhaesebroeck B, McCulloch CA, Oudit GY. PI3Kα-regulated gelsolin activity is a critical determinant of cardiac cytoskeletal remodeling and heart disease. Nat Commun 2018; 9:5390. [PMID: 30568254 PMCID: PMC6300608 DOI: 10.1038/s41467-018-07812-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 11/28/2018] [Indexed: 12/21/2022] Open
Abstract
Biomechanical stress and cytoskeletal remodeling are key determinants of cellular homeostasis and tissue responses to mechanical stimuli and injury. Here we document the increased activity of gelsolin, an actin filament severing and capping protein, in failing human hearts. Deletion of gelsolin prevents biomechanical stress-induced adverse cytoskeletal remodeling and heart failure in mice. We show that phosphatidylinositol (3,4,5)-triphosphate (PIP3) lipid suppresses gelsolin actin-severing and capping activities. Accordingly, loss of PI3Kα, the key PIP3-producing enzyme in the heart, increases gelsolin-mediated actin-severing activities in the myocardium in vivo, resulting in dilated cardiomyopathy in response to pressure-overload. Mechanical stretching of adult PI3Kα-deficient cardiomyocytes disrupts the actin cytoskeleton, which is prevented by reconstituting cells with PIP3. The actin severing and capping activities of recombinant gelsolin are effectively suppressed by PIP3. Our data identify the role of gelsolin-driven cytoskeletal remodeling in heart failure in which PI3Kα/PIP3 act as negative regulators of gelsolin activity.
Collapse
Affiliation(s)
- Vaibhav B Patel
- Division of Cardiology, Department of Medicine, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Department of Physiology and Pharmacology and Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, HMRB-71, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| | - Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
| | - Xueyi Chen
- Division of Cardiology, Department of Medicine, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
| | - Faqi Wang
- Division of Cardiology, Department of Medicine, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
| | - Manish Paul
- Department of Biotechnology, North Orissa University, Baripada, 757003, Odisha, India
| | - Dong Fan
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Department of Physiology, University of Alberta, HMRC-407, 116 St 85 Ave, Edmonton, AB T6G 2S2, Canada
| | - Brent A McLean
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Department of Physiology, University of Alberta, HMRC-407, 116 St 85 Ave, Edmonton, AB T6G 2S2, Canada
| | - Ratnadeep Basu
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Department of Physiology, University of Alberta, HMRC-407, 116 St 85 Ave, Edmonton, AB T6G 2S2, Canada
| | - Pu Zhang
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Department of Physiology, University of Alberta, HMRC-407, 116 St 85 Ave, Edmonton, AB T6G 2S2, Canada
| | - Saumya Shah
- Division of Cardiology, Department of Medicine, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
| | - John F Dawson
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, ON, N1G 2W1, Canada
- Centre of Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - W Glen Pyle
- Centre of Cardiovascular Investigations, University of Guelph, Guelph, ON, N1G 2W1, Canada
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Mousumi Hazra
- Department of Botany and Microbiology, Gurukula Kangri University, Haridwar, 249404, Uttarakhand, India
| | - Zamaneh Kassiri
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada
- Department of Physiology, University of Alberta, HMRC-407, 116 St 85 Ave, Edmonton, AB T6G 2S2, Canada
| | - Saugata Hazra
- Department of Biotechnology, Indian Institute of Technology, Roorkee, 247667, Uttarakhand, India
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, Uttarakhand, India
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, University College London, London, WC1E 6BT, England, UK
| | - Christopher A McCulloch
- Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Toronto, ON, M5S 3E2, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada.
- Mazankowski Alberta Heart Institute, University of Alberta, 2C2, 8440-112 St, Edmonton, AB T6G 2B7, Canada.
- Department of Physiology, University of Alberta, HMRC-407, 116 St 85 Ave, Edmonton, AB T6G 2S2, Canada.
| |
Collapse
|
16
|
Abstract
In the heart, cardiac muscle fibers curve creating zones of membrane forces resulting in regions of mechanotransduction. This study uses the finite difference method to solve the mechanical bidomain equations numerically for a complex fiber geometry. The magnitude of the active tension T is constant but its direction makes an angle with the x-axis that varies with position. Differences between the intracellular and extracellular displacements result from the bidomain behavior of the tissue that gives rise to forces on the integrin proteins in the membrane. The long-term goal is to use the mechanical bidomain model to suggest experiments and make predictions about growth and remodeling in the heart.
Collapse
|
17
|
Yuan H, Marzban B, Kit Parker K. Myofibrils in Cardiomyocytes Tend to Assemble Along the Maximal Principle Stress Directions. J Biomech Eng 2018; 139:2653368. [PMID: 28857113 DOI: 10.1115/1.4037795] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Indexed: 11/08/2022]
Abstract
The mechanisms underlying the spatial organization of self-assembled myofibrils in cardiac tissues remain incompletely understood. By modeling cells as elastic solids under active cytoskeletal contraction, we found a good correlation between the predicted maximal principal stress directions and the in vitro myofibril orientations in individual cardiomyocytes. This implies that actomyosin fibers tend to assemble along the maximal tensile stress (MTS) directions. By considering the dynamics of focal adhesion and myofibril formation in the model, we showed that different patterns of myofibril organizations in mature versus immature cardiomyocytes can be explained as the consequence of the different levels of force-dependent remodeling of focal adhesions. Further, we applied the mechanics model to cell pairs and showed that the myofibril organizations can be regulated by a combination of multiple factors including cell shape, cell-substrate adhesions, and cell-cell adhesions. This mechanics model can guide the rational design in cardiac tissue engineering where recapitulating in vivo myofibril organizations is crucial to the contractile function of the heart.
Collapse
Affiliation(s)
- Hongyan Yuan
- Department of Mechanical, Industrial and Systems Engineering, University of Rhode Island, Kingston, RI 02881 e-mail:
| | - Bahador Marzban
- Department of Mechanical, Industrial and Systems Engineering, University of Rhode Island, Kingston, RI 02881
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138 e-mail:
| |
Collapse
|
18
|
Urmaliya V, Franchelli G. A multidimensional sight on cardiac failure: uncovered from structural to molecular level. Heart Fail Rev 2018; 22:357-370. [PMID: 28474325 DOI: 10.1007/s10741-017-9610-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Heart failure is one of the leading causes of death, with high mortality rate within 5 years after diagnosis. Treatment and prognosis options for heart failure primarily targeted on hemodynamic and neurohumoral components that drive progressive deterioration of the heart. However, given the multifactorial background that eventually leads to the "phenotype" named heart failure, better insight into the various components may lead to personalized treatment opportunities. Indeed, currently used criteria to diagnose and/or classify heart failure are possibly too focused on phenotypic improvement rather than the molecular driver of the disease and could therefore be further refined by integrating the leap of molecular and cellular knowledge. The ambiguity of the ejection fraction-based classification criteria became evident with development of advanced molecular techniques and the dawn of omics disciplines which introduced the idea that disease is caused by a myriad of cellular and molecular processes rather than a single event or pathway. The fact that different signaling pathways may underlie similar clinical manifestations calls for a more holistic study of heart failure. In this context, the systems biology approach can offer a better understanding of how different components of a system are altered during disease and how they interact with each other, potentially leading to improved diagnosis and classification of this condition. This review is aimed at addressing heart failure through a multilayer approach that covers individually some of the anatomical, morphological, functional, and tissue aspects, with focus on cellular and subcellular features as an alternative insight into new therapeutic opportunities.
