1
|
Mir M, Chen J, Patel A, Pinezich MR, Hudock MR, Yoon A, Diane M, O'Neill J, Bacchetta M, Vunjak-Novakovic G, Kim J. Bioimpedance measurements of fibrotic and acutely injured lung tissues. Acta Biomater 2025; 194:270-287. [PMID: 39870150 PMCID: PMC11877686 DOI: 10.1016/j.actbio.2025.01.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/12/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
In injured and diseased tissues, changes in molecular and cellular compositions, as well as tissue architecture, lead to alterations in both physiological and physical characteristics. Notably, the electrical properties of tissues, which can be characterized as bioelectrical impedance (bioimpedance), are closely linked to the health and pathological conditions of the tissues. This highlights the significant role of quantitatively characterizing these electrical properties in improving the accuracy and speed of diagnosis and prognosis. In this study, we investigate how diseases, injuries, and physical conditions can affect the electrical properties of lung tissues, using both rat and human lung tissue samples. Results showed that rat lung and trachea tissues exhibit a frequency-dependent behavior to alternating current (AC) across the frequency range of 0.1-300 kHz. The bioimpedance of the lung tissue increased with the level of aeration of the lung, which was manipulated by altering alveolar pressure (PALV: 1-15 cmH2O; bioimpedance level: 1.2-2.8 kΩ; AC frequency: 2 kHz). This increase is mainly because air is electrically nonconductive. The bioimpedance of rat lungs injured via intratracheal aspiration of hydrochloric acid (HCl; volume: 1 mL; AC frequency: 2 kHz) decreased by at least 82 % compared to that of healthy control lungs due to accumulation of fluids inside the airspace of the injured lungs. Moreover, using decellularized lung tissues, we determined the contributions of cellular components and tissue extracellular matrix (ECM) on the electrical characteristics of the lung tissues. Specifically, we observed a considerable increase in bioimpedance in fibrotic human lung tissues due to excessive ECM deposition (healthy: 70.8 Ω ± 10.2 Ω, fibrotic: 132.1 Ω ± 15.8 Ω, frequency: 2 kHz). Overall, the findings of this study can enhance our understanding of the correlation between electrical properties and pathological lung conditions, thereby improving diagnostic and prognostic capabilities and aiding in the treatment of lung diseases and injuries. STATEMENT OF SIGNIFICANCE: The bioelectrical properties of tissue are closely linked to both its physiological and physical characteristics. This underscores the importance of quantitatively characterizing these properties to improve the accuracy and speed of diagnosis and prognosis. In this study, we investigate how the bioelectrical properties of lung tissues are affected by different physical states and pathological conditions using rat and human lung tissues. As the burden of lung diseases continues to increase, our findings can contribute to improved treatment outcomes by enabling accurate and rapid assessment of lung tissue conditions.
Collapse
Affiliation(s)
- Mohammad Mir
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Jiawen Chen
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Aneri Patel
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Meghan R Pinezich
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Maria R Hudock
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Alexander Yoon
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Mohamed Diane
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - John O'Neill
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Matthew Bacchetta
- Department of Cardiac Surgery, Vanderbilt University, Nashville, TN, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, USA; Department of Medicine, Columbia University, New York, NY, USA
| | - Jinho Kim
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA.
| |
Collapse
|
2
|
Braidotti N, Demontis G, Conti M, Andolfi L, Ciubotaru CD, Sbaizero O, Cojoc D. The local mechanosensitive response of primary cardiac fibroblasts is influenced by the microenvironment mechanics. Sci Rep 2024; 14:10365. [PMID: 38710778 PMCID: PMC11074268 DOI: 10.1038/s41598-024-60685-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/26/2024] [Indexed: 05/08/2024] Open
Abstract
Cardiac fibroblasts (CFs) are essential for preserving myocardial integrity and function. They can detect variations in cardiac tissue stiffness using various cellular mechanosensors, including the Ca2+ permeable mechanosensitive channel Piezo1. Nevertheless, how CFs adapt the mechanosensitive response to stiffness changes remains unclear. In this work we adopted a multimodal approach, combining the local mechanical stimulation (from 10 pN to 350 nN) with variations of culture substrate stiffness. We found that primary rat CFs cultured on stiff (GPa) substrates showed a broad Piezo1 distribution in the cell with particular accumulation at the mitochondria membrane. CFs displayed a force-dependent behavior in both calcium uptake and channel activation probability, showing a threshold at 300 nN, which involves both cytosolic and mitochondrial Ca2+ mobilization. This trend decreases as the myofibroblast phenotype within the cell population increases, following a possible Piezo1 accumulation at focal adhesion sites. In contrast, the inhibition of fibroblasts to myofibroblasts transition with soft substrates (kPa) considerably reduces both mechanically- and chemically-induced Piezo1 activation and expression. Our findings shed light on how Piezo1 function and expression are regulated by the substrate stiffness and highlight its involvement in the environment-mediated modulation of CFs mechanosensitivity.
