1
|
Niro F, Fernandes S, Cassani M, Apostolico M, Oliver-De La Cruz J, Pereira-Sousa D, Pagliari S, Vinarsky V, Zdráhal Z, Potesil D, Pustka V, Pompilio G, Sommariva E, Rovina D, Maione AS, Bersanini L, Becker M, Rasponi M, Forte G. Fibrotic extracellular matrix impacts cardiomyocyte phenotype and function in an iPSC-derived isogenic model of cardiac fibrosis. Transl Res 2024; 273:58-77. [PMID: 39025226 DOI: 10.1016/j.trsl.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/14/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
Cardiac fibrosis occurs following insults to the myocardium and is characterized by the abnormal accumulation of non-compliant extracellular matrix (ECM), which compromises cardiomyocyte contractile activity and eventually leads to heart failure. This phenomenon is driven by the activation of cardiac fibroblasts (cFbs) to myofibroblasts and results in changes in ECM biochemical, structural and mechanical properties. The lack of predictive in vitro models of heart fibrosis has so far hampered the search for innovative treatments, as most of the cellular-based in vitro reductionist models do not take into account the leading role of ECM cues in driving the progression of the pathology. Here, we devised a single-step decellularization protocol to obtain and thoroughly characterize the biochemical and micro-mechanical properties of the ECM secreted by activated cFbs differentiated from human induced pluripotent stem cells (iPSCs). We activated iPSC-derived cFbs to the myofibroblast phenotype by tuning basic fibroblast growth factor (bFGF) and transforming growth factor beta 1 (TGF-β1) signalling and confirmed that activated cells acquired key features of myofibroblast phenotype, like SMAD2/3 nuclear shuttling, the formation of aligned alpha-smooth muscle actin (α-SMA)-rich stress fibres and increased focal adhesions (FAs) assembly. Next, we used Mass Spectrometry, nanoindentation, scanning electron and confocal microscopy to unveil the characteristic composition and the visco-elastic properties of the abundant, collagen-rich ECM deposited by cardiac myofibroblasts in vitro. Finally, we demonstrated that the fibrotic ECM activates mechanosensitive pathways in iPSC-derived cardiomyocytes, impacting on their shape, sarcomere assembly, phenotype, and calcium handling properties. We thus propose human bio-inspired decellularized matrices as animal-free, isogenic cardiomyocyte culture substrates recapitulating key pathophysiological changes occurring at the cellular level during cardiac fibrosis.
Collapse
Affiliation(s)
- Francesco Niro
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Soraia Fernandes
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Marco Cassani
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Monica Apostolico
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Jorge Oliver-De La Cruz
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
| | - Daniel Pereira-Sousa
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; Masaryk University, Faculty of Medicine, Department of Biomedical Sciences, Brno 62500, Czech Republic
| | - Stefania Pagliari
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK
| | - Vladimir Vinarsky
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno
| | - Zbyněk Zdráhal
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - David Potesil
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Vaclav Pustka
- Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| | - Giulio Pompilio
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy; Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milan, Italy
| | - Elena Sommariva
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Davide Rovina
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | - Angela Serena Maione
- Unit of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino-IRCCS, Milan, Italy
| | | | | | - Marco Rasponi
- Department of Electronics, Informatics and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC), St Anne's University Hospital Brno; School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, UK.
| |
Collapse
|
2
|
Erhardt J, Ludwig S, Brock J, Hörning M. Native mechano-regulative matrix properties stabilize alternans dynamics and reduce spiral wave stabilization in cardiac tissue. FRONTIERS IN NETWORK PHYSIOLOGY 2024; 4:1443156. [PMID: 39381499 PMCID: PMC11458432 DOI: 10.3389/fnetp.2024.1443156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024]
Abstract
The stability of wave conduction in the heart is strongly related to the proper interplay between the electrophysiological activation and mechanical contraction of myocytes and extracellular matrix (ECM) properties. In this study, we statistically compare bioengineered cardiac tissues cultured on soft hydrogels ( E ≃ 12 kPa) and rigid glass substrates by focusing on the critical threshold of alternans, network-physiological tissue properties, and the formation of stable spiral waves that manifest after wave breakups. For the classification of wave dynamics, we use an improved signal oversampling technique and introduce simple probability maps to identify and visualize spatially concordant and discordant alternans as V- and X-shaped probability distributions. We found that cardiac tissues cultured on ECM-mimicking soft hydrogels show a lower variability of the calcium transient durations among cells in the tissue. This lowers the likelihood of forming stable spiral waves because of the larger dynamical range that tissues can be stably entrained with to form alternans and larger spatial spiral tip movement that increases the chance of self-termination on the tissue boundary. Conclusively, we show that a dysfunction in the excitation-contraction coupling dynamics facilitates life-threatening arrhythmic states such as spiral waves and, thus, highlights the importance of the network-physiological interplay between contractile myocytes and the ECM.
Collapse
Affiliation(s)
| | | | | | - Marcel Hörning
- Institute of Biomaterials and Biomolecular Systems, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
3
|
Gallemí-Pérez A, Tarantola M. Electric Cell-Substrate Impedance Sensing as a Tool to Characterize Wound Healing Dynamics. Methods Mol Biol 2024; 2828:119-145. [PMID: 39147975 DOI: 10.1007/978-1-0716-4023-4_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The electric cell-substrate impedance sensing (ECIS) is a well-established technique that allows for the real-time monitoring of cell cultures growing on gold-electrodes embedded in culture dishes. Its foundation lays on the insulating effect that cells present against the free-flow of electrons, as these passive electrical properties generate a characteristic complex impedance spectrum when a small-amplitude, non-invasive alternating current (AC) is provided through the electrodes, the living cells, and the culture media in the culture ware. In addition, it possesses the ability to create a wound that is highly confined to the electrode area by simply increasing the amplitude of the AC current in dependence of the pre-resistor strength for a defined pulse duration and at a specific frequency. Therefore, it represents a controlled and reproducible tool to carry out in vitro wound healing experiments. Accordingly, in this methods protocol, the use of the ECIS will be described in the context of the wound healing research: cardiac 3T3 fibroblasts will be wounded and their recovery dynamics analyzed based on the typical methodologies applied to the processing of ECIS data. In addition, cellular micromotions will be evaluated. Finally, fluorescence immunostaining of ECIS samples will be described in order to showcase the potential of the ECIS in combination with other well-established techniques to add further knowledge depth to the understanding of the complex wound healing dynamics.
Collapse
Affiliation(s)
- Aina Gallemí-Pérez
- Max Planck Institute for Dynamics and Self-Organisation, Göttingen, Germany.
| | - Marco Tarantola
- Max Planck Institute for Dynamics and Self-Organisation, Göttingen, Germany
| |
Collapse
|
4
|
House A, Cornick J, Butt Q, Guvendiren M. Elastomeric platform with surface wrinkling patterns to control cardiac cell alignment. J Biomed Mater Res A 2023; 111:1228-1242. [PMID: 36762538 DOI: 10.1002/jbm.a.37511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/07/2022] [Accepted: 01/29/2023] [Indexed: 02/11/2023]
Abstract
There is a growing interest in creating 2D cardiac tissue models that display native extracellular matrix (ECM) cues of the heart tissue. Cellular alignment alone is known to be a crucial cue for cardiac tissue development by regulating cell-cell and cell-ECM interactions. In this study, we report a simple and robust approach to create lamellar surface wrinkling patterns enabling spatial control of pattern dimensions with a wide range of pattern amplitude (A ≈ 2-55 μm) and wavelength (λ ≈ 35-100 μm). For human cardiomyocytes (hCMs) and human cardiac fibroblasts (hCFs), our results indicate that the degree of cellular alignment and pattern recognition are correlated with pattern A and λ. We also demonstrate fabrication of devices composed of micro-well arrays with user-defined lamellar patterns on the bottom surface of each well for high-throughput screening studies. Results from a screening study indicate that cellular alignment is strongly diminished with increasing seeding density. In another study, we show our ability to vary hCM/hCF seeding ratio for each well to create co-culture systems where seeding ratio is independent of cellular alignment.
Collapse
Affiliation(s)
- Andrew House
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Jason Cornick
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Quratulain Butt
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| | - Murat Guvendiren
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, New Jersey, USA
| |
Collapse
|
5
|
Liu W, Han JL, Tomek J, Bub G, Entcheva E. Simultaneous Widefield Voltage and Dye-Free Optical Mapping Quantifies Electromechanical Waves in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. ACS PHOTONICS 2023; 10:1070-1083. [PMID: 37096210 PMCID: PMC10119986 DOI: 10.1021/acsphotonics.2c01644] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Indexed: 05/03/2023]
Abstract
Coupled electromechanical waves define a heart's function in health and diseases. Optical mapping of electrical waves using fluorescent labels offers mechanistic insights into cardiac conduction abnormalities. Dye-free/label-free mapping of mechanical waves presents an attractive non-invasive alternative. In this study, we developed a simultaneous widefield voltage and interferometric dye-free optical imaging methodology that was used as follows: (1) to validate dye-free optical mapping for quantification of cardiac wave properties in human iPSC-cardiomyocytes (CMs); (2) to demonstrate low-cost optical mapping of electromechanical waves in hiPSC-CMs using recent near-infrared (NIR) voltage sensors and orders of magnitude cheaper miniature industrial CMOS cameras; (3) to uncover previously underexplored frequency- and space-varying parameters of cardiac electromechanical waves in hiPSC-CMs. We find similarity in the frequency-dependent responses of electrical (NIR fluorescence-imaged) and mechanical (dye-free-imaged) waves, with the latter being more sensitive to faster rates and showing steeper restitution and earlier appearance of wavefront tortuosity. During regular pacing, the dye-free-imaged conduction velocity and electrical wave velocity are correlated; both modalities are sensitive to pharmacological uncoupling and dependent on gap-junctional protein (connexins) determinants of wave propagation. We uncover the strong frequency dependence of the electromechanical delay (EMD) locally and globally in hiPSC-CMs on a rigid substrate. The presented framework and results offer new means to track the functional responses of hiPSC-CMs inexpensively and non-invasively for counteracting heart disease and aiding cardiotoxicity testing and drug development.