Collapse
Affiliation(s)
- Vijay Urmaliya
- Discovery Sciences, Janssen Research & Development, Beerse, Belgium.
| | | |
Collapse
|
19
|
Chopra A, Kutys ML, Zhang K, Polacheck WJ, Sheng CC, Luu RJ, Eyckmans J, Hinson JT, Seidman JG, Seidman CE, Chen CS. Force Generation via β-Cardiac Myosin, Titin, and α-Actinin Drives Cardiac Sarcomere Assembly from Cell-Matrix Adhesions. Dev Cell 2018; 44:87-96.e5. [PMID: 29316444 DOI: 10.1016/j.devcel.2017.12.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/16/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022]
Abstract
Truncating mutations in the sarcomere protein titin cause dilated cardiomyopathy due to sarcomere insufficiency. However, it remains mechanistically unclear how these mutations decrease sarcomere content in cardiomyocytes. Utilizing human induced pluripotent stem cell-derived cardiomyocytes, CRISPR/Cas9, and live microscopy, we characterize the fundamental mechanisms of human cardiac sarcomere formation. We observe that sarcomerogenesis initiates at protocostameres, sites of cell-extracellular matrix adhesion, where nucleation and centripetal assembly of α-actinin-2-containing fibers provide a template for the fusion of Z-disk precursors, Z bodies, and subsequent striation. We identify that β-cardiac myosin-titin-protocostamere form an essential mechanical connection that transmits forces required to direct α-actinin-2 centripetal fiber assembly and sarcomere formation. Titin propagates diastolic traction stresses from β-cardiac myosin, but not α-cardiac myosin or non-muscle myosin II, to protocostameres during sarcomerogenesis. Ablating protocostameres or decoupling titin from protocostameres abolishes sarcomere assembly. Together these results identify the mechanical and molecular components critical for human cardiac sarcomerogenesis.
Collapse
Affiliation(s)
- Anant Chopra
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Matthew L Kutys
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Kehan Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - William J Polacheck
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - Calvin C Sheng
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Rebeccah J Luu
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Cardiology Center, University of Connecticut Health, Farmington, CT 06030, USA.
| | - Jonathan G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; The Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Aboelkassem Y. Stokes flow patterns induced by a single cardiac cell. Comput Biol Med 2017; 86:65-74. [PMID: 28511120 DOI: 10.1016/j.compbiomed.2017.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/15/2017] [Accepted: 05/01/2017] [Indexed: 11/27/2022]
Abstract
Stokes flow motions induced by a beating single cardiac cell (cardiomyocyte) are obtained numerically using the method of fundamental solutions (MFS). A two-dimensional meshfree-Stokeslets computational framework is used to solve the Stokes governing equations around an isolated cardiomyocyte. An approximate beating kinematical model is derived and used to approximate the cell-length shortening over a complete cardiac cycle. The induced flow patterns have been found to be characterized by the presence of counter-rotating vortices at both cell's edges. These vortical flow structures are clearly shown by rendering the velocity streamlines. The static pressure contours are also calculated at different time snapshots during both contraction and relaxation phases of the beating motion. The pressure signal is calculated at a point in the neighborhood of cell surface to capture the induced normal stress (traction) by the cell morphological motions to the surrounding fluid medium. The presented results have shown that, cells with a slightly different shortening/beating profile can induce different flow field. This implies that, each cell is characterized by a unique flow pattern "signature", which potentially can be correlated to the sub-cellular excitation-contraction processes of cardiac cells.
Collapse
Affiliation(s)
- Yasser Aboelkassem
- Institute for Computational Medicine, Department of Biomedical Engineering Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
21
|
Lister Z, Rayner KJ, Suuronen EJ. How Biomaterials Can Influence Various Cell Types in the Repair and Regeneration of the Heart after Myocardial Infarction. Front Bioeng Biotechnol 2016; 4:62. [PMID: 27486578 PMCID: PMC4948030 DOI: 10.3389/fbioe.2016.00062] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/01/2016] [Indexed: 12/15/2022] Open
Abstract
The healthy heart comprises many different cell types that work together to preserve optimal function. However, in a diseased heart the function of one or more cell types is compromised which can lead to many adverse events, one of which is myocardial infarction (MI). Immediately after MI, the cardiac environment is characterized by excessive cardiomyocyte death and inflammatory signals leading to the recruitment of macrophages to clear the debris. Proliferating fibroblasts then invade, and a collagenous scar is formed to prevent rupture. Better functional restoration of the heart is not achieved due to the limited regenerative capacity of cardiac tissue. To address this, biomaterial therapy is being investigated as an approach to improve regeneration in the infarcted heart, as they can possess the potential to control cell function in the infarct environment and limit the adverse compensatory changes that occur post-MI. Over the past decade, there has been considerable research into the development of biomaterials for cardiac regeneration post-MI; and various effects have been observed on different cell types depending on the biomaterial that is applied. Biomaterial treatment has been shown to enhance survival, improve function, promote proliferation, and guide the mobilization and recruitment of different cells in the post-MI heart. This review will provide a summary on the biomaterials developed to enhance cardiac regeneration and remodeling post-MI with a focus on how they control macrophages, cardiomyocytes, fibroblasts, and endothelial cells. A better understanding of how a biomaterial interacts with the different cell types in the heart may lead to the development of a more optimized biomaterial therapy for cardiac regeneration.
Collapse
Affiliation(s)
- Zachary Lister
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Katey J Rayner
- Atherosclerosis, Genomics and Cell Biology Group, University of Ottawa Heart Institute , Ottawa, ON , Canada
| | - Erik J Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
22
|
Ennomani H, Letort G, Guérin C, Martiel JL, Cao W, Nédélec F, De La Cruz EM, Théry M, Blanchoin L. Architecture and Connectivity Govern Actin Network Contractility. Curr Biol 2016; 26:616-26. [PMID: 26898468 DOI: 10.1016/j.cub.2015.12.069] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 11/25/2015] [Accepted: 12/24/2015] [Indexed: 01/03/2023]
Abstract
Actomyosin contractility plays a central role in a wide range of cellular processes, including the establishment of cell polarity, cell migration, tissue integrity, and morphogenesis during development. The contractile response is variable and depends on actomyosin network architecture and biochemical composition. To determine how this coupling regulates actomyosin-driven contraction, we used a micropatterning method that enables the spatial control of actin assembly. We generated a variety of actin templates and measured how defined actin structures respond to myosin-induced forces. We found that the same actin filament crosslinkers either enhance or inhibit the contractility of a network, depending on the organization of actin within the network. Numerical simulations unified the roles of actin filament branching and crosslinking during actomyosin contraction. Specifically, we introduce the concept of "network connectivity" and show that the contractions of distinct actin architectures are described by the same master curve when considering their degree of connectivity. This makes it possible to predict the dynamic response of defined actin structures to transient changes in connectivity. We propose that, depending on the connectivity and the architecture, network contraction is dominated by either sarcomeric-like or buckling mechanisms. More generally, this study reveals how actin network contractility depends on its architecture under a defined set of biochemical conditions.