Collapse
Affiliation(s)
- Nicoletta Braidotti
- Department of Physics, University of Trieste, Via A. Valerio 2, 34127, Trieste, Italy
- CNR-Istituto Officina dei Materiali (IOM), SS 14 km 163.5, Area Science Park Basovizza, 34149, Trieste, Italy
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, Via L. Giorgieri 1, 34127, Trieste, Italy
| | - Giorgia Demontis
- Department of Physics, University of Trieste, Via A. Valerio 2, 34127, Trieste, Italy
- CNR-Istituto Officina dei Materiali (IOM), SS 14 km 163.5, Area Science Park Basovizza, 34149, Trieste, Italy
| | - Martina Conti
- CNR-Istituto Officina dei Materiali (IOM), SS 14 km 163.5, Area Science Park Basovizza, 34149, Trieste, Italy
| | - Laura Andolfi
- CNR-Istituto Officina dei Materiali (IOM), SS 14 km 163.5, Area Science Park Basovizza, 34149, Trieste, Italy
| | - Catalin Dacian Ciubotaru
- CNR-Istituto Officina dei Materiali (IOM), SS 14 km 163.5, Area Science Park Basovizza, 34149, Trieste, Italy
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, Via A. Valerio 6/A, 34127, Trieste, Italy
| | - Dan Cojoc
- CNR-Istituto Officina dei Materiali (IOM), SS 14 km 163.5, Area Science Park Basovizza, 34149, Trieste, Italy.
| |
Collapse
|
3
|
Tsuji Y, Ogata T, Mochizuki K, Tamura S, Morishita Y, Takamatsu T, Matoba S, Tanaka H. Myofibroblasts impair myocardial impulse propagation by heterocellular connexin43 gap-junctional coupling through micropores. Front Physiol 2024; 15:1352911. [PMID: 38465264 PMCID: PMC10920281 DOI: 10.3389/fphys.2024.1352911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/07/2024] [Indexed: 03/12/2024] Open
Abstract
Aim: Composite population of myofibroblasts (MFs) within myocardial tissue is known to alter impulse propagation, leading to arrhythmias. However, it remains unclear whether and how MFs alter their propagation patterns when contacting cardiomyocytes (CMs) without complex structural insertions in the myocardium. We attempted to unveil the effects of the one-sided, heterocellular CM-MF connection on the impulse propagation of CM monolayers without the spatial insertion of MFs as an electrical or mechanical obstacle. Methods and results: We evaluated fluo8-based spatiotemporal patterns in impulse propagation of neonatal rat CM monolayers cultured on the microporous membrane having 8-μm diameter pores with co-culture of MFs or CMs on the reverse membrane side (CM-MF model or CM-CM model, respectively). During consecutive pacing at 1 or 2 Hz, the CM monolayers exhibited forward impulse propagation from the pacing site with a slower conduction velocity (θ) and a larger coefficient of directional θ variation in the CM-MF model than that in the CM-CM model in a frequency-dependent manner (2 Hz >1 Hz). The localized placement of an MF cluster on the reverse side resulted in an abrupt segmental depression of the impulse propagation of the upper CM layer, causing a spatiotemporally non-uniform pattern. Dye transfer of the calcein loaded in the upper CM layer to the lower MF layer was attenuated by the gap-junction inhibitor heptanol. Immunocytochemistry identified definitive connexin 43 (Cx43) between the CMs and MFs in the membrane pores. MF-selective Cx43 knockdown in the MF layer improved both the velocity and uniformity of propagation in the CM monolayer. Conclusion: Heterocellular Cx43 gap junction coupling of CMs with MFs alters the spatiotemporal patterns of myocardial impulse propagation, even in the absence of spatially interjacent and mechanosensitive modulations by MFs. Moreover, MFs can promote pro-arrhythmogenic impulse propagation when in face-to-face contact with the myocardium that arises in the healing infarct border zone.