Collapse
Affiliation(s)
- Wei Liu
- Department
of Biomedical Engineering, George Washington
University, Washington, D.C. 20052, United States
| | - Julie L. Han
- Department
of Biomedical Engineering, George Washington
University, Washington, D.C. 20052, United States
| | - Jakub Tomek
- Department
of Pharmacology, University of California−Davis, Davis, California 95616, United States
| | - Gil Bub
- Department
of Physiology, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Emilia Entcheva
- Department
of Biomedical Engineering, George Washington
University, Washington, D.C. 20052, United States
| |
Collapse
|
6
|
Chen Y, Chan JPY, Wu J, Li R, Santerre JP. Compatibility and function of human induced pluripotent stem cell derived cardiomyocytes on an electrospun nanofibrous scaffold, generated from an ionomeric polyurethane composite. J Biomed Mater Res A 2022; 110:1932-1943. [PMID: 35851742 DOI: 10.1002/jbm.a.37428] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/27/2022] [Accepted: 07/01/2022] [Indexed: 11/06/2022]
Abstract
Synthetic scaffolds are needed for generating organized neo-myocardium constructs to promote functional tissue repair. This study investigated the biocompatibility of an elastomeric electrospun degradable polar/hydrophobic/ionic polyurethane (D-PHI) composite scaffold with human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). The composite material was electrospun to generate scaffolds, with nanofibres oriented in aligned or random directions. These features enabled the authors to evaluate the effect of characteristic elements which mimic that of the native extracellular matrix (alignment, chemical heterogeneity, and fiber topography) on hiPSC-CMs activity. The functional nature of the hiPSC-CM cultured on gelatin and Matrigel-coated scaffolds were assessed, investigating the influence of protein interactions with the synthetic substrate on subsequent cell phenotype. After 7 days of culture, high hiPSC-CM viability was observed on the scaffolds. The cells on the aligned scaffold were elongated and demonstrated aligned sarcomeres that oriented parallel to the direction of the fibers, while the cells on random scaffolds and a tissue culture polystyrene (TCPS) control did not exhibit such an organized morphology. The hiPSC-CMs cultured on the scaffolds and TCPS expressed similar levels of cardiac troponin-T, but there was a higher expression of ventricular myosin light chain-2 on the D-PHI composite scaffolds versus TCPS, indicating a higher proportion of hiPSC-CM exhibiting a ventricular cardiomyocyte like phenotype. Within 7 days, the hiPSC-CMs on aligned scaffolds and TCPS beat synchronously and had similar conductive velocities. These preliminary results show that aligned D-PHI elastomeric scaffolds allow hiPSC-CMs to demonstrate important cardiomyocytes characteristics, critical to enabling their future potential use for cardiac tissue regeneration.
Collapse
Affiliation(s)
- Yizhou Chen
- Institute of Biomedical Engineering, University of Toronto Toronto Ontario Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research University of Toronto Toronto Ontario Canada
| | - Jennifer P. Y. Chan
- Institute of Biomedical Engineering, University of Toronto Toronto Ontario Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research University of Toronto Toronto Ontario Canada
- Baylis Medical Mississauga Ontario Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network Toronto Ontario Canada
| | - Ren‐Ke Li
- Toronto General Hospital Research Institute, University Health Network Toronto Ontario Canada
| | - J. Paul Santerre
- Institute of Biomedical Engineering, University of Toronto Toronto Ontario Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research University of Toronto Toronto Ontario Canada
- Toronto General Hospital Research Institute, University Health Network Toronto Ontario Canada
- Faculty of Dentistry University of Toronto Toronto Ontario Canada
| |
Collapse
|
7
|
Boehm M, Tian X, Ali MK, Mao Y, Ichimura K, Zhao M, Kuramoto K, Dannewitz Prosseda S, Fajardo G, Dufva MJ, Qin X, Kheyfets VO, Bernstein D, Reddy S, Metzger RJ, Zamanian RT, Haddad F, Spiekerkoetter E. Improving Right Ventricular Function by Increasing BMP Signaling with FK506. Am J Respir Cell Mol Biol 2021; 65:272-287. [PMID: 33938785 PMCID: PMC8485990 DOI: 10.1165/rcmb.2020-0528oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/20/2021] [Indexed: 11/24/2022] Open
Abstract
Right ventricular (RV) function is the predominant determinant of survival in patients with pulmonary arterial hypertension (PAH). In preclinical models, pharmacological activation of BMP (bone morphogenetic protein) signaling with FK506 (tacrolimus) improved RV function by decreasing RV afterload. FK506 therapy further stabilized three patients with end-stage PAH. Whether FK506 has direct effects on the pressure-overloaded right ventricle is yet unknown. We hypothesized that increasing cardiac BMP signaling with FK506 improves RV structure and function in a model of fixed RV afterload after pulmonary artery banding (PAB). Direct cardiac effects of FK506 on the microvasculature and RV fibrosis were studied after surgical PAB in wild-type and heterozygous Bmpr2 mutant mice. RV function and strain were assessed longitudinally via cardiac magnetic resonance imaging during continuous FK506 infusion. Genetic lineage tracing of endothelial cells (ECs) was performed to assess the contribution of ECs to fibrosis. Molecular mechanistic studies were performed in human cardiac fibroblasts and ECs. In mice, low BMP signaling in the right ventricle exaggerated PAB-induced RV fibrosis. FK506 therapy restored cardiac BMP signaling, reduced RV fibrosis in a BMP-dependent manner independent from its immunosuppressive effect, preserved RV capillarization, and improved RV function and strain over the time course of disease. Endothelial mesenchymal transition was a rare event and did not significantly contribute to cardiac fibrosis after PAB. Mechanistically, FK506 required ALK1 in human cardiac fibroblasts as a BMPR2 co-receptor to reduce TGFβ1-induced proliferation and collagen production. Our study demonstrates that increasing cardiac BMP signaling with FK506 improves RV structure and function independent from its previously described beneficial effects on pulmonary vascular remodeling.
Collapse
Affiliation(s)
- Mario Boehm
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardio-Pulmonary Institute, Justus-Liebig-University Giessen, German Center for Lung Research (DZL), Giessen, Germany
| | - Xuefei Tian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Md Khadem Ali
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Yuqiang Mao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Kenzo Ichimura
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Mingming Zhao
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Kazuya Kuramoto
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Svenja Dannewitz Prosseda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
| | - Giovanni Fajardo
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Melanie J. Dufva
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado; and
- Department of Pediatrics, Section of Cardiology, Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Xulei Qin
- Cardiovascular Institute, and
- Department of Cardiovascular Medicine, Stanford University, Stanford, California
| | - Vitaly O. Kheyfets
- Department of Bioengineering, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado; and
- Department of Pediatrics, Section of Cardiology, Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Daniel Bernstein
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Sushma Reddy
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Ross J. Metzger
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Division of Cardiology, Department of Pediatrics
- Cardiovascular Institute, and
| | - Roham T. Zamanian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardiovascular Institute, and
| | - Francois Haddad
- Cardiovascular Institute, and
- Department of Cardiovascular Medicine, Stanford University, Stanford, California
| | - Edda Spiekerkoetter
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
- Vera Moulton Wall Center for Pulmonary Vascular Disease
- Cardiovascular Institute, and
| |
Collapse
|
8
|
Schussler O, Chachques JC, Alifano M, Lecarpentier Y. Key Roles of RGD-Recognizing Integrins During Cardiac Development, on Cardiac Cells, and After Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:179-203. [PMID: 34342855 DOI: 10.1007/s12265-021-10154-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Cardiac cells interact with the extracellular matrix (ECM) proteins through integrin mechanoreceptors that control many cellular events such as cell survival, apoptosis, differentiation, migration, and proliferation. Integrins play a crucial role in cardiac development as well as in cardiac fibrosis and hypertrophy. Integrins recognize oligopeptides present on ECM proteins and are involved in three main types of interaction, namely with collagen, laminin, and the oligopeptide RGD (Arg-Gly-Asp) present on vitronectin and fibronectin proteins. To date, the specific role of integrins recognizing the RGD has not been addressed. In this review, we examine their role during cardiac development, their role on cardiac cells, and their upregulation during pathological processes such as heart fibrosis and hypertrophy. We also examine their role in regenerative and angiogenic processes after myocardial infarction (MI) in the peri-infarct area. Specific targeting of these integrins may be a way of controlling some of these pathological events and thereby improving medical outcomes.
Collapse
Affiliation(s)
- Olivier Schussler
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.
| | - Juan C Chachques
- Department of Cardiac Surgery Pompidou Hospital, Laboratory of Biosurgical Research, Carpentier Foundation, University Paris Descartes, 75015, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.,INSERM U1138 Team "Cancer, Immune Control, and Escape", Cordeliers Research Center, University of Paris, Paris, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| |
Collapse
|
9
|
Iwakura T, Marschner JA, Zhao ZB, Świderska MK, Anders HJ. Electric cell-substrate impedance sensing in kidney research. Nephrol Dial Transplant 2021; 36:216-223. [PMID: 31598727 DOI: 10.1093/ndt/gfz191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/18/2019] [Indexed: 12/20/2022] Open
Abstract
Electric cell-substrate impedance sensing (ECIS) is a quantitative, label-free, non-invasive analytical method allowing continuous monitoring of the behaviour of adherent cells by online recording of transcellular impedance. ECIS offers a wide range of practical applications to study cell proliferation, migration, differentiation, toxicity and monolayer barrier integrity. All of these applications are relevant for basic kidney research, e.g. on endothelial cells, tubular and glomerular epithelial cells. This review gives an overview on the fundamental principles of the ECIS technology. We name strengths and remaining hurdles for practical applications, present an ECIS array reuse protocol, and review its past, present and potential future contributions to preclinical kidney research.