Collapse
Affiliation(s)
- Hajer Ennomani
- Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Vegétale, CNRS/CEA/UGA/INRA, Grenoble 38054, France
| | - Gaëlle Letort
- Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Vegétale, CNRS/CEA/UGA/INRA, Grenoble 38054, France
| | - Christophe Guérin
- Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Vegétale, CNRS/CEA/UGA/INRA, Grenoble 38054, France
| | - Jean-Louis Martiel
- Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Vegétale, CNRS/CEA/UGA/INRA, Grenoble 38054, France
| | - Wenxiang Cao
- Department of Molecular Biophysics and Biochemistry, Yale University, 260 Whitney Avenue, New Haven, CT 06520-8114, USA
| | - François Nédélec
- Cell Biology and Biophysics Unit, EMBL, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Enrique M De La Cruz
- Department of Molecular Biophysics and Biochemistry, Yale University, 260 Whitney Avenue, New Haven, CT 06520-8114, USA
| | - Manuel Théry
- Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Vegétale, CNRS/CEA/UGA/INRA, Grenoble 38054, France; Unité de thérapie Cellulaire, Hopital Saint-Louis, Avenue Claude Vellefaux, Paris 75010, France.
| | - Laurent Blanchoin
- Biosciences & Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Vegétale, CNRS/CEA/UGA/INRA, Grenoble 38054, France.
| |
Collapse
|
23
|
Neves JS, Leite-Moreira AM, Neiva-Sousa M, Almeida-Coelho J, Castro-Ferreira R, Leite-Moreira AF. Acute Myocardial Response to Stretch: What We (don't) Know. Front Physiol 2016; 6:408. [PMID: 26779036 PMCID: PMC4700209 DOI: 10.3389/fphys.2015.00408] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/11/2015] [Indexed: 12/24/2022] Open
Abstract
Myocardial stretch, as result of acute hemodynamic overload, is one of the most frequent challenges to the heart and the ability of the heart to intrinsically adapt to it is essential to prevent circulatory congestion. In this review, we highlight the historical background, the currently known mechanisms, as well as the gaps in the understanding of this physiological response. The systolic adaptation to stretch is well-known for over 100 years, being dependent on an immediate increase in contractility—known as the Frank-Starling mechanism—and a further progressive increase—the slow force response. On the other hand, its diastolic counterpart remains largely unstudied. Mechanosensors are structures capable of perceiving mechanical signals and activating pathways that allow their transduction into biochemical responses. Although the connection between these structures and stretch activated pathways remains elusive, we emphasize those most likely responsible for the initiation of the acute response. Calcium-dependent pathways, including angiotensin- and endothelin-related pathways; and cGMP-dependent pathways, comprising the effects of nitric oxide and cardiac natriuretic hormones, embody downstream signaling. The ischemic setting, a paradigmatic situation of acute hemodynamic overload, is also touched upon. Despite the relevant knowledge accumulated, there is much that we still do not know. The quest for further understanding the myocardial response to acute stretch may provide new insights, not only in its physiological importance, but also in the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- João S Neves
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - André M Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - Manuel Neiva-Sousa
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - João Almeida-Coelho
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - Ricardo Castro-Ferreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| | - Adelino F Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine of the University of Porto Porto, Portugal
| |
Collapse
|
24
|
Denning C, Borgdorff V, Crutchley J, Firth KSA, George V, Kalra S, Kondrashov A, Hoang MD, Mosqueira D, Patel A, Prodanov L, Rajamohan D, Skarnes WC, Smith JGW, Young LE. Cardiomyocytes from human pluripotent stem cells: From laboratory curiosity to industrial biomedical platform. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1728-48. [PMID: 26524115 PMCID: PMC5221745 DOI: 10.1016/j.bbamcr.2015.10.014] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 10/12/2015] [Accepted: 10/20/2015] [Indexed: 12/14/2022]
Abstract
Cardiomyocytes from human pluripotent stem cells (hPSCs-CMs) could revolutionise biomedicine. Global burden of heart failure will soon reach USD $90bn, while unexpected cardiotoxicity underlies 28% of drug withdrawals. Advances in hPSC isolation, Cas9/CRISPR genome engineering and hPSC-CM differentiation have improved patient care, progressed drugs to clinic and opened a new era in safety pharmacology. Nevertheless, predictive cardiotoxicity using hPSC-CMs contrasts from failure to almost total success. Since this likely relates to cell immaturity, efforts are underway to use biochemical and biophysical cues to improve many of the ~30 structural and functional properties of hPSC-CMs towards those seen in adult CMs. Other developments needed for widespread hPSC-CM utility include subtype specification, cost reduction of large scale differentiation and elimination of the phenotyping bottleneck. This review will consider these factors in the evolution of hPSC-CM technologies, as well as their integration into high content industrial platforms that assess structure, mitochondrial function, electrophysiology, calcium transients and contractility. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Chris Denning
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom.
| | - Viola Borgdorff
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - James Crutchley
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Karl S A Firth
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Vinoj George
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Spandan Kalra
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Alexander Kondrashov
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Minh Duc Hoang
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Diogo Mosqueira
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Asha Patel
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Ljupcho Prodanov
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Divya Rajamohan
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - William C Skarnes
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, United Kingdom
| | - James G W Smith
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| | - Lorraine E Young
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, NG7 2RD, United Kingdom
| |
Collapse
|
25
|
|
26
|
Singh P, Sharma R, McElhanon K, Allen CD, Megyesi JK, Beneš H, Singh SP. Sulforaphane protects the heart from doxorubicin-induced toxicity. Free Radic Biol Med 2015; 86:90-101. [PMID: 26025579 PMCID: PMC4554811 DOI: 10.1016/j.freeradbiomed.2015.05.028] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 05/04/2015] [Accepted: 05/19/2015] [Indexed: 01/08/2023]
Abstract
Cardiotoxicity is one of the major side effects encountered during cancer chemotherapy with doxorubicin (DOX) and other anthracyclines. Previous studies have shown that oxidative stress caused by DOX is one of the primary mechanisms for its toxic effects on the heart. Since the redox-sensitive transcription factor, Nrf2, plays a major role in protecting cells from the toxic metabolites generated during oxidative stress, we examined the effects of the phytochemical sulforaphane (SFN), a potent Nrf2-activating agent, on DOX-induced cardiotoxicity. These studies were carried out both in vitro and in vivo using rat H9c2 cardiomyoblast cells and wild type 129/sv mice, and involved SFN pretreatment followed by SFN administration during DOX exposure. SFN treatment protected H9c2 cells from DOX cytotoxicity and also resulted in restored cardiac function and a significant reduction in DOX-induced cardiomyopathy and mortality in mice. Specificity of SFN induction of Nrf2 and protection of H9c2 cells was demonstrated in Nrf2 knockdown experiments. Cardiac accumulation of 4-hydroxynonenal (4-HNE) protein adducts, due to lipid peroxidation following DOX-induced oxidative stress, was significantly attenuated by SFN treatment. The respiratory function of cardiac mitochondria isolated from mice exposed to DOX alone was repressed, while SFN treatment with DOX significantly elevated mitochondrial respiratory complex activities. Co-administration of SFN reversed the DOX-associated reduction in nuclear Nrf2 binding activity and restored cardiac expression of Nrf2-regulated genes at both the RNA and protein levels. Together, our results demonstrate for the first time that the Nrf2 inducer, SFN, has the potential to provide protection against DOX-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Preeti Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Rajendra Sharma
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kevin McElhanon
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Charles D Allen
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA
| | - Judit K Megyesi
- Central Arkansas Veterans Healthcare System, Little Rock, AR, USA; Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Helen Beneš
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sharda P Singh
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.