Collapse
Affiliation(s)
- Yumika Tsuji
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takehiro Ogata
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kentaro Mochizuki
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shoko Tamura
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuma Morishita
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuro Takamatsu
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Medical Photonics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation and, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Faculty of Health and Medical Sciences, Kyoto University of Advanced Science, Kyoto, Japan
| |
Collapse
|
4
|
Ghosh A, Sekar A, Saravanan S, Sriram CS, Pandurangi UM. An unusual site of ablation for a ventricular tachycardia. J Arrhythm 2024; 40:160-165. [PMID: 38333384 PMCID: PMC10848586 DOI: 10.1002/joa3.12954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/18/2023] [Accepted: 10/28/2023] [Indexed: 02/10/2024] Open
Abstract
Scar-related ventricular tachycardia (VT) ablation involves localizing the critical isthmuses by overdrive pacing maneuvers and three-dimensional activation mapping. Implantable prosthetic devices have been known to complicate this by covering sites of potential isthmuses. We herein present a sentinel report of scar-VT ablation with a protected isthmus localized over an endothelialized post-myocardial infarction ventricular septal defect occluder device.
Collapse
Affiliation(s)
- Anindya Ghosh
- Department of Cardiac Electrophysiology and Pacing, Arrhythmia Heart Failure AcademyThe Madras Medical MissionChennaiTamil NaduIndia
| | - Anbarasan Sekar
- Department of Cardiac Electrophysiology and Pacing, Arrhythmia Heart Failure AcademyThe Madras Medical MissionChennaiTamil NaduIndia
| | | | - Chenni S. Sriram
- Division of CardiologySub‐section of Electrophysiology, Children's Hospital of Michigan and Detroit Medical CenterDetroitMichiganUSA
| | - Ulhas M. Pandurangi
- Department of Cardiac Electrophysiology and Pacing, Arrhythmia Heart Failure AcademyThe Madras Medical MissionChennaiTamil NaduIndia
| |
Collapse
|
5
|
Iwamiya S, Ihara K, Furukawa T, Sasano T. Sacubitril/valsartan attenuates atrial conduction disturbance and electrophysiological heterogeneity with ameliorating fibrosis in mice. Front Cardiovasc Med 2024; 11:1341601. [PMID: 38312235 PMCID: PMC10834649 DOI: 10.3389/fcvm.2024.1341601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/02/2024] [Indexed: 02/06/2024] Open
Abstract
Background Sacubitril/valsartan (SacVal) has been shown to improve the prognosis of heart failure; however, whether SacVal reduces the occurrence of atrial fibrillation (AF) in heart failure has not yet been elucidated. In this study, we aimed to determine whether SacVal is effective in reducing the occurrence of AF in heart failure and identify the underlying mechanism of its electrophysiological effect in mice. Methods Adult male mice underwent transverse aortic constriction, followed by SacVal, valsartan, or vehicle treatment for two weeks. Electrophysiological study (EPS) and optical mapping were performed to assess the susceptibility to AF and the atrial conduction properties, and fibrosis was investigated using heart tissue and isolated cardiac fibroblasts (CFs). Results EPS analysis revealed that AF was significantly less inducible in SacVal-treated mice than in vehicle-treated mice. Optical mapping of the atrium showed that SacVal-treated and valsartan-treated mice restored the prolonged action potential duration (APD); however, only SacVal-treated mice showed the restoration of decreased conduction velocity (CV) compared to vehicle-treated mice. In addition, the electrophysiological distribution analysis demonstrated that heterogeneous electrophysiological properties were rate-dependent and increased heterogeneity was closely related to the susceptibility to AF. SacVal attenuated the increased heterogeneity of CV at short pacing cycle length in atria, whereas Val could not. Histological and molecular evaluation showed that SacVal exerted the anti-fibrotic effect on the atria. An in vitro study of CFs treated with natriuretic peptides and LBQ657, the metabolite and active form of sacubitril, revealed that C-type natriuretic peptide (CNP) combined with LBQ657 had an additional anti-fibrotic effect on CFs. Conclusions Our results demonstrated that SacVal can improve the conduction disturbance and heterogeneity through the attenuation of fibrosis in murine atria and reduce the susceptibility of AF in heart failure with pressure overload, which might be attributed to the enhanced function of CNP.