Collapse
Affiliation(s)
- Takamasa Iwakura
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany.,Internal Medicine I, Division of Nephrology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Julian A Marschner
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Zhi Bo Zhao
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Monika Katarzyna Świderska
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| | - Hans-Joachim Anders
- Renal Division, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, LMU Munich, München, Germany
| |
Collapse
|
10
|
Development of a drug screening system using three-dimensional cardiac tissues containing multiple cell types. Sci Rep 2021; 11:5654. [PMID: 33707655 PMCID: PMC7952584 DOI: 10.1038/s41598-021-85261-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 02/28/2021] [Indexed: 01/22/2023] Open
Abstract
We hypothesized that an appropriate ratio of cardiomyocytes, fibroblasts, endothelial cells, and extracellular matrix (ECM) factors would be required for the development of three-dimensional cardiac tissues (3D-CTs) as drug screening systems. To verify this hypothesis, ECM-coated human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), ECM-coated cardiac fibroblasts (CFs), and uncoated cardiac endothelial cells (CEs) were mixed in the following ratios: 10:0:0 (10CT), 7:2:1 (7CT), 5:4:1 (5CT), and 2:7:1 (2CT). The expression of cardiac-, fibroblasts-, and endothelial-specific markers was assessed by FACS, qPCR, and immunostaining while that of ECM-, cell adhesion-, and ion channel-related genes was examined by qPCR. Finally, the contractile properties of the tissues were evaluated in the absence or presence of E-4031 and isoproterenol. The expression of ECM- and adhesion-related genes significantly increased, while that of ion channel-related genes significantly decreased with the CF proportion. Notably, 7CT showed the greatest contractility of all 3D-CTs. When exposed to E-4031 (hERG K channel blocker), 7CT and 5CT showed significantly decreased contractility and increased QT prolongation. Moreover, 10CT and 7CT exhibited a stronger response to isoproterenol than did the other 3D-CTs. Finally, 7CT showed the highest drug sensitivity among all 3D-CTs. In conclusion, 3D-CTs with an appropriate amount of fibroblasts/endothelial cells (7CT in this study) are suitable drug screening systems, e.g. for the detection of drug-induced arrhythmia.
Collapse
|
11
|
Yu J, Cai P, Chen X. Structural Regulation of Myocytes in Engineered Healthy and Diseased Cardiac Models. ACS APPLIED BIO MATERIALS 2021; 4:267-276. [DOI: 10.1021/acsabm.0c01270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jing Yu
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Pingqiang Cai
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| | - Xiaodong Chen
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Lab for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, 639798 Singapore
| |
Collapse
|
12
|
Sridhar KC, Hersch N, Dreissen G, Merkel R, Hoffmann B. Calcium mediated functional interplay between myocardial cells upon laser-induced single-cell injury: an in vitro study of cardiac cell death signaling mechanisms. Cell Commun Signal 2020; 18:191. [PMID: 33371897 PMCID: PMC7771078 DOI: 10.1186/s12964-020-00689-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 11/23/2020] [Indexed: 01/09/2023] Open
Abstract
Background The electromechanical function of myocardial tissue depends on the intercellular communication between cardiomyocytes (CMs) as well as their crosstalk with other cell types. Cell injury, and subsequent death trigger inflammation as in myocardial infarction (MI) resulting in myocardial remodeling. Although mechanisms underlying myocardial cell death have been studied so far, the signaling events following single cell death and spontaneous response of connected cells in the myocardial tissue is still barely understood. Methods Here, we investigated the effect of laser-induced single cell death on Calcium (Ca2+) concentrations and transport in myocardial cell clusters in vitro. Spatial and temporal changes in intracellular Ca2+ concentrations [Ca2+]i were studied using a fluorescent calcium indicator, Fluo-4AM. Spontaneous signaling events following cell death were studied in rat embryonic cardiomyocytes and non-myocytes using separate cell culture systems. Results Cell death triggered spontaneous increase in intracellular Ca2+ levels ([Ca2+]i) of surrounding cells. The spread of the observed propagating Ca2+ signal was slow and sustained in myocytes while it was rapid and transient in fibroblasts (Fbs). Further, sustained high Ca2+ levels temporarily impaired the contractility in CMs. The cell-type specific effect of ablation was confirmed using separate cultures of CMs and Fbs. Comparing Ca2+ propagation speed in myocytes and fibroblasts, we argue for a diffusion-driven Ca2+ propagation in myocytes, but not in fibroblasts. Radial and sequential Ca2+ diffusion across the CMs through cell–cell contacts and presence of Cx43-based intercellular junctions indicated a gap junction flow of Ca2+. Conclusions These findings illustrate the spontaneous Ca2+-mediated functional interplay in myocardial cell clusters upon mechanical injury and, further, the difference in Ca2+ signaling in cardiomyocytes and fibroblasts. Video Abstract
Collapse
Affiliation(s)
- Krishna Chander Sridhar
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Nils Hersch
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Georg Dreissen
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Rudolf Merkel
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Forschungszentrum Jülich, 52425, Jülich, Germany
| | - Bernd Hoffmann
- Institute of Biological Information Processing, IBI-2: Mechanobiology, Forschungszentrum Jülich, 52425, Jülich, Germany.
| |
Collapse
|
13
|
Pretorius D, Kahn-Krell AM, LaBarge WC, Lou X, Kannappan R, Pollard AE, Fast VG, Berry JL, Eberhardt AW, Zhang J. Fabrication and characterization of a thick, viable bi-layered stem cell-derived surrogate for future myocardial tissue regeneration. Biomed Mater 2020; 16. [PMID: 33053512 DOI: 10.1088/1748-605x/abc107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Cardiac tissue surrogates show promise for restoring mechanical and electrical function in infarcted left ventricular (LV) myocardium. For these cardiac surrogates to be useful in vivo, they are required to support synchronous and forceful contraction over the infarcted region. These design requirements necessitate a thickness sufficient to produce a useful contractile force, an area large enough to cover an infarcted region, and prevascularization to overcome diffusion limitations. Attempts to meet these requirements have been hampered by diffusion limits of oxygen and nutrients (100-200 μm) leading to necrotic regions.This study demonstrates a novel layer-by-layer (LbL) fabrication method used to produce tissue surrogates that meet these requirements and mimic normal myocardium in form and function. Thick (1.5-2 mm) LbL cardiac tissues created from human induced pluripotent stem cell-derived cardiomyocytes and endothelial cells were assessed, in vitro, over a four week period for viability (< 5.6 ± 1.4 % nectrotic cells), cell morphology, viscoelastic properties and functionality. Viscoelastic properties of the cardiac surrogates were determined via stress relaxation response modeling and compared to native murine LV tissue. Viscoelastic characterization showed that the generalized Maxwell model of order 4 described the samples well (0.7 < R2 < 0.98). Functional performance assessment showed enhanced t-tubule network development, gap junction communication as well as conduction velocity (16.9 ± 2.3 cm s-1). These results demonstrate that LbL fabrication can be utilized successfully in creating complex, functional cardiac surrogates for therapeutic applications.
Collapse
Affiliation(s)
- Danielle Pretorius
- Biomedical Engineering, The University of Alabama at Birmingham, Volker Hall Room G094, 1670 University Blvd, Birmingham, Alabama, 35294-2182, UNITED STATES
| | - Asher M Kahn-Krell
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Wesley C LaBarge
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Xi Lou
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Ramaswamy Kannappan
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Andrew E Pollard
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Vladimir G Fast
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Joel L Berry
- School of Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Alan W Eberhardt
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| | - Jianyi Zhang
- Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, Alabama, UNITED STATES
| |
Collapse
|
14
|
Kim H, Witt H, Oswald TA, Tarantola M. Adhesion of Epithelial Cells to PNIPAm Treated Surfaces for Temperature-Controlled Cell-Sheet Harvesting. ACS APPLIED MATERIALS & INTERFACES 2020; 12:33516-33529. [PMID: 32631046 PMCID: PMC7467562 DOI: 10.1021/acsami.0c09166] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Stimuli responsive polymer coatings are a common motive for designing surfaces for cell biological applications. In the present study, we have characterized temperature dependent adhesive properties of poly(N-isopropylacrylamide) (PNIPAm) microgel coated surfaces (PMS) using various atomic force microscopy based approaches. We imaged and quantified the material properties of PMS upon a temperature switch using quantitative AFM imaging but also employed single-cell force spectroscopy (SCFS) before and after decreasing the temperature to assess the forces and work of initial adhesion between cells and PMS. We performed a detailed analysis of steps in the force-distance curves. Finally, we applied colloid probe atomic force microscopy (CP-AFM) to analyze the adhesive properties of two major components of the extracellular matrix to PMS under temperature control, namely collagen I and fibronectin. In combination with confocal imaging, we could show that these two ECM components differ in their detachment properties from PNIPAm microgel films upon cell harvesting, and thus gained a deeper understanding of cell-sheet maturation and harvesting process and the involved partial ECM dissolution.