| |
Collapse
|
27
|
Bogatan S, Cevik D, Demidov V, Vanderploeg J, Panchbhaya A, Vitkin A, Jacobs JR. Talin Is Required Continuously for Cardiomyocyte Remodeling during Heart Growth in Drosophila. PLoS One 2015; 10:e0131238. [PMID: 26110760 PMCID: PMC4482443 DOI: 10.1371/journal.pone.0131238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 05/30/2015] [Indexed: 12/26/2022] Open
Abstract
Mechanotransduction of tension can govern the remodeling of cardiomyocytes during growth or cardiomyopathy. Tension is signaled through the integrin adhesion complexes found at muscle insertions and costameres but the relative importance of signalling during cardiomyocyte growth versus remodelling has not been assessed. Employing the Drosophila cardiomyocyte as a genetically amenable model, we depleted the levels of Talin, a central component of the integrin adhesion complex, at different stages of heart growth and remodeling. We demonstrate a continuous requirement for Talin during heart growth to maintain the one-to-one apposition of myofibril ends between cardiomyocytes. Retracted myofibrils cannot regenerate appositions to adjacent cells after restoration of normal Talin expression, and the resulting deficit reduces heart contraction and lifespan. Reduction of Talin during heart remodeling after hatching or during metamorphosis results in pervasive degeneration of cell contacts, myofibril length and number, for which restored Talin expression is insufficient for regeneration. Resultant dilated cardiomyopathy results in a fibrillating heart with poor rhythmicity. Cardiomyocytes have poor capacity to regenerate deficits in myofibril orientation and insertion, despite an ongoing capacity to remodel integrin based adhesions.
Collapse
Affiliation(s)
- Simina Bogatan
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Duygu Cevik
- Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Valentin Demidov
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jessica Vanderploeg
- Department of Biology, Taylor University, Euler Science Complex, 236 W. Reade Ave, Upland, IN, 46989, United States of America
| | | | - Alex Vitkin
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - J. Roger Jacobs
- Department of Biology, McMaster University, Hamilton, ON, Canada
- * E-mail:
| |
Collapse
|
28
|
Rodriguez ML, Graham BT, Pabon LM, Han SJ, Murry CE, Sniadecki NJ. Measuring the contractile forces of human induced pluripotent stem cell-derived cardiomyocytes with arrays of microposts. J Biomech Eng 2015; 136:051005. [PMID: 24615475 DOI: 10.1115/1.4027145] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/10/2014] [Indexed: 12/31/2022]
Abstract
Human stem cell-derived cardiomyocytes hold promise for heart repair, disease modeling, drug screening, and for studies of developmental biology. All of these applications can be improved by assessing the contractility of cardiomyocytes at the single cell level. We have developed an in vitro platform for assessing the contractile performance of stem cell-derived cardiomyocytes that is compatible with other common endpoints such as microscopy and molecular biology. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were seeded onto elastomeric micropost arrays in order to characterize the contractile force, velocity, and power produced by these cells. We assessed contractile function by tracking the deflection of microposts beneath an individual hiPSC-CM with optical microscopy. Immunofluorescent staining of these cells was employed to assess their spread area, nucleation, and sarcomeric structure on the microposts. Following seeding of hiPSC-CMs onto microposts coated with fibronectin, laminin, and collagen IV, we found that hiPSC-CMs on laminin coatings demonstrated higher attachment, spread area, and contractile velocity than those seeded on fibronectin or collagen IV coatings. Under optimized conditions, hiPSC-CMs spread to an area of approximately 420 μm2, generated systolic forces of approximately 15 nN/cell, showed contraction and relaxation rates of 1.74 μm/s and 1.46 μm/s, respectively, and had a peak contraction power of 29 fW. Thus, elastomeric micropost arrays can be used to study the contractile strength and kinetics of hiPSC-CMs. This system should facilitate studies of hiPSC-CM maturation, disease modeling, and drug screens as well as fundamental studies of human cardiac contraction.
Collapse
|
29
|
Backes C, Rühle F, Stoll M, Haas J, Frese K, Franke A, Lieb W, Wichmann HE, Weis T, Kloos W, Lenhof HP, Meese E, Katus H, Meder B, Keller A. Systematic permutation testing in GWAS pathway analyses: identification of genetic networks in dilated cardiomyopathy and ulcerative colitis. BMC Genomics 2014; 15:622. [PMID: 25052024 PMCID: PMC4223581 DOI: 10.1186/1471-2164-15-622] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/17/2014] [Indexed: 12/20/2022] Open
Abstract
Background Genome wide association studies (GWAS) are applied to identify genetic loci, which are associated with complex traits and human diseases. Analogous to the evolution of gene expression analyses, pathway analyses have emerged as important tools to uncover functional networks of genome-wide association data. Usually, pathway analyses combine statistical methods with a priori available biological knowledge. To determine significance thresholds for associated pathways, correction for multiple testing and over-representation permutation testing is applied. Results We systematically investigated the impact of three different permutation test approaches for over-representation analysis to detect false positive pathway candidates and evaluate them on genome-wide association data of Dilated Cardiomyopathy (DCM) and Ulcerative Colitis (UC). Our results provide evidence that the gold standard - permuting the case–control status – effectively improves specificity of GWAS pathway analysis. Although permutation of SNPs does not maintain linkage disequilibrium (LD), these permutations represent an alternative for GWAS data when case–control permutations are not possible. Gene permutations, however, did not add significantly to the specificity. Finally, we provide estimates on the required number of permutations for the investigated approaches. Conclusions To discover potential false positive functional pathway candidates and to support the results from standard statistical tests such as the Hypergeometric test, permutation tests of case control data should be carried out. The most reasonable alternative was case–control permutation, if this is not possible, SNP permutations may be carried out. Our study also demonstrates that significance values converge rapidly with an increasing number of permutations. By applying the described statistical framework we were able to discover axon guidance, focal adhesion and calcium signaling as important DCM-related pathways and Intestinal immune network for IgA production as most significant UC pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
30
|
Di Scipio F, Sprio A, Folino A, Carere M, Salamone P, Yang Z, Berrone M, Prat M, Losano G, Rastaldo R, Berta G. Injured cardiomyocytes promote dental pulp mesenchymal stem cell homing. Biochim Biophys Acta Gen Subj 2014; 1840:2152-61. [DOI: 10.1016/j.bbagen.2014.03.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 02/28/2014] [Accepted: 03/05/2014] [Indexed: 02/07/2023]
|
31
|
Kaushik G, Engler AJ. From stem cells to cardiomyocytes: the role of forces in cardiac maturation, aging, and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 126:219-42. [PMID: 25081620 DOI: 10.1016/b978-0-12-394624-9.00009-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stem cell differentiation into a variety of lineages is known to involve signaling from the extracellular niche, including from the physical properties of that environment. What regulates stem cell responses to these cues is there ability to activate different mechanotransductive pathways. Here, we will review the structures and pathways that regulate stem cell commitment to a cardiomyocyte lineage, specifically examining proteins within muscle sarcomeres, costameres, and intercalated discs. Proteins within these structures stretch, inducing a change in their phosphorylated state or in their localization to initiate different signals. We will also put these changes in the context of stem cell differentiation into cardiomyocytes, their subsequent formation of the chambered heart, and explore negative signaling that occurs during disease.