Collapse
Affiliation(s)
- Satoshi Iwamiya
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kensuke Ihara
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsushi Furukawa
- Department of Bio-Informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tetsuo Sasano
- Department of Cardiovascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
6
|
Song Q, Alvarez-Laviada A, Schrup SE, Reilly-O'Donnell B, Entcheva E, Gorelik J. Opto-SICM framework combines optogenetics with scanning ion conductance microscopy for probing cell-to-cell contacts. Commun Biol 2023; 6:1131. [PMID: 37938652 PMCID: PMC10632396 DOI: 10.1038/s42003-023-05509-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
We present a novel framework, Opto-SICM, for studies of cellular interactions in live cells with high spatiotemporal resolution. The approach combines scanning ion conductance microscopy, SICM, and cell-type-specific optogenetic interrogation. Light-excitable cardiac fibroblasts (FB) and myofibroblasts (myoFB) were plated together with non-modified cardiomyocytes (CM) and then paced with periodic illumination. Opto-SICM reveals the extent of FB/myoFB-CM cell-cell contacts and the dynamic changes over time not visible by optical microscopy. FB-CM pairs have lower gap junctional expression of connexin-43 and higher contact dynamism compared to myoFB-CM pairs. The responsiveness of CM to pacing via FB/myoFB depends on the dynamics of the contact but not on the area. The non-responding pairs have higher net cell-cell movement at the contact. These findings are relevant to cardiac disease states, where adverse remodeling leads to abnormal electrical excitation of CM. The Opto-SICM framework can be deployed to offer new insights on cellular and subcellular interactions in various cell types, in real-time.
Collapse
Affiliation(s)
- Qianqian Song
- Imperial College London, Du Cane road, W12 0NN, London, UK
| | | | - Sarah E Schrup
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | | | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA.
| | - Julia Gorelik
- Imperial College London, Du Cane road, W12 0NN, London, UK.
| |
Collapse
|
7
|
Takaya H, Comtois-Bona M, Spasojevic A, Cortes D, Variola F, Liang W, Ruel M, Suuronen EJ, Alarcon EI. BEaTS-β: an open-source electromechanical bioreactor for simulating human cardiac disease conditions. Front Bioeng Biotechnol 2023; 11:1253602. [PMID: 37781536 PMCID: PMC10540188 DOI: 10.3389/fbioe.2023.1253602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Heart disease remains the leading cause of worldwide mortality. Although the last decades have broadened our understanding of the biology behind the pathologies of heart disease, ex vivo systems capable of mimicking disease progression and abnormal heart function using human cells remain elusive. In this contribution, an open-access electromechanical system (BEaTS-β) capable of mimicking the environment of cardiac disease is reported. BEaTS-β was designed using computer-aided modeling to combine tunable electrical stimulation and mechanical deformation of cells cultured on a flexible elastomer. To recapitulate the clinical scenario of a heart attack more closely, in designing BEaTS-β we considered a device capable to operate under hypoxic conditions. We tested human induced pluripotent stem cell-derived cardiomyocytes, fibroblasts, and coronary artery endothelial cells in our simulated myocardial infarction environment. Our results indicate that, under simulated myocardium infarction, there was a decrease in maturation of cardiomyocytes, and reduced survival of fibroblasts and coronary artery endothelial cells. The open access nature of BEaTS-β will allow for other investigators to use this platform to investigate cardiac cell biology or drug therapeutic efficacy in vitro under conditions that simulate arrhythmia and/or myocardial infarction.