Collapse
Affiliation(s)
- Hyejeong Kim
- Max Planck Institute
for Dynamics and Self Organization (MPIDS), Am Fassberg 17, 37077 Göttingen, Germany
| | - Hannes Witt
- Max Planck Institute
for Dynamics and Self Organization (MPIDS), Am Fassberg 17, 37077 Göttingen, Germany
| | - Tabea A. Oswald
- Institute of Organic and Biomolecular Chemistry, University of Göttingen, Tammannstrasse 2, 37077 Göttingen, Germany
| | - Marco Tarantola
- Max Planck Institute
for Dynamics and Self Organization (MPIDS), Am Fassberg 17, 37077 Göttingen, Germany
- Institute for Dynamics of Complex Systems, University of Göttingen, Friedrich-Hund Platz 1, 37073 Göttingen, Germany
- E-mail: . Phone: +49-551-5176-316
| |
Collapse
|
15
|
Arai S, Lloyd K, Takahashi T, Mammoto K, Miyazawa T, Tamura K, Kaneko T, Ishida K, Moriyama Y, Mitsui T. Dynamic Properties of Heart Fragments from Different Regions and Their Synchronization. Bioengineering (Basel) 2020; 7:E81. [PMID: 32751255 PMCID: PMC7552607 DOI: 10.3390/bioengineering7030081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/17/2020] [Accepted: 07/23/2020] [Indexed: 12/29/2022] Open
Abstract
The dynamic properties of the heart differ based on the regions that effectively circulate blood throughout the body with each heartbeat. These properties, including the inter-beat interval (IBI) of autonomous beat activity, are retained even in in vitro tissue fragments. However, details of beat dynamics have not been well analyzed, particularly at the sub-mm scale, although such dynamics of size are important for regenerative medicine and computational studies of the heart. We analyzed the beat dynamics in sub-mm tissue fragments from atria and ventricles of hearts obtained from chick embryos over a period of 40 h. The IBI and contraction speed differed by region and atrial fragments retained their values for a longer time. The major finding of this study is synchronization of these fragment pairs physically attached to each other. The probability of achieving this and the time required differ for regional pairs: atrium-atrium, ventricle-ventricle, or atrium-ventricle. Furthermore, the time required to achieve 1:1 synchronization does not depend on the proximity of initial IBI of paired fragments. Various interesting phenomena, such as 1:n synchronization and a reentrant-like beat sequence, are revealed during synchronization. Finally, our observation of fragment dynamics indicates that mechanical motion itself contributes to the synchronization of atria.
Collapse
Affiliation(s)
- Shin Arai
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Kento Lloyd
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Tomonori Takahashi
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Kazuki Mammoto
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Takashi Miyazawa
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Kei Tamura
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Tomoyuki Kaneko
- Department of Frontier Bioscience, Hosei University, Koganei, Tokyo 184-8584, Japan;
| | - Kentaro Ishida
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Yuuta Moriyama
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| | - Toshiyuki Mitsui
- Department of Physics and Mathematics, College of Science and Engineering, Aoyama Gakuin University, Kanagawa 252-5258, Japan; (S.A.); (K.L.); (T.T.); (K.M.); (T.M.); (K.T.); (K.I.); (Y.M.)
| |
Collapse
|
16
|
Quagliariello V, Vecchione R, De Capua A, Lagreca E, Iaffaioli RV, Botti G, Netti PA, Maurea N. Nano-Encapsulation of Coenzyme Q10 in Secondary and Tertiary Nano-Emulsions for Enhanced Cardioprotection and Hepatoprotection in Human Cardiomyocytes and Hepatocytes During Exposure to Anthracyclines and Trastuzumab. Int J Nanomedicine 2020; 15:4859-4876. [PMID: 32764923 PMCID: PMC7359894 DOI: 10.2147/ijn.s245170] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/18/2020] [Indexed: 12/19/2022] Open
Abstract
Introduction CoenzymeQ10 (CoQ10) is a well-known antioxidant and anti-inflammatory agent with cardioprotective properties. However, clinical trials based on its oral administration have failed to provide significant effect on cardiac functionality. The main limitation of CoQ10 is based on its very low oral bioavailability and instability that limit dramatically its effects as a cardioprotective agent. Herein, we loaded CoQ10 in high bioavailable nano-emulsions (NEs) coated with chitosan or chitosan and hyaluronic acid in order to improve its performance. Methods We tested cardioprotective and hepatoprotective effects of CoQ10-loaded nano-carriers against Doxorubicin and Trastuzumab toxicities in cardiomyocytes and liver cells through analysis of cell viability, lipid peroxidation, expression of leukotrienes, p65/NF-kB and pro-inflammatory cytokines involved in anticancer-induced cardio and hepatotoxicity. Results Nano-carriers showed high stability and loading ability and increased cell viability both in hepatocytes and cardiomyocytes during anticancer treatments. We observed that these effects are mediated by the inhibition of lipid peroxidation and reduction of the inflammation. CoQ10-loaded nano-emulsions showed also strong anti-inflammatory effects reducing leukotriene B4 and p65/NF-κB expression and Interleukin 1β and 6 production during anticancer treatments. Discussion Anthracyclines and Human epidermal growth factor receptor (HER2) inhibitors have shown significant anticancer effects in clinical practice but their use is characterized by cardiotoxicity and hepatotoxicity. Nano-carriers loaded with CoQ10 showed cardio and hepatoprotective properties mediated by reduction of oxidative damages and pro-inflammatory mediators. These results set the stage for preclinical studies of cardio and hepatoprotection in HER2+ breast cancer-bearing mice treated with Doxorubicin and Trastuzumab.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, Napoli, Italia
| | - Raffaele Vecchione
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | - Alberta De Capua
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | - Elena Lagreca
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | | | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, Napoli, Italia
| | - Paolo A Netti
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, Napoli, Italia
| |
Collapse
|
17
|
Pitoulis FG, Watson SA, Perbellini F, Terracciano CM. Myocardial slices come to age: an intermediate complexity in vitro cardiac model for translational research. Cardiovasc Res 2020; 116:1275-1287. [PMID: 31868875 PMCID: PMC7243278 DOI: 10.1093/cvr/cvz341] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/31/2019] [Accepted: 12/19/2019] [Indexed: 12/17/2022] Open
Abstract
Although past decades have witnessed significant reductions in mortality of heart failure together with advances in our understanding of its cellular, molecular, and whole-heart features, a lot of basic cardiac research still fails to translate into clinical practice. In this review we examine myocardial slices, a novel model in the translational arena. Myocardial slices are living ultra-thin sections of heart tissue. Slices maintain the myocardium's native function (contractility, electrophysiology) and structure (multicellularity, extracellular matrix) and can be prepared from animal and human tissue. The discussion begins with the history and current advances in the model, the different interlaboratory methods of preparation and their potential impact on results. We then contextualize slices' advantages and limitations by comparing it with other cardiac models. Recently, sophisticated methods have enabled slices to be cultured chronically in vitro while preserving the functional and structural phenotype. This is more timely now than ever where chronic physiologically relevant in vitro platforms for assessment of therapeutic strategies are urgently needed. We interrogate the technological developments that have permitted this, their limitations, and future directions. Finally, we look into the general obstacles faced by the translational field, and how implementation of research systems utilizing slices could help in resolving these.
Collapse
Affiliation(s)
- Fotios G Pitoulis
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Samuel A Watson
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Filippo Perbellini
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies, Hannover, Germany
| | - Cesare M Terracciano
- Laboratory of Cell Electrophysiology, Department of Myocardial Function, Imperial College London, National Heart and Lung Institute, 4th Floor ICTEM Building Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
18
|
Slotvitsky MM, Tsvelaya VA, Podgurskaya AD, Agladze KI. Formation of an electrical coupling between differentiating cardiomyocytes. Sci Rep 2020; 10:7774. [PMID: 32385315 PMCID: PMC7210299 DOI: 10.1038/s41598-020-64581-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/08/2020] [Indexed: 01/02/2023] Open
Abstract
Human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) serve as an indispensable platform for the study of human cardiovascular disease is human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs). While the possibility of reproducing rare pathologies, patient-specific selection of drugs, and other issues concerning single cardiomyocytes have been well studied, little attention has been paid to the properties of the whole syncytium of CMs, in which both the functionality of individual cells and the distribution of electrophysiological connections between them are essential. The aim of this work is to directly study the ability of hiPSC-CMs to form a functional syncytium that can stably conduct an excitation wave. For that purpose, syncytium forming hiPSC-CMs were harvested and seeded (transferred) on a new substrate on different days of differentiation. The excitation conduction in a sample was characterized by the stability of the wavefront using optical mapping data. We found that the cells transferred before the 20th day of differentiation were able to organize a functional syncytium capable of further development and stable excitation conduction at high stimulation frequencies, while the cells transferred after 20 days did not form a homogeneous syncytium, and multiple instabilities of the propagating wavefront were observed with the possibility of reentry formation.
Collapse
Affiliation(s)
- M M Slotvitsky
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation
| | - V A Tsvelaya
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation
| | - A D Podgurskaya
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation
| | - K I Agladze
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141700, Russian Federation. .,M.F. Vladimirsky Moscow Regional Clinical Research Institute, Moscow, 129110, Russian Federation.
| |
Collapse
|
19
|
Turaga D, Matthys OB, Hookway TA, Joy DA, Calvert M, McDevitt TC. Single-Cell Determination of Cardiac Microtissue Structure and Function Using Light Sheet Microscopy. Tissue Eng Part C Methods 2020; 26:207-215. [PMID: 32111148 DOI: 10.1089/ten.tec.2020.0020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Native cardiac tissue is composed of heterogeneous cell populations that work cooperatively for proper tissue function; thus, engineered tissue models have moved toward incorporating multiple cardiac cell types in an effort to recapitulate native multicellular composition and organization. Cardiac tissue models composed of stem cell-derived cardiomyocytes (CMs) require inclusion of non-myocytes to promote stable tissue formation, yet the specific contributions of the supporting non-myocyte population on the parenchymal CMs and cardiac microtissues have to be fully dissected. This gap can be partly attributed to limitations in technologies able to accurately study the individual cellular structure and function that comprise intact three-dimensional (3D) tissues. The ability to interrogate the cell-cell interactions in 3D tissue constructs has been restricted by conventional optical imaging techniques that fail to adequately penetrate multicellular microtissues with sufficient spatial resolution. Light sheet fluorescence microscopy (LSFM) overcomes these constraints to enable single-cell resolution structural and functional imaging of intact cardiac microtissues. Multicellular spatial distribution analysis of heterotypic cardiac cell populations revealed that CMs and cardiac fibroblasts were randomly distributed throughout 3D microtissues. Furthermore, calcium imaging of live cardiac microtissues enabled single-cell detection of CM calcium activity, which showed that functional heterogeneity correlated with spatial location within the tissues. This study demonstrates that LSFM can be utilized to determine single-cell spatial and functional interactions of multiple cell types within intact 3D engineered microtissues, thereby facilitating the determination of structure-function relationships at both tissue-level and single-cell resolution. Impact statement The ability to achieve single-cell resolution by advanced three-dimensional light imaging techniques enables exquisite new investigation of multicellular analyses in native and engineered tissues. In this study, light sheet fluorescence microscopy was used to define structure-function relationships of distinct cell types in engineered cardiac microtissues by determining heterotypic cell distributions and interactions throughout the tissues as well as by assessing regional differences in calcium handing functional properties at the individual cardiomyocyte level.