Collapse
Affiliation(s)
- Gaurav Kaushik
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
32
|
Chopra A, Murray ME, Byfield FJ, Mendez MG, Halleluyan R, Restle DJ, Raz-Ben Aroush D, Galie PA, Pogoda K, Bucki R, Marcinkiewicz C, Prestwich GD, Zarembinski TI, Chen CS, Puré E, Kresh JY, Janmey PA. Augmentation of integrin-mediated mechanotransduction by hyaluronic acid. Biomaterials 2014; 35:71-82. [PMID: 24120037 PMCID: PMC3930571 DOI: 10.1016/j.biomaterials.2013.09.066] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 09/20/2013] [Indexed: 12/26/2022]
Abstract
Changes in tissue and organ stiffness occur during development and are frequently symptoms of disease. Many cell types respond to the stiffness of substrates and neighboring cells in vitro and most cell types increase adherent area on stiffer substrates that are coated with ligands for integrins or cadherins. In vivo cells engage their extracellular matrix (ECM) by multiple mechanosensitive adhesion complexes and other surface receptors that potentially modify the mechanical signals transduced at the cell/ECM interface. Here we show that hyaluronic acid (also called hyaluronan or HA), a soft polymeric glycosaminoglycan matrix component prominent in embryonic tissue and upregulated during multiple pathologic states, augments or overrides mechanical signaling by some classes of integrins to produce a cellular phenotype otherwise observed only on very rigid substrates. The spread morphology of cells on soft HA-fibronectin coated substrates, characterized by formation of large actin bundles resembling stress fibers and large focal adhesions resembles that of cells on rigid substrates, but is activated by different signals and does not require or cause activation of the transcriptional regulator YAP. The fact that HA production is tightly regulated during development and injury and frequently upregulated in cancers characterized by uncontrolled growth and cell movement suggests that the interaction of signaling between HA receptors and specific integrins might be an important element in mechanical control of development and homeostasis.
Collapse
Affiliation(s)
- Anant Chopra
- Dept. of Cardiothoracic Surgery, Drexel Univ. College of Med, Philadelphia, PA, USA; Dept. of Bioengineering, Univ. of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Roth BJ. Boundary Layers and the Distribution of Membrane Forces Predicted by the Mechanical Bidomain Model. MECHANICS RESEARCH COMMUNICATIONS 2013; 50:12-16. [PMID: 23772096 PMCID: PMC3678842 DOI: 10.1016/j.mechrescom.2013.02.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The mechanical bidomain model is a mathematical description of the elastic properties of cardiac tissue. The unique feature of the bidomain model is that it is a macroscopic continuum representation of tissue that nevertheless accounts for the intracellular and extracellular spaces individually, thereby focusing on mechanical forces arising across the cell membrane. In this paper, the mechanical bidomain model describes a two-dimensional sheet of cardiac tissue undergoing a uniform active tension. At the boundary, the tissue sheet is free to move. Analytical solutions are found for the intracellular and extracellular displacements and pressures. The model predicts that membrane forces, which may be responsible for phenomena such as mechanotransduction and remodeling, are large near the tissue boundary, and fall off rapidly with distance from the boundary.
Collapse
Affiliation(s)
- Bradley J Roth
- Department of Physics Oakland University Rochester, Michigan
| |
Collapse
|
34
|
Dongaonkar RM, Stewart RH, Quick CM, Uray KL, Cox CS, Laine GA. Award article: Microcirculatory Society Award for Excellence in Lymphatic Research: time course of myocardial interstitial edema resolution and associated left ventricular dysfunction. Microcirculation 2013; 19:714-22. [PMID: 22708850 DOI: 10.1111/j.1549-8719.2012.00204.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Although the causal relationship between acute myocardial edema and cardiac dysfunction has been established, resolution of myocardial edema and subsequent recovery of cardiac function have not been established. The time to resolve myocardial edema and the degree that cardiac function is depressed after edema resolves are not known. We therefore characterized temporal changes in cardiac function as acute myocardial edema formed and resolved. METHODS Acute myocardial edema was induced in the canine model by elevating coronary sinus pressure for three hours. Myocardial water content and cardiac function were determined before and during coronary sinus pressure elevation, and after coronary sinus pressure restoration. RESULTS Although no change in systolic properties was detected, accumulation of water in myocardial interstitium was associated with increased diastolic stiffness. When coronary sinus pressure was relieved, myocardial edema resolved within 180 minutes. Diastolic stiffness, however, remained significantly elevated compared with baseline values, and cardiac function remained compromised. CONCLUSIONS The present work suggests that the cardiac dysfunction caused by the formation of myocardial edema may persist after myocardial edema resolves. With the advent of new imaging techniques to quantify myocardial edema, this insight provides a new avenue for research to detect and treat a significant cause of cardiac dysfunction.
Collapse
Affiliation(s)
- Ranjeet M Dongaonkar
- Michael E DeBakey Institute, Texas A&M University, College Station, Texas 77843-4466, USA
| | | | | | | | | | | |
Collapse
|
35
|
Novaes RD, Penitente AR, Gonçalves RV, Talvani A, Peluzio MCG, Neves CA, Natali AJ, Maldonado IRSC. Trypanosoma cruzi infection induces morphological reorganization of the myocardium parenchyma and stroma, and modifies the mechanical properties of atrial and ventricular cardiomyocytes in rats. Cardiovasc Pathol 2013; 22:270-9. [PMID: 23541389 DOI: 10.1016/j.carpath.2012.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/01/2012] [Accepted: 12/05/2012] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND This study investigates morphofunctional adaptations of the heart stroma and parenchyma in rats that are chronically infected with Trypanosoma cruzi. METHODS Four-month-old male Wistar rats were randomized into control (n=14) and infected (n=14) groups. Infected animals were inoculated with T. cruzi Y strain. After 9 weeks, the animals were euthanized, and the right atrium (RA) and left ventricle (LV) were removed for biochemical, stereological, and cardiomyocyte mechanical analyses. RESULTS Infected animals presented cardiomyocyte atrophy and myocardial fibrosis. For these animals, the total volume, length, surface area, and cross-sectional area of cardiomyocytes were significantly reduced, and the total interstitial and collagen volumes were significantly increased in the RA and LV compared to the controls. The total volume and length of blood vessels were significantly increased in the LV, and the total blood vessel surface area was significantly higher in the RA of infected animals. RA and LV cardiomyocytes from infected animals exhibited a significant reduction in cell shortening (43.02% and 24.98%, respectively), prolongation of the time to the peak of contraction (17.09%) and the time to half relaxation (23.68%) compared to non-infected animals. Lipid hydroperoxides, but not mineral concentrations, were significantly increased in the RA and LV from infected animals, showing an inverse correlation with cell shortening. CONCLUSIONS T. cruzi infection induces global structural remodeling of the RA and LV in rats. This remodeling coexists with cardiomyocyte contractility dysfunction, which is possibly related to the abnormal organization of the myocardial stroma and increased cellular lipid peroxidation.