Collapse
Affiliation(s)
- Hiroki Takaya
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Maxime Comtois-Bona
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Biomedical Mechanical Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Ana Spasojevic
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - David Cortes
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Biomedical Mechanical Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Fabio Variola
- Department of Mechanical Engineering, University of Ottawa, Ottawa, ON, Canada
| | - Wenbin Liang
- Cardiac Electrophysiology Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Marc Ruel
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Erik J. Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Emilio I. Alarcon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
8
|
Johnson RD, Lei M, McVey JH, Camelliti P. Human myofibroblasts increase the arrhythmogenic potential of human induced pluripotent stem cell-derived cardiomyocytes. Cell Mol Life Sci 2023; 80:276. [PMID: 37668685 PMCID: PMC10480244 DOI: 10.1007/s00018-023-04924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023]
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have the potential to remuscularize infarcted hearts but their arrhythmogenicity remains an obstacle to safe transplantation. Myofibroblasts are the predominant cell-type in the infarcted myocardium but their impact on transplanted hiPSC-CMs remains poorly defined. Here, we investigate the effect of myofibroblasts on hiPSC-CMs electrophysiology and Ca2+ handling using optical mapping of advanced human cell coculture systems mimicking cell-cell interaction modalities. Human myofibroblasts altered the electrophysiology and Ca2+ handling of hiPSC-CMs and downregulated mRNAs encoding voltage channels (KV4.3, KV11.1 and Kir6.2) and SERCA2a calcium pump. Interleukin-6 was elevated in the presence of myofibroblasts and direct stimulation of hiPSC-CMs with exogenous interleukin-6 recapitulated the paracrine effects of myofibroblasts. Blocking interleukin-6 reduced the effects of myofibroblasts only in the absence of physical contact between cell-types. Myofibroblast-specific connexin43 knockdown reduced functional changes in contact cocultures only when combined with interleukin-6 blockade. This provides the first in-depth investigation into how human myofibroblasts modulate hiPSC-CMs function, identifying interleukin-6 and connexin43 as paracrine- and contact-mediators respectively, and highlighting their potential as targets for reducing arrhythmic risk in cardiac cell therapy.
Collapse
Affiliation(s)
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - John H McVey
- School of Biosciences, University of Surrey, Guildford, UK
| | | |
Collapse
|
9
|
Baena-Montes JM, Kraśny MJ, O’Halloran M, Dunne E, Quinlan LR. In Vitro Models for Improved Therapeutic Interventions in Atrial Fibrillation. J Pers Med 2023; 13:1237. [PMID: 37623487 PMCID: PMC10455620 DOI: 10.3390/jpm13081237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Atrial fibrillation is the most common type of cardiac arrhythmias in humans, mostly caused by hyper excitation of specific areas in the atrium resulting in dyssynchronous atrial contractions, leading to severe consequences such as heart failure and stroke. Current therapeutics aim to target this condition through both pharmacological and non-pharmacological approaches. To test and validate any of these treatments, an appropriate preclinical model must be carefully chosen to refine and optimise the therapy features to correctly reverse this condition. A broad range of preclinical models have been developed over the years, with specific features and advantages to closely mimic the pathophysiology of atrial fibrillation. In this review, currently available models are described, from traditional animal models and in vitro cell cultures to state-of-the-art organoids and organs-on-a-chip. The advantages, applications and limitations of each model are discussed, providing the information to select the appropriate model for each research application.
Collapse
Affiliation(s)
- Jara M. Baena-Montes
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
| | - Marcin J. Kraśny
- Smart Sensors Lab, Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Martin O’Halloran
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
- Electrical & Electronic Engineering, School of Engineering, University of Galway, H91 TK33 Galway, Ireland
| | - Eoghan Dunne
- Translational Medical Device Lab (TMDLab), Lambe Institute for Translational Research, School of Medicine, University of Galway, H91 TK33 Galway, Ireland
| | - Leo R. Quinlan
- Physiology and Cellular Physiology Research Laboratory, School of Medicine, Human Biology Building, University of Galway, H91 TK33 Galway, Ireland
- CÚRAM SFI Centre for Research in Medical Devices, University of Galway, H91 TK33 Galway, Ireland
| |
Collapse
|
10
|
Belanger K, Koppes AN, Koppes RA. Impact of Non-Muscle Cells on Excitation-Contraction Coupling in the Heart and the Importance of In Vitro Models. Adv Biol (Weinh) 2023; 7:e2200117. [PMID: 36216583 DOI: 10.1002/adbi.202200117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/07/2022] [Indexed: 05/13/2023]
Abstract
Excitation-coupling (ECC) is paramount for coordinated contraction to maintain sufficient cardiac output. The study of ECC regulation has primarily been limited to cardiomyocytes (CMs), which conduct voltage waves via calcium fluxes from one cell to another, eliciting contraction of the atria followed by the ventricles. CMs rapidly transmit ionic flux via gap junction proteins, predominantly connexin 43. While the expression of connexin isoforms has been identified in each of the individual cell populations comprising the heart, the formation of gap junctions with nonmuscle cells (i.e., macrophages and Schwann cells) has gained new attention. Evaluating nonmuscle contributions to ECC in vivo or in situ remains difficult and necessitates the development of simple, yet biomimetic in vitro models to better understand and prevent physiological dysfunction. Standard 2D cell culture often consists of homogenous cell populations and lacks the dynamic mechanical environment of native tissue, confounding the phenotypic and proteomic makeup of these highly mechanosensitive cell populations in prolonged culture conditions. This review will highlight the recent developments and the importance of new microphysiological systems to better understand the complex regulation of ECC in cardiac tissue.