Collapse
Affiliation(s)
| | - Oriane B Matthys
- Gladstone Institutes, San Francisco, California
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California
| | | | - David A Joy
- Gladstone Institutes, San Francisco, California
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California
| | | | - Todd C McDevitt
- Gladstone Institutes, San Francisco, California
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| |
Collapse
|
20
|
Mehrotra S, de Melo BAG, Hirano M, Keung W, Li RA, Mandal BB, Shin SR. Nonmulberry Silk Based Ink for Fabricating Mechanically Robust Cardiac Patches and Endothelialized Myocardium-on-a-Chip Application. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1907436. [PMID: 33071707 PMCID: PMC7566555 DOI: 10.1002/adfm.201907436] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Indexed: 05/20/2023]
Abstract
Bioprinting holds great promise towards engineering functional cardiac tissue constructs for regenerative medicine and as drug test models. However, it is highly limited by the choice of inks that require maintaining a balance between the structure and functional properties associated with the cardiac tissue. In this regard, we have developed a novel and mechanically robust biomaterial-ink based on non-mulberry silk fibroin protein. The silk-based ink demonstrated suitable mechanical properties required in terms of elasticity and stiffness (~40 kPa) for developing clinically relevant cardiac tissue constructs. The ink allowed the fabrication of stable anisotropic scaffolds using a dual crosslinking method, which were able to support formation of aligned sarcomeres, high expression of gap junction proteins as connexin-43, and maintain synchronously beating of cardiomyocytes. The printed constructs were found to be non-immunogenic in vitro and in vivo. Furthermore, delving into an innovative method for fabricating a vascularized myocardial tissue-on-a-chip, the silk-based ink was used as supporting hydrogel for encapsulating human induced pluripotent stem cell derived cardiac spheroids (hiPSC-CSs) and creating perfusable vascularized channels via an embedded bioprinting technique. We confirmed the ability of silk-based supporting hydrogel towards maturation and viability of hiPSC-CSs and endothelial cells, and for applications in evaluating drug toxicity.
Collapse
Affiliation(s)
- Shreya Mehrotra
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Bruna A. G. de Melo
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA
- Department of Engineering of Materials and Bioprocesses, School of Chemical Engineering, University of Campinas, Campinas, SP 13083-852, Brazil
| | - Minoru Hirano
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA
- Future Vehicle Research Department, Toyota Research Institute North America, Toyota Motor North America Inc., 1555 Woodridge Ave Ann Arbor, MI 48105, USA
| | - Wendy Keung
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong
| | - Ronald A. Li
- Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Hong Kong
| | - Biman B. Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
- Center for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA 02139, USA
| |
Collapse
|
21
|
Orozco P, Montoya Y, Bustamante J. Development of endomyocardial fibrosis model using a cell patterning technique: In vitro interaction of cell coculture of 3T3 fibroblasts and RL-14 cardiomyocytes. PLoS One 2020; 15:e0229158. [PMID: 32092082 PMCID: PMC7039516 DOI: 10.1371/journal.pone.0229158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/30/2020] [Indexed: 01/07/2023] Open
Abstract
Cardiac functions can be altered by changes in the microstructure of the heart, i.e., remodeling of the cardiac tissue, which may activate pathologies such as hypertrophy, dilation, or cardiac fibrosis. Cardiac fibrosis can develop due to an excessive deposition of extracellular matrix proteins, which are products of the activation of fibroblasts. In this context, the anatomical-histological change may interfere with the functioning of the cardiac tissue, which requires specialized cells for its operation. The purpose of the present study was to determine the cellular interactions and morphological changes in cocultures of 3T3 fibroblasts and RL-14 cardiomyocytes via the generation of a platform an in vitro model. For this purpose, a platform emulating the biological characteristics of endomyocardial fibrosis was generated using a cell patterning technique to study morphological cellular changes in compact and irregular patterns of fibrosis. It was found that cellular patterns emulating the geometrical distributions of endomyocardial fibrosis generated morphological changes after interaction of the RL-14 cardiomyocytes with the 3T3 fibroblasts. Through this study, it was possible to evaluate biological characteristics such as cell proliferation, adhesion, and spatial distribution, which are directly related to the type of emulated endomyocardial fibrosis. This research concluded that fibroblasts inhibited the proliferation of cardiomyocytes via their interaction with specific microarchitectures. This behavior is consistent with the histopathological distribution of cardiac fibrosis; therefore, the platform developed in this research could be useful for the in vitro assessment of cellular microdomains. This would allow for the experimental determination of interactions with drugs, substrates, or biomaterials within the engineering of cardiac tissues.
Collapse
Affiliation(s)
- Paola Orozco
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
| | - Yuliet Montoya
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
- Comité de Trabajo de Bioingeniería Cardiovascular, Sociedad Colombiana de Cardiología y Cirugía Cardiovascular, Bogotá, Colombia
| | - John Bustamante
- Centro de Bioingeniería, Grupo de Dinámica Cardiovascular, Universidad Pontificia Bolivariana, Medellín, Colombia
- Comité de Trabajo de Bioingeniería Cardiovascular, Sociedad Colombiana de Cardiología y Cirugía Cardiovascular, Bogotá, Colombia
| |
Collapse
|
22
|
Jang Y, Choi SC, Lim DS, Kim JH, Kim J, Park Y. Modulating cardiomyocyte and fibroblast interaction using layer-by-layer deposition facilitates synchronisation of cardiac macro tissues. SOFT MATTER 2020; 16:428-434. [PMID: 31799582 DOI: 10.1039/c9sm01531k] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Maturation and synchronisation of heart cells, including cardiomyocytes and fibroblasts, are essential to develop functional biomimetic cardiac tissues for regenerative medicine and drug discovery. Synchronisation of cells in the biomimetic cardiac tissue requires the structural integrity and functional maturation of cardiomyocytes with other cell types. However, it is challenging to synchronise the beating of macroscale cardiac tissues and induce maturation of cardiomyocytes derived from stem cells. Here, we developed a simple assembly technology to modulate cell-cell interactions by combining layer-by-layer (LBL) deposition and centrifugation of cells with collagen type I to control cell-cell interactions for the preparation of cardiac macro tissues (CMTs). We found that maturation of cardiomyocytes in CMTs was largely enhanced by growth factors FGF-4 and ascorbic acid, but synchronisation of cardiac beating required LBL deposition of cardiomyocytes and cardiac fibroblasts in addition to the growth factors during the maturation process. Our findings have important implications because incorporation of cardiac fibroblasts into the cardiomyocyte layer is a prerequisite for synchronised beating of macroscale cardiac tissues in addition to growth factors to facilitate maturation of stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
- Yongjun Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Korea.
| | | | | | | | | | | |
Collapse
|
23
|
Jang Y, Jung DJ, Choi SC, Lim DS, Kim JH, Jeoung GS, Kim J, Park Y. Multidimensional assembly using layer-by-layer deposition for synchronized cardiac macro tissues. RSC Adv 2020; 10:18806-18815. [PMID: 35693693 PMCID: PMC9122566 DOI: 10.1039/d0ra01577f] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/04/2020] [Accepted: 04/29/2020] [Indexed: 12/13/2022] Open
Abstract
The fabrication of biomimetic structures for tissues and organs is emerging in the fields of biomedical engineering and precision medicine. While current progress in biomedical research provides a number of biofabrication methods, the construction of multi-dimensional cardiac tissue is highly challenging due to difficulties in the maturation and synchronization of cardiomyocytes (CMs) in conjunction with other types of cells, such as myofibroblasts and endothelial cells. Here, we show a simple fabrication methodology to construct multi-dimensional cardiac macro tissue (mCMT) by layer-by-layer (LBL) deposition of cells on micro patterned PDMS. mCMTs formed by LBL deposition of pluripotent stem cell (PSC)-derived cardiomyocytes and cardiac fibroblasts formed 3D patterned structures with synchronized beating characteristics. We also demonstrate that cardiac maturation factors such as the gene expression of MLC2v and cTNI and formation of sarcomeres in mCMTs were significantly enhanced by LBL deposition and growth factors during the maturation process. Fabrication of matured mCMTs with synchronized beating enables providing an efficient platform for evaluating the efficacy and toxicity of drug candidates. These results have important implications because mCMTs are applicable to diverse in vitro studies and drug screening methods that require tissue-like structures and functions in a physiological environment. We fabricated a cardiac macro tissue with synchronized beating by layer-by-layer deposition and evaluated the effect of drug candidates.![]()
Collapse
Affiliation(s)
- Yongjun Jang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Korea
| | - Da Jung Jung
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Seung-Cheol Choi
- Departments of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Korea
| | - Do-Sun Lim
- Departments of Cardiology, Cardiovascular Center, Korea University Anam Hospital, Seoul, Korea
| | - Jong-Hoon Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | - Gi Seok Jeoung
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Korea
| | - Jongseong Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
24
|
Hookway TA, Matthys OB, Mendoza-Camacho FN, Rains S, Sepulveda JE, Joy DA, McDevitt TC. Phenotypic Variation Between Stromal Cells Differentially Impacts Engineered Cardiac Tissue Function. Tissue Eng Part A 2019; 25:773-785. [PMID: 30968748 DOI: 10.1089/ten.tea.2018.0362] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
IMPACT STATEMENT Understanding the relationship between parenchymal and supporting cell populations is paramount to recapitulate the multicellular complexity of native tissues. Incorporation of stromal cells is widely recognized to be necessary for the stable formation of stem cell-derived cardiac tissues; yet, the types of stromal cells used have varied widely. This study systematically characterized several stromal populations and found that stromal phenotype and morphology was highly variable depending on cell source and exerted differential impacts on cardiac tissue function and induced pluripotent stem cell-cardiomyocyte phenotype. Therefore, the choice of supporting stromal population can differentially impact the phenotypic or functional performance of engineered cardiac tissues.