Collapse
Affiliation(s)
- Rômulo D Novaes
- Department of General Biology, Federal University of Viçosa, MG, 36570-000, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
The extracellular matrix (ECM) is a complex entity containing a large portfolio of structural proteins, signaling molecules, and proteases. Changes in the overall integrity and activational state of these ECM constituents can contribute to tissue structure and function, which is certainly true of the myocardium. Changes in the expression patterns and activational states of a family of ECM proteolytic enzymes, the matrix metalloproteinases (MMPs), have been identified in all forms of left ventricle remodeling and can be a contributory factor in the progression to heart failure. However, new clinical and basic research has identified some surprising and unpredicted changes in MMP profiles in left ventricle remodeling processes, such as with pressure or volume overload, as well as with myocardial infarction. From these studies, it has become recognized that proteolytic processing of signaling molecules by certain MMP types, particularly the transmembrane MMPs, actually may facilitate ECM accumulation and modulate fibroblast transdifferentiation; both are critical events in adverse left ventricle remodeling. Based on the ever-increasing substrates and diversity of biological actions of MMPs, it is likely that continued research about the relationship of left ventricle remodeling in this family of proteases will yield new insights into the ECM remodeling process and new therapeutic targets.
Collapse
Affiliation(s)
- Francis G Spinale
- Cardiovascular Translational Research Center, CBA, University of South Carolina School of Medicine, 6439 Garners Ferry Rd, Columbia, SC, USA.
| | | | | |
Collapse
|
37
|
Abstract
The mechanical bidomain model is a new mathematical description of the elastic behavior of cardiac tissue. Its primary advantage over previous models is that it accounts for forces acting across the cell membrane arising form differences in the displacement of the intracellular and extracellular spaces. In this review, I describe the development of the mechanical bidomain model. I emphasize new predictions of the model, such as the existence of boundary layers at the tissue surface where the membrane forces are large, and pressure differences between the intracellular and extracellular spaces. Although the theoretical analysis is quite mathematical, I highlight the types of experiments that could be used to test the model predictions. Finally, I present open questions about the mechanical bidomain model that may be productive future directions for research.
Collapse
|
38
|
Transcriptome profiling of peripheral blood cells identifies potential biomarkers for doxorubicin cardiotoxicity in a rat model. PLoS One 2012; 7:e48398. [PMID: 23209553 PMCID: PMC3507887 DOI: 10.1371/journal.pone.0048398] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 09/25/2012] [Indexed: 02/08/2023] Open
Abstract
Aims Doxorubicin (DOX), a widely used anticancer agent, can cause an unpredictable cardiac toxicity which remains a major limitation in cancer chemotherapy. There is a need for noninvasive, sensitive and specific biomarkers which will allow identifying patients at risk for DOX-induced cardiotoxicity to prevent permanent cardiac damage. The aim of this study was to investigate whether the expression of specific genes in the peripheral blood can be used as surrogate marker(s) for DOX-induced cardiotoxicity. Methods/Results Rats were treated with a single dose of DOX similar to one single dose that is often administered in humans. The cardiac and peripheral blood mononuclear cells (PBMCs) genome-wide expression profiling were examined using Illumina microarrays. The results showed 4,409 differentially regulated genes (DRG) in the hearts and 4,120 DRG in PBMC. Of these 2411 genes were similarly DRG (SDRG) in both the heart and PBMC. Pathway analysis of the three datasets of DRG using Gene Ontology (GO) enrichment analysis and Ingenuity Pathways Analysis (IPA) showed that most of the genes in these datasets fell into pathways related to oxidative stress response and protein ubiquination. IPA search for potential eligible biomarkers for cardiovascular disease within the SDRG list revealed 188 molecules. Conclusions We report the first in-depth comparison of DOX-induced global gene expression profiles of hearts and PBMCs. The high similarity between the gene expression profiles of the heart and PBMC induced by DOX indicates that the PBMC transcriptome may serve as a surrogate marker of DOX-induced cardiotoxicity. Future directions of this research will include analysis of PBMC expression profiles of cancer patients treated with DOX-based chemotherapy to identify the cardiotoxicity risk, predict DOX-treatment response and ultimately to allow individualized anti-cancer therapy.
Collapse
|
39
|
Goldsmith EC, Bradshaw AD, Spinale FG. Cellular mechanisms of tissue fibrosis. 2. Contributory pathways leading to myocardial fibrosis: moving beyond collagen expression. Am J Physiol Cell Physiol 2012; 304:C393-402. [PMID: 23174564 DOI: 10.1152/ajpcell.00347.2012] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
While the term "fibrosis" can be misleading in terms of the complex patterns and processes of myocardial extracellular matrix (ECM) remodeling, fibrillar collagen accumulation is a common consequence of relevant pathophysiological stimuli, such as pressure overload (PO) and myocardial infarction (MI). Fibrillar collagen accumulation in both PO and MI is predicated on a number of diverse cellular and extracellular events, which include changes in fibroblast phenotype (transdifferentiation), posttranslational processing and assembly, and finally, degradation. The expansion of a population of transformed fibroblasts/myofibroblasts is a significant cellular event with respect to ECM remodeling in both PO and MI. The concept that this cellular expansion within the myocardial ECM may be due, at least in part, to endothelial-mesenchymal transformation and thereby not dissimilar to events observed in cancer progression holds intriguing future possibilities. Studies regarding determinants of procollagen processing, such as procollagen C-endopeptidase enhancer (PCOLCE), and collagen assembly, such as the secreted protein acidic and rich in cysteine (SPARC), have identified potential new targets for modifying the fibrotic response in both PO and MI. Finally, the transmembrane matrix metalloproteinases, such as MMP-14, underscore the diversity and complexity of this ECM proteolytic family as this protease can degrade the ECM as well as induce a profibrotic response. The growing recognition that the myocardial ECM is a dynamic entity containing a diversity of matricellular and nonstructural proteins as well as proteases and that the fibrillar collagens can change in structure and content in a rapid temporal fashion has opened up new avenues for modulating what was once considered an irreversible event--myocardial fibrosis.
Collapse
Affiliation(s)
- Edie C Goldsmith
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29208, USA
| | | | | |
Collapse
|
40
|
Shachar M, Benishti N, Cohen S. Effects of mechanical stimulation induced by compression and medium perfusion on cardiac tissue engineering. Biotechnol Prog 2012; 28:1551-9. [PMID: 22961835 DOI: 10.1002/btpr.1633] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 07/31/2012] [Indexed: 11/12/2022]
Abstract
Cardiac tissue engineering presents a challenge due to the complexity of the muscle tissue and the need for multiple signals to induce tissue regeneration in vitro. We investigated the effects of compression (1 Hz, 15% strain) combined with fluid shear stress (10(-2) -10(-1) dynes/cm(2) ) provided by medium perfusion on the outcome of cardiac tissue engineering. Neonatal rat cardiac cells were seeded in Arginine-Glycine-Aspartate (RGD)-attached alginate scaffolds, and the constructs were cultivated in a compression bioreactor. A daily, short-term (30 min) compression (i.e., "intermittent compression") for 4 days induced the formation of cardiac tissue with typical striation, while in the continuously compressed constructs (i.e., "continuous compression"), the cells remained spherical. By Western blot, on day 4 the expression of the gap junction protein connexin 43 was significantly greater in the "intermittent compression" constructs and the cardiomyocyte markers (α-actinin and N-cadherin) showed a trend of better preservation compared to the noncompressed constructs. This regime of compression had no effect on the proliferation of nonmyocyte cells, which maintained low expression level of proliferating cell nuclear antigen. Elevated secretion levels of basic fibroblast growth factor and transforming growth factor-β in the daily, intermittently compressed constructs likely attributed to tissue formation. Our study thus establishes the formation of an improved cardiac tissue in vitro, when induced by combined mechanical signals of compression and fluid shear stress provided by perfusion.