Collapse
Affiliation(s)
- Kirstie Belanger
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Abigail N Koppes
- Department of Bioengineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
- Department of Biology, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Ryan A Koppes
- Department of Chemical Engineering, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| |
Collapse
|
11
|
Wang EY, Zhao Y, Okhovatian S, Smith JB, Radisic M. Intersection of stem cell biology and engineering towards next generation in vitro models of human fibrosis. Front Bioeng Biotechnol 2022; 10:1005051. [PMID: 36338120 PMCID: PMC9630603 DOI: 10.3389/fbioe.2022.1005051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/26/2022] [Indexed: 08/31/2023] Open
Abstract
Human fibrotic diseases constitute a major health problem worldwide. Fibrosis involves significant etiological heterogeneity and encompasses a wide spectrum of diseases affecting various organs. To date, many fibrosis targeted therapeutic agents failed due to inadequate efficacy and poor prognosis. In order to dissect disease mechanisms and develop therapeutic solutions for fibrosis patients, in vitro disease models have gone a long way in terms of platform development. The introduction of engineered organ-on-a-chip platforms has brought a revolutionary dimension to the current fibrosis studies and discovery of anti-fibrotic therapeutics. Advances in human induced pluripotent stem cells and tissue engineering technologies are enabling significant progress in this field. Some of the most recent breakthroughs and emerging challenges are discussed, with an emphasis on engineering strategies for platform design, development, and application of machine learning on these models for anti-fibrotic drug discovery. In this review, we discuss engineered designs to model fibrosis and how biosensor and machine learning technologies combine to facilitate mechanistic studies of fibrosis and pre-clinical drug testing.
Collapse
Affiliation(s)
- Erika Yan Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Jacob B. Smith
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
12
|
Siles-Paredes JG, Crowley CJ, Fenton FH, Bhatia N, Iravanian S, Sandoval I, Pollnow S, Dössel O, Salinet J, Uzelac I. Circle Method for Robust Estimation of Local Conduction Velocity High-Density Maps From Optical Mapping Data: Characterization of Radiofrequency Ablation Sites. Front Physiol 2022; 13:794761. [PMID: 36035466 PMCID: PMC9417315 DOI: 10.3389/fphys.2022.794761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 06/15/2022] [Indexed: 01/10/2023] Open
Abstract
Conduction velocity (CV) slowing is associated with atrial fibrillation (AF) and reentrant ventricular tachycardia (VT). Clinical electroanatomical mapping systems used to localize AF or VT sources as ablation targets remain limited by the number of measuring electrodes and signal processing methods to generate high-density local activation time (LAT) and CV maps of heterogeneous atrial or trabeculated ventricular endocardium. The morphology and amplitude of bipolar electrograms depend on the direction of propagating electrical wavefront, making identification of low-amplitude signal sources commonly associated with fibrotic area difficulty. In comparison, unipolar electrograms are not sensitive to wavefront direction, but measurements are susceptible to distal activity. This study proposes a method for local CV calculation from optical mapping measurements, termed the circle method (CM). The local CV is obtained as a weighted sum of CV values calculated along different chords spanning a circle of predefined radius centered at a CV measurement location. As a distinct maximum in LAT differences is along the chord normal to the propagating wavefront, the method is adaptive to the propagating wavefront direction changes, suitable for electrical conductivity characterization of heterogeneous myocardium. In numerical simulations, CM was validated characterizing modeled ablated areas as zones of distinct CV slowing. Experimentally, CM was used to characterize lesions created by radiofrequency ablation (RFA) on isolated hearts of rats, guinea pig, and explanted human hearts. To infer the depth of RFA-created lesions, excitation light bands of different penetration depths were used, and a beat-to-beat CV difference analysis was performed to identify CV alternans. Despite being limited to laboratory