Collapse
Affiliation(s)
- Tracy A Hookway
- 1 Gladstone Institute of Cardiovascular Disease, San Francisco, California
| | - Oriane B Matthys
- 1 Gladstone Institute of Cardiovascular Disease, San Francisco, California.,2 UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California
| | | | - Sarah Rains
- 1 Gladstone Institute of Cardiovascular Disease, San Francisco, California.,3 Department of Bioengineering, University of Texas at Dallas, Richardson, Texas
| | - Jessica E Sepulveda
- 1 Gladstone Institute of Cardiovascular Disease, San Francisco, California.,4 Biological Sciences Department, Humboldt State University, Arcata, California
| | - David A Joy
- 1 Gladstone Institute of Cardiovascular Disease, San Francisco, California.,2 UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, California
| | - Todd C McDevitt
- 1 Gladstone Institute of Cardiovascular Disease, San Francisco, California.,5 Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| |
Collapse
|
25
|
Bernhardt M, Nicolas JD, Osterhoff M, Mittelstädt H, Reuss M, Harke B, Wittmeier A, Sprung M, Köster S, Salditt T. A beamline-compatible STED microscope for combined visible-light and X-ray studies of biological matter. JOURNAL OF SYNCHROTRON RADIATION 2019; 26:1144-1151. [PMID: 31274438 DOI: 10.1107/s1600577519004089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 03/26/2019] [Indexed: 06/09/2023]
Abstract
A dedicated stimulated emission depletion (STED) microscope had been designed and implemented into the Göttingen Instrument for Nano-Imaging with X-rays (GINIX) at the synchrotron beamline P10 of the PETRA III storage ring (DESY, Hamburg). The microscope was installed on the same optical table used for X-ray holography and scanning small-angle X-ray scattering (SAXS). Scanning SAXS was implemented with the Kirkpatrick-Baez (KB) nano-focusing optics of GINIX, while X-ray holography used a combined KB and X-ray waveguide optical system for full-field projection recordings at a defocus position of the object. The STED optical axis was aligned (anti-)parallel to the focused synchrotron beam and was laterally displaced from the KB focus. This close proximity between the STED and the X-ray probe enabled in situ combined recordings on the same biological cell, tissue or any other biomolecular sample, using the same environment and mounting. Here, the instrumentation and experimental details of this correlative microscopy approach are described, as first published in our preceding work [Bernhardt et al. (2018), Nat. Commun. 9, 3641], and the capabilities of correlative STED microscopy, X-ray holography and scanning SAXS are illustrated by presenting additional datasets on cardiac tissue cells with labeled actin cytoskeleton.
Collapse
Affiliation(s)
- Marten Bernhardt
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Jan David Nicolas
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Markus Osterhoff
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Haugen Mittelstädt
- Abberior Instruments, Hans-Adolf-Krebs-Weg 1, D-37077 Göttingen, Germany
| | - Matthias Reuss
- Abberior Instruments, Hans-Adolf-Krebs-Weg 1, D-37077 Göttingen, Germany
| | - Benjamin Harke
- Abberior Instruments, Hans-Adolf-Krebs-Weg 1, D-37077 Göttingen, Germany
| | - Andrew Wittmeier
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Michael Sprung
- Deutsches Elektronen-Synchrotron (DESY), Notkestrasse 47c, D-22607 Hamburg, Germany
| | - Sarah Köster
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| | - Tim Salditt
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077 Göttingen, Germany
| |
Collapse
|
26
|
Kudryashova N, Nizamieva A, Tsvelaya V, Panfilov AV, Agladze KI. Self-organization of conducting pathways explains electrical wave propagation in cardiac tissues with high fraction of non-conducting cells. PLoS Comput Biol 2019; 15:e1006597. [PMID: 30883540 PMCID: PMC6438583 DOI: 10.1371/journal.pcbi.1006597] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/28/2019] [Accepted: 02/04/2019] [Indexed: 02/04/2023] Open
Abstract
Cardiac fibrosis occurs in many forms of heart disease and is considered to be one of the main arrhythmogenic factors. Regions with a high density of fibroblasts are likely to cause blocks of wave propagation that give rise to dangerous cardiac arrhythmias. Therefore, studies of the wave propagation through these regions are very important, yet the precise mechanisms leading to arrhythmia formation in fibrotic cardiac tissue remain poorly understood. Particularly, it is not clear how wave propagation is organized at the cellular level, as experiments show that the regions with a high percentage of fibroblasts (65-75%) are still conducting electrical signals, whereas geometric analysis of randomly distributed conducting and non-conducting cells predicts connectivity loss at 40% at the most (percolation threshold). To address this question, we used a joint in vitro-in silico approach, which combined experiments in neonatal rat cardiac monolayers with morphological and electrophysiological computer simulations. We have shown that the main reason for sustainable wave propagation in highly fibrotic samples is the formation of a branching network of cardiomyocytes. We have successfully reproduced the morphology of conductive pathways in computer modelling, assuming that cardiomyocytes align their cytoskeletons to fuse into cardiac syncytium. The electrophysiological properties of the monolayers, such as conduction velocity, conduction blocks and wave fractionation, were reproduced as well. In a virtual cardiac tissue, we have also examined the wave propagation at the subcellular level, detected wavebreaks formation and its relation to the structure of fibrosis and, thus, analysed the processes leading to the onset of arrhythmias. Cardiac arrhythmias are one of the major causes of death in the industrialized world. The most dangerous ones are often caused by the blocks of propagation of electrical signals. One of the common factors that contribute to the likelihood of these blocks, is a condition called cardiac fibrosis. In fibrosis, excitable cardiac tissue is partially replaced with the inexcitable and non-conducting connective tissue. The precise mechanisms leading to arrhythmia formation in fibrotic cardiac tissue remain poorly understood. Therefore, it is important to study wave propagation in fibrosis from cellular to tissue level. In this paper, we study tissues with high densities of non-conducting cells in experiments and computer simulations. We have observed a paradoxical ability of the tissue with extremely high portion of non-conducting cells (up to 75%) to conduct electrical signals and contract synchronously, whereas geometric analysis of randomly distributed cells predicted connectivity loss at 40% at the most. To explain this phenomenon, we have studied the patterns that cardiac cells form in the tissue and reproduced their self-organisation in a computer model. Our virtual model also took into account the polygonal shapes of the spreading cells and explained high arrhythmogenicity of fibrotic tissue.
Collapse
Affiliation(s)
- Nina Kudryashova
- Laboratory of Biophysics of Excitable Systems, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Aygul Nizamieva
- Laboratory of Biophysics of Excitable Systems, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Valeriya Tsvelaya
- Laboratory of Biophysics of Excitable Systems, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Alexander V. Panfilov
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
- Laboratory of Computational Biology and Medicine, Ural Federal University, Ekaterinburg, Russia
| | - Konstantin I. Agladze
- Laboratory of Biophysics of Excitable Systems, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- * E-mail:
| |
Collapse
|
27
|
Nguyen DT, Nagarajan N, Zorlutuna P. Effect of Substrate Stiffness on Mechanical Coupling and Force Propagation at the Infarct Boundary. Biophys J 2018; 115:1966-1980. [PMID: 30473015 PMCID: PMC6303235 DOI: 10.1016/j.bpj.2018.08.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 07/15/2018] [Accepted: 08/20/2018] [Indexed: 12/17/2022] Open
Abstract
Heterogeneous intercellular coupling plays a significant role in mechanical and electrical signal transmission in the heart. Although many studies have investigated the electrical signal conduction between myocytes and nonmyocytes within the heart muscle tissue, there are not many that have looked into the mechanical counterpart. This study aims to investigate the effect of substrate stiffness and the presence of cardiac myofibroblasts (CMFs) on mechanical force propagation across cardiomyocytes (CMs) and CMFs in healthy and heart-attack-mimicking matrix stiffness conditions. The contractile forces generated by the CMs and their propagation across the CMFs were measured using a bio-nanoindenter integrated with fluorescence microscopy for fast calcium imaging. Our results showed that softer substrates facilitated stronger and further signal transmission. Interestingly, the presence of the CMFs attenuated the signal propagation in a stiffness-dependent manner. Stiffer substrates with CMFs present attenuated the signal ∼24-32% more compared to soft substrates with CMFs, indicating a synergistic detrimental effect of increased matrix stiffness and increased CMF numbers after myocardial infarction on myocardial function. Furthermore, the beating pattern of the CMF movement at the CM-CMF boundary also depended on the substrate stiffness, thereby influencing the waveform of the propagation of CM-generated contractile forces. We performed computer simulations to further understand the occurrence of different force transmission patterns and showed that cell-matrix focal adhesions assembled at the CM-CMF interfaces, which differs depending on the substrates stiffness, play important roles in determining the efficiency and mechanism of signal transmission. In conclusion, in addition to substrate stiffness, the degree and type of cell-cell and cell-matrix interactions, affected by the substrate stiffness, influence mechanical signal conduction between myocytes and nonmyocytes in the heart muscle tissue.