Collapse
Affiliation(s)
- Michal Shachar
- The Avram and Stella Goldstein-Goren Dept. of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | | | | |
Collapse
|
41
|
Chopra A, Patel A, Shieh AC, A. Janmey P, Kresh JY. α-Catenin localization and sarcomere self-organization on N-cadherin adhesive patterns are myocyte contractility driven. PLoS One 2012; 7:e47592. [PMID: 23077648 PMCID: PMC3471892 DOI: 10.1371/journal.pone.0047592] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 09/12/2012] [Indexed: 12/18/2022] Open
Abstract
The N-cadherin (N-cad) complex plays a crucial role in cardiac cell structure and function. Cadherins are adhesion proteins linking adjacent cardiac cells and, like integrin adhesions, are sensitive to force transmission. Forces through these adhesions are capable of eliciting structural and functional changes in myocytes. Compared to integrins, the mechanisms of force transduction through cadherins are less explored. α-catenin is a major component of the cadherin-catenin complex, thought to provide a link to the cell actin cytoskeleton. Using N-cad micropatterned substrates in an adhesion constrainment model, the results from this study show that α-catenin localizes to regions of highest internal stress in myocytes. This localization suggests that α-catenin acts as an adaptor protein associated with the cadherin mechanosensory apparatus, which is distinct from mechanosensing through integrins. Myosin inhibition in cells bound by integrins to fibronectin-coated patterns disrupts myofibiril organization, whereas on N-cad coated patterns, myosin inhibition leads to better organized myofibrils. This result indicates that the two adhesion systems provide independent mechanisms for regulating myocyte structural organization.
Collapse
Affiliation(s)
- Anant Chopra
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Akash Patel
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Adrian C. Shieh
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Paul A. Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JYK); (PAJ)
| | - J. Yasha Kresh
- Department of Cardiothoracic Surgery, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail: (JYK); (PAJ)
| |
Collapse
|
42
|
Nabavi N, Pustylnik S, Harrison RE. Rab GTPase mediated procollagen trafficking in ascorbic acid stimulated osteoblasts. PLoS One 2012; 7:e46265. [PMID: 23050002 PMCID: PMC3458846 DOI: 10.1371/journal.pone.0046265] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 08/30/2012] [Indexed: 12/24/2022] Open
Abstract
Despite advances in investigating functional aspects of osteoblast (OB) differentiation, especially studies on how bone proteins are deposited and mineralized, there has been little research on the intracellular trafficking of bone proteins during OB differentiation. Collagen synthesis and secretion is the major function of OBs and is markedly up-regulated upon ascorbic acid (AA) stimulation, significantly more so than in fibroblast cells. Understanding the mechanism by which collagen is mobilized in specialized OB cells is important for both basic cell biology and diseases involving defects in bone protein secretion and deposition. Protein trafficking along the exocytic and endocytic pathways is aided by many molecules, with Rab GTPases being master regulators of vesicle targeting. In this study, we used microarray analysis to identify the Rab GTPases that are up-regulated during a 5-day AA differentiation of OBs, namely Rab1, Rab3d, and Rab27b. Further, we investigated the role of identified Rabs in regulating the trafficking of collagen from the site of synthesis in the ER to the Golgi and ultimately to the plasma membrane utilizing Rab dominant negative (DN) expression. We also observed that experimental halting of biosynthetic trafficking by these mutant Rabs initiated proteasome-mediated degradation of procollagen and ceased global protein translation. Acute expression of Rab1 and Rab3d DN constructs partially alleviated this negative feedback mechanism and resulted in impaired ER to Golgi trafficking of procollagen. Similar expression of Rab27b DN constructs resulted in dispersed collagen vesicles which may represent failed secretory vesicles sequestered in the cytosol. A significant and strong reduction in extracellular collagen levels was also observed implicating the functional importance of Rab1, Rab3d and Rab27b in these major collagen-producing cells.
Collapse
Affiliation(s)
- Noushin Nabavi
- Department of Cell and Systems Biology, University of Tronto Scarborought, Toronto, Ontario, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Sofia Pustylnik
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
| | - Rene E. Harrison
- Department of Cell and Systems Biology, University of Tronto Scarborought, Toronto, Ontario, Canada
- Department of Biological Sciences, University of Toronto Scarborough, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
43
|
Simmons CS, Petzold BC, Pruitt BL. Microsystems for biomimetic stimulation of cardiac cells. LAB ON A CHIP 2012; 12:3235-48. [PMID: 22782590 DOI: 10.1039/c2lc40308k] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The heart is a complex integrated system that leverages mechanoelectrical signals to synchronize cardiomyocyte contraction and push blood throughout the body. The correct magnitude, timing, and distribution of these signals is critical for proper functioning of the heart; aberrant signals can lead to acute incidents, long-term pathologies, and even death. Due to the heart's limited regenerative capacity and the wide variety of pathologies, heart disease is often studied in vitro. However, it is difficult to accurately replicate the cardiac environment outside of the body. Studying the biophysiology of the heart in vitro typically consists of studying single cells in a tightly controlled static environment or whole tissues in a complex dynamic environment. Micro-electromechanical systems (MEMS) allow us to bridge these two extremes by providing increasing complexity for cell culture without having to use a whole tissue. Here, we carefully describe the electromechanical environment of the heart and discuss MEMS specifically designed to replicate these stimulation modes. Strengths, limitations and future directions of various designs are discussed for a variety of applications.
Collapse
Affiliation(s)
- Chelsey S Simmons
- Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | | | | |
Collapse
|
44
|
Sheehy SP, Grosberg A, Parker KK. The contribution of cellular mechanotransduction to cardiomyocyte form and function. Biomech Model Mechanobiol 2012; 11:1227-39. [PMID: 22772714 DOI: 10.1007/s10237-012-0419-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 06/25/2012] [Indexed: 01/07/2023]
Abstract
Myocardial development is regulated by an elegantly choreographed ensemble of signaling events mediated by a multitude of intermediates that take a variety of forms. Cellular differentiation and maturation are a subset of vertically integrated processes that extend over several spatial and temporal scales to create a well-defined collective of cells that are able to function cooperatively and reliably at the organ level. Early efforts to understand the molecular mechanisms of cardiomyocyte fate determination focused primarily on genetic and chemical mediators of this process. However, increasing evidence suggests that mechanical interactions between the extracellular matrix (ECM) and cell surface receptors as well as physical interactions between neighboring cells play important roles in regulating the signaling pathways controlling the developmental processes of the heart. Interdisciplinary efforts have made it apparent that the influence of the ECM on cellular behavior occurs through a multitude of physical mechanisms, such as ECM boundary conditions, elasticity, and the propagation of mechanical signals to intracellular compartments, such as the nucleus. In addition to experimental studies, a number of mathematical models have been developed that attempt to capture the interplay between cells and their local microenvironment and the influence these interactions have on cellular self-assembly and functional behavior. Nevertheless, many questions remain unanswered concerning the mechanism through which physical interactions between cardiomyocytes and their environment are translated into biochemical cellular responses and how these signaling modalities can be utilized in vitro to fabricate myocardial tissue constructs from stem cell-derived cardiomyocytes that more faithfully represent their in vivo counterpart. These studies represent a broad effort to characterize biological form as a conduit for information transfer that spans the nanometer length scale of proteins to the meter length scale of the patient and may yield new insights into the contribution of mechanotransduction into heart development and disease.