Collapse
Affiliation(s)
- Dung Trung Nguyen
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Neerajha Nagarajan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana.
| |
Collapse
|
28
|
Many Cells Make Life Work-Multicellularity in Stem Cell-Based Cardiac Disease Modelling. Int J Mol Sci 2018; 19:ijms19113361. [PMID: 30373227 PMCID: PMC6274721 DOI: 10.3390/ijms19113361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cardiac disease causes 33% of deaths worldwide but our knowledge of disease progression is still very limited. In vitro models utilising and combining multiple, differentiated cell types have been used to recapitulate the range of myocardial microenvironments in an effort to delineate the mechanical, humoral, and electrical interactions that modulate the cardiac contractile function in health and the pathogenesis of human disease. However, due to limitations in isolating these cell types and changes in their structure and function in vitro, the field is now focused on the development and use of stem cell-derived cell types, most notably, human-induced pluripotent stem cell-derived CMs (hiPSC-CMs), in modelling the CM function in health and patient-specific diseases, allowing us to build on the findings from studies using animal and adult human CMs. It is becoming increasingly appreciated that communications between cardiomyocytes (CMs), the contractile cell of the heart, and the non-myocyte components of the heart not only regulate cardiac development and maintenance of health and adult CM functions, including the contractile state, but they also regulate remodelling in diseases, which may cause the chronic impairment of the contractile function of the myocardium, ultimately leading to heart failure. Within the myocardium, each CM is surrounded by an intricate network of cell types including endothelial cells, fibroblasts, vascular smooth muscle cells, sympathetic neurons, and resident macrophages, and the extracellular matrix (ECM), forming complex interactions, and models utilizing hiPSC-derived cell types offer a great opportunity to investigate these interactions further. In this review, we outline the historical and current state of disease modelling, focusing on the major milestones in the development of stem cell-derived cell types, and how this technology has contributed to our knowledge about the interactions between CMs and key non-myocyte components of the heart in health and disease, in particular, heart failure. Understanding where we stand in the field will be critical for stem cell-based applications, including the modelling of diseases that have complex multicellular dysfunctions.
Collapse
|
29
|
Bernhardt M, Nicolas JD, Osterhoff M, Mittelstädt H, Reuss M, Harke B, Wittmeier A, Sprung M, Köster S, Salditt T. Correlative microscopy approach for biology using X-ray holography, X-ray scanning diffraction and STED microscopy. Nat Commun 2018; 9:3641. [PMID: 30194418 PMCID: PMC6128893 DOI: 10.1038/s41467-018-05885-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/30/2018] [Indexed: 12/31/2022] Open
Abstract
We present a correlative microscopy approach for biology based on holographic X-ray imaging, X-ray scanning diffraction, and stimulated emission depletion (STED) microscopy. All modalities are combined into the same synchrotron endstation. In this way, labeled and unlabeled structures in cells are visualized in a complementary manner. We map out the fluorescently labeled actin cytoskeleton in heart tissue cells and superimpose the data with phase maps from X-ray holography. Furthermore, an array of local far-field diffraction patterns is recorded in the regime of small-angle X-ray scattering (scanning SAXS), which can be interpreted in terms of biomolecular shape and spatial correlations of all contributing scattering constituents. We find that principal directions of anisotropic diffraction patterns coincide to a certain degree with the actin fiber directions and that actin stands out in the phase maps from holographic recordings. In situ STED recordings are proposed to formulate models for diffraction data based on co-localization constraints.
Collapse
Affiliation(s)
- M Bernhardt
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077, Göttingen, Germany
| | - J-D Nicolas
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077, Göttingen, Germany
| | - M Osterhoff
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077, Göttingen, Germany
| | - H Mittelstädt
- Abberior Instruments, Hans-Adolf-Krebs-Weg 1, D-37077, Göttingen, Germany
| | - M Reuss
- Abberior Instruments, Hans-Adolf-Krebs-Weg 1, D-37077, Göttingen, Germany
| | - B Harke
- Abberior Instruments, Hans-Adolf-Krebs-Weg 1, D-37077, Göttingen, Germany
| | - A Wittmeier
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077, Göttingen, Germany
| | - M Sprung
- Deutsches Elektronen-Synchrotron (DESY), Notkestraße 47c, D-22607, Hamburg, Germany
| | - S Köster
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077, Göttingen, Germany
| | - T Salditt
- Institut für Röntgenphysik, Universität Göttingen, Friedrich-Hund-Platz 1, D-37077, Göttingen, Germany.
| |
Collapse
|
30
|
Matsuda Y, Takahashi K, Kamioka H, Naruse K. Human gingival fibroblast feeder cells promote maturation of induced pluripotent stem cells into cardiomyocytes. Biochem Biophys Res Commun 2018; 503:1798-1804. [PMID: 30060947 DOI: 10.1016/j.bbrc.2018.07.116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 07/23/2018] [Indexed: 11/26/2022]
Abstract
The use of human induced pluripotent stem (iPS) cells has been investigated in multiple regenerative medicine studies. However, although methods for efficient differentiation of iPS cells into heart tissues have been devised, it remains difficult to obtain cardiac tissue with high contractility. Herein, we established a method for differentiating iPS cells into highly contractile cardiomyocytes (CMs), and demonstrate that the use of human gingival fibroblasts (HGFs) as a feeder cells promotes maturation of iPS-derived CMs (iPS-CMs) in vitro. After CM differentiation of iPS cells, iPS-CMs showed increased mRNA expression of the CM specific maker cardiac troponin T (cTnT) in the absence and presence (on-feeder condition) of cocultured HGFs, and decreased expression of pluripotent markers was observed under both conditions. Protein expression of cTnT was also observed in immunocytochemical analyses, although on-feeder CMs showed comparatively robust sarcomere structure and significantly stronger contractility than feederless cardiomyocytes, suggesting that HGF feeder cells facilitate CM differentiation of iPS cells.
Collapse
Affiliation(s)
- Yusuke Matsuda
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan; Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
31
|
Nicolas JD, Bernhardt M, Schlick SF, Tiburcy M, Zimmermann WH, Khan A, Markus A, Alves F, Toischer K, Salditt T. X-ray diffraction imaging of cardiac cells and tissue. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 144:151-165. [PMID: 29914693 DOI: 10.1016/j.pbiomolbio.2018.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/18/2018] [Accepted: 05/25/2018] [Indexed: 12/30/2022]
Abstract
With the development of advanced focusing optics for x-rays, we can now use x-ray beams with spot sizes in the micro- or nanometer range to scan cells and large areas of tissues and continuously record the diffraction signals. From this data, x-ray scattering maps or so-called x-ray darkfield images are computed showing how different types of cells or regions of tissues differ in their diffraction intensity. At the same time a diffraction pattern is available for each scan point which encodes the local nanostructure, averaged over many contributing constituents illuminated by the beam. In this work we have exploited these new capabilities of scanning x-ray diffraction to investigate cardiac muscle cells as well as cardiac tissue. We give examples of how cardiac cells, especially living, cultured cells, can be prepared to be compatible with the instrumentation constraints of nano- or micro-diffraction instruments. Furthermore, we show how the developmental stage, ranging from neonatal to adult cells, as well as the final preparation state of the cardiomyocytes influences the recorded scattering signal and how these diffraction signals compare to the structure of a fully developed cardiac muscle.
Collapse
Affiliation(s)
- Jan-David Nicolas
- Universität Göttingen, Institut für Röntgenphysik, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany
| | - Marten Bernhardt
- Universität Göttingen, Institut für Röntgenphysik, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany
| | - Susanne F Schlick
- Universitätsmedizin Göttingen, Institut für Pharmakologie und Toxikologie, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Malte Tiburcy
- Universitätsmedizin Göttingen, Institut für Pharmakologie und Toxikologie, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Universitätsmedizin Göttingen, Institut für Pharmakologie und Toxikologie, Robert-Koch-Str. 40, 37075 Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Oudenarder Straße 16, 13347 Berlin, Germany
| | - Amara Khan
- Max-Planck-Institut für Experimentelle Medizin, Hermann-Rein-Straße 3, 37075 Göttingen, Germany; Universitätsmedizin Göttingen, Institut für Diagnostische und Interventionelle Radiologie, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Andrea Markus
- Max-Planck-Institut für Experimentelle Medizin, Hermann-Rein-Straße 3, 37075 Göttingen, Germany
| | - Frauke Alves
- Max-Planck-Institut für Experimentelle Medizin, Hermann-Rein-Straße 3, 37075 Göttingen, Germany; Universitätsmedizin Göttingen, Institut für Diagnostische und Interventionelle Radiologie, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Karl Toischer
- Universitätsmedizin Göttingen, Klinik für Kardiologie und Pneumologie, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Tim Salditt
- Universität Göttingen, Institut für Röntgenphysik, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
| |
Collapse
|
32
|
Parviz M, Toshniwal P, Viola HM, Hool LC, Fear PMW, Wood FM, Gaus K, Iyer KS, Gooding JJ. Real-Time Bioimpedance Sensing of Antifibrotic Drug Action in Primary Human Cells. ACS Sens 2017; 2:1482-1490. [PMID: 28871791 DOI: 10.1021/acssensors.7b00442] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Fibrotic diseases are among the most serious health issues with severe burdens due to their chronic nature and a large number of patients suffering from the debilitating effects and long-term sequelae. Collagenase treatment is a nonsurgical option but has limited results. To date, there is no potent noninvasive solution for fibrosis. Part of the reason for this is the lack of appropriate in vitro live cell screening tools to assess the efficacy of new therapeutical agents. Here, we demonstrate the utility of a cell-based electrochemical impedance biosensor platform to screen the efficacy of potential antifibrotic compounds. The platform employs a label-free and noninvasive strategy to detect the progression of fibrosis and the potency of the antifibrotic molecules in real-time. The fundamental principle that governs this novel system is that dynamic changes in cell shape and adhesion during fibrosis can be measured accurately by monitoring the changes in the impedance. This is achieved by growing the cells on a transparent interdigitated indium tin oxide (ITO) electrodes. It was demonstrated by monitoring the efficacy of a model antifibrotic compound, PXS64, on cells collected from patients with Dupuytren's contracture. We confirmed the validity of the developed biochemical impedance biosensor as an tool for in vitro screening of antifibrotic compounds and provided quantitative information on subcellular influences of the examined chemical molecules using correlative microscopy analyses that monitor the average cell area, cell morphology, and the amount and directionality of the deposited extracellular matrix protein collagen and measurement of cytosolic Ca2+ changes.
Collapse
Affiliation(s)
| | | | | | - Livia C. Hool
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
| | - P. Mark W. Fear
- Burns
Service of Western Australia, Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | - Fiona M. Wood
- Burns
Service of Western Australia, Royal Perth Hospital, Perth, Western Australia 6000, Australia
| | | | | | | |
Collapse
|
33
|
Zykov V, Krekhov A, Bodenschatz E. Geometrical factors in propagation block and spiral wave initiation. CHAOS (WOODBURY, N.Y.) 2017; 27:093923. [PMID: 28964142 DOI: 10.1063/1.4999473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 06/28/2017] [Indexed: 05/27/2023]
Abstract
Many theoretical and experimental studies indicate that a propagation block represents an important factor in spiral wave initiation in excitable media. The analytical and numerical results we obtained for a generic two-component reaction-diffusion system demonstrate quantitative conditions for the propagation block in a one-dimensional and a two-dimensional medium due to a sharp spatial increase of the medium's excitability or the coupling strength above a certain critical value. Here, we prove that this critical value strongly depends on the medium parameters and the geometry of the inhomogeneity. For an exemplary two-dimensional medium, we show how the propagation block can be used to initiate spiral waves by a specific choice of the size and shape of the medium's inhomogeneity.
Collapse
Affiliation(s)
- Vladimir Zykov
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
| | - Alexei Krekhov
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
| | - Eberhard Bodenschatz
- Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
| |
Collapse
|
34
|
Figtree GA, Bubb KJ, Tang O, Kizana E, Gentile C. Vascularized Cardiac Spheroids as Novel 3D in vitro Models to Study Cardiac Fibrosis. Cells Tissues Organs 2017; 204:191-198. [DOI: 10.1159/000477436] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2017] [Indexed: 12/21/2022] Open
Abstract
Spheroid cultures are among the most explored cellular biomaterials used in cardiovascular research, due to their improved integration of biochemical and physiological features of the heart in a defined architectural three-dimensional microenvironment when compared to monolayer cultures. To further explore the potential use of spheroid cultures for research, we engineered a novel in vitro model of the heart with vascularized cardiac spheroids (VCSs), by coculturing cardiac myocytes, endothelial cells, and fibroblasts isolated from dissociated rat neonatal hearts (aged 1-3 days) in hanging drop cultures. To evaluate the validity of VCSs in recapitulating pathophysiological processes typical of the in vivo heart, such as cardiac fibrosis, we then treated VCSs with transforming growth factor beta 1 (TGFβ1), a known profibrotic agent. Our mRNA analysis demonstrated that TGFβ1-treated VCSs present elevated levels of expression of connective tissue growth factor, fibronectin, and TGFβ1 when compared to control cultures. We demonstrated a dramatic increase in collagen deposition following TGFβ1 treatment in VCSs in the PicroSirius Red-stained sections. Doxorubicin, a renowned cardiotoxic and profibrotic agent, triggered apoptosis and disrupted vascular networks in VCSs. Taken together, our findings demonstrate that VCSs are a valid model for the study of the mechanisms involved in cardiac fibrosis, with the potential to be used to investigate novel mechanisms and therapeutics for treating and preventing cardiac fibrosis in vitro.
Collapse
|
35
|
Fast propagation regions cause self-sustained reentry in excitable media. Proc Natl Acad Sci U S A 2017; 114:1281-1286. [PMID: 28123066 DOI: 10.1073/pnas.1611475114] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Self-sustained waves of electrophysiological activity can cause arrhythmia in the heart. These reentrant excitations have been associated with spiral waves circulating around either an anatomically defined weakly conducting region or a functionally determined core. Recently, an ablation procedure has been clinically introduced that stops atrial fibrillation of the heart by destroying the electrical activity at the spiral core. This is puzzling because the tissue at the anatomically defined spiral core would already be weakly conducting, and a further decrease should not improve the situation. In the case of a functionally determined core, an ablation procedure should even further stabilize the rotating wave. The efficacy of the procedure thus needs explanation. Here, we show theoretically that fundamentally in any excitable medium a region with a propagation velocity faster than its surrounding can act as a nucleation center for reentry and can anchor an induced spiral wave. Our findings demonstrate a mechanistic underpinning for the recently developed ablation procedure. Our theoretical results are based on a very general and widely used two-component model of an excitable medium. Moreover, the important control parameters used to realize conditions for the discovered phenomena are applicable to quite different multicomponent models.
Collapse
|
36
|
Judd J, Lovas J, Huang GN. Isolation, Culture and Transduction of Adult Mouse Cardiomyocytes. J Vis Exp 2016. [PMID: 27685811 DOI: 10.3791/54012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Cultured cardiomyocytes can be used to study cardiomyocyte biology using techniques that are complementary to in vivo systems. For example, the purity and accessibility of in vitro culture enables fine control over biochemical analyses, live imaging, and electrophysiology. Long-term culture of cardiomyocytes offers access to additional experimental approaches that cannot be completed in short term cultures. For example, the in vitro investigation of dedifferentiation, cell cycle re-entry, and cell division has thus far largely been restricted to rat cardiomyocytes, which appear to be more robust in long-term culture. However, the availability of a rich toolset of transgenic mouse lines and well-developed disease models make mouse systems attractive for cardiac research. Although several reports exist of adult mouse cardiomyocyte isolation, few studies demonstrate their long-term culture. Presented here, is a step-by-step method for the isolation and long-term culture of adult mouse cardiomyocytes. First, retrograde Langendorff perfusion is used to efficiently digest the heart with proteases, followed by gravity sedimentation purification. After a period of dedifferentiation following isolation, the cells gradually attach to the culture and can be cultured for weeks. Adenovirus cell lysate is used to efficiently transduce the isolated cardiomyocytes. These methods provide a simple, yet powerful model system to study cardiac biology.
Collapse
Affiliation(s)
- Justin Judd
- Cardiovascular Research Institute, University of California, San Francisco
| | - Jonathan Lovas
- Cardiovascular Research Institute, University of California, San Francisco
| | - Guo N Huang
- Cardiovascular Research Institute, University of California, San Francisco;
| |
Collapse
|
37
|
Novel therapeutic strategies targeting fibroblasts and fibrosis in heart disease. Nat Rev Drug Discov 2016; 15:620-638. [PMID: 27339799 DOI: 10.1038/nrd.2016.89] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our understanding of the functions of cardiac fibroblasts has moved beyond their roles in heart structure and extracellular matrix generation and now includes their contributions to paracrine, mechanical and electrical signalling during ontogenesis and normal cardiac activity. Fibroblasts also have central roles in pathogenic remodelling during myocardial ischaemia, hypertension and heart failure. As key contributors to scar formation, they are crucial for tissue repair after interventions including surgery and ablation. Novel experimental approaches targeting cardiac fibroblasts are promising potential therapies for heart disease. Indeed, several existing drugs act, at least partially, through effects on cardiac connective tissue. This Review outlines the origins and roles of fibroblasts in cardiac development, homeostasis and disease; illustrates the involvement of fibroblasts in current and emerging clinical interventions; and identifies future targets for research and development.
Collapse
|
38
|
Pellman J, Zhang J, Sheikh F. Myocyte-fibroblast communication in cardiac fibrosis and arrhythmias: Mechanisms and model systems. J Mol Cell Cardiol 2016; 94:22-31. [PMID: 26996756 DOI: 10.1016/j.yjmcc.2016.03.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/27/2016] [Accepted: 03/14/2016] [Indexed: 12/17/2022]
Abstract
Development of cardiac fibrosis and arrhythmias is controlled by the activity of and communication between cardiomyocytes and fibroblasts in the heart. Myocyte-fibroblast interactions occur via both direct and indirect means including paracrine mediators, extracellular matrix interactions, electrical modulators, mechanical junctions, and membrane nanotubes. In the diseased heart, cardiomyocyte and fibroblast ratios and activity, and thus myocyte-fibroblast interactions, change and are thought to contribute to the course of disease including development of fibrosis and arrhythmogenic activity. Fibroblasts have a developing role in modulating cardiomyocyte electrical and hypertrophic activity, however gaps in knowledge regarding these interactions still exist. Research in this field has necessitated the development of unique approaches to isolate and control myocyte-fibroblast interactions. Numerous methods for 2D and 3D co-culture systems have been developed, while a growing part of this field is in the use of better tools for in vivo systems including cardiomyocyte and fibroblast specific Cre mouse lines for cell type specific genetic ablation. This review will focus on (i) mechanisms of myocyte-fibroblast communication and their effects on disease features such as cardiac fibrosis and arrhythmias as well as (ii) methods being used and currently developed in this field.
Collapse
Affiliation(s)
- Jason Pellman
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jing Zhang
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Farah Sheikh
- Department of Medicine, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|