Collapse
Affiliation(s)
- Sean P Sheehy
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, School of Engineering and Applied Sciences, Harvard University, Pierce Hall Rm. 321, 29 Oxford St., Cambridge, MA 02138, USA
| | | | | |
Collapse
|
45
|
Balse E, Steele DF, Abriel H, Coulombe A, Fedida D, Hatem SN. Dynamic of Ion Channel Expression at the Plasma Membrane of Cardiomyocytes. Physiol Rev 2012; 92:1317-58. [DOI: 10.1152/physrev.00041.2011] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac myocytes are characterized by distinct structural and functional entities involved in the generation and transmission of the action potential and the excitation-contraction coupling process. Key to their function is the specific organization of ion channels and transporters to and within distinct membrane domains, which supports the anisotropic propagation of the depolarization wave. This review addresses the current knowledge on the molecular actors regulating the distinct trafficking and targeting mechanisms of ion channels in the highly polarized cardiac myocyte. In addition to ubiquitous mechanisms shared by other excitable cells, cardiac myocytes show unique specialization, illustrated by the molecular organization of myocyte-myocyte contacts, e.g., the intercalated disc and the gap junction. Many factors contribute to the specialization of the cardiac sarcolemma and the functional expression of cardiac ion channels, including various anchoring proteins, motors, small GTPases, membrane lipids, and cholesterol. The discovery of genetic defects in some of these actors, leading to complex cardiac disorders, emphasizes the importance of trafficking and targeting of ion channels to cardiac function. A major challenge in the field is to understand how these and other actors work together in intact myocytes to fine-tune ion channel expression and control cardiac excitability.
Collapse
Affiliation(s)
- Elise Balse
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David F. Steele
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Hugues Abriel
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Alain Coulombe
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David Fedida
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Stéphane N. Hatem
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| |
Collapse
|
46
|
Tong Z, Jia X. Biomaterials-Based Strategies for the Engineering of Mechanically Active Soft Tissues. MRS COMMUNICATIONS 2012; 2:31-39. [PMID: 25250199 PMCID: PMC4169986 DOI: 10.1557/mrc.2012.4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 03/09/2012] [Indexed: 06/03/2023]
Abstract
Load-bearing, mechanically active tissues are routinely subjected to non-linear mechanical deformations. Consequently, these tissues exhibit complex mechanical properties and unique tissue organizations. Successful engineering of mechanically active tissues relies on the integration of the mechanical sensing mechanism found in the native tissues into polymeric scaffolds. Intelligent biomaterials that closely mimic the structural organizations and multi-scale responsiveness of the natural extracellular matrices (ECM), when strategically combined with multipotent cells and dynamic culture devices that generate physiologically relevant physical forces, will lead to the creation of artificial tissues that are mechanically robust and biologically functional.
Collapse
Affiliation(s)
- Zhixiang Tong
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Delaware Biotechnology Institute, University of Delaware, Newark, DE 19716
| | - Xinqiao Jia
- Department of Materials Science and Engineering, Department of Biomedical Engineering, Delaware Biotechnology Institute, University of Delaware, Newark, DE 19716
| |
Collapse
|
47
|
2D and 3D-organized cardiac cells shows differences in cellular morphology, adhesion junctions, presence of myofibrils and protein expression. PLoS One 2012; 7:e38147. [PMID: 22662278 PMCID: PMC3360656 DOI: 10.1371/journal.pone.0038147] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Accepted: 04/30/2012] [Indexed: 12/03/2022] Open
Abstract
Cardiac cells are organized in vivo in a complex tridimensional structural organization that is crucial for heart function. While in vitro studies can reveal details about cardiac cell biology, usually cells are grown on simplified two-dimensional (2D) environments. To address these differences, we established a cardiac cell culture composed of both 2D and three-dimensional (3D)-organized cells. Our results shows significant differences between the two culture contexts in relation to the overall morphology of the cells, contraction ability, proliferation rate, presence of intercellular adhesion structures, organization of myofibrils, mitochondria morphology, endoplasmic reticulum contents, cytoskeletal filaments and extracellular matrix distribution, and expression of markers of cardiac differentiation. Cardiac cells grown in 2D-context displayed a flattened and well spread shape, were mostly isolated and their cytoplasm was filled with a large network of microfilaments and microtubules. In contrast, 3D-cells were smaller in size, were always in close contact with each other with several cellular junctions, and displayed a less conspicuous cytoskeletal network. 3D-cells had more mitochondria and myofibrils and these cells contract spontaneously more often than 2D-cells. On the other hand, endoplasmic reticulum membranes were present in higher amounts in 2D-cells when compared to 3D-cells. The expression of desmin, cadherin and alpha-actinin was higher in 3D-aggregates compared to 2D-spread cells. These findings indicate that the tridimensional environment in which the cardiac cells are grown influence several aspects of cardiac differentiation, including cell adhesion, cell shape, myofibril assembly, mitochondria contents and protein expression. We suggest that the use of this cardiac culture model, with 2D and 3D-context cells, could be useful for studies on the effects of different drugs, or growth factors, giving valuable information on the biological response of cells grown in different spatial organizations.
Collapse
|
48
|
Punal VM, Roth BJ. A perturbation solution of the mechanical bidomain model. Biomech Model Mechanobiol 2011; 11:995-1000. [PMID: 22200886 DOI: 10.1007/s10237-011-0368-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 12/13/2011] [Indexed: 02/06/2023]
Abstract
This research focuses on finding analytical solutions to the mechanical bidomain model for cardiac tissue. In particular, a perturbation expansion is used to analyze the equations, with the perturbation parameter being inversely proportional to the spring constant coupling the intracellular and extracellular spaces. The results indicate that the intracellular and extracellular pressures are not equal and that the two spaces can move relative to each other. This calculation is complicated enough to illustrate the implications of the mechanical bidomain model but is nevertheless simple enough to solve analytically. One application of the calculation is to the mechanical behavior of active cardiac tissue surrounding an ischemic region.
Collapse
Affiliation(s)
- Vanessa M Punal
- Department of Physics, Oakland University, Rochester, MI 48309, USA.
| | | |
Collapse
|
49
|
Kojic S, Radojkovic D, Faulkner G. Muscle ankyrin repeat proteins: their role in striated muscle function in health and disease. Crit Rev Clin Lab Sci 2011; 48:269-94. [DOI: 10.3109/10408363.2011.643857] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
de Tombe PP, Granzier HL. The cytoskeleton and the cellular transduction of mechanical strain in the heart: a special issue. Pflugers Arch 2011; 462:1-2. [PMID: 21594569 DOI: 10.1007/s00424-011-0976-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 01/15/2023]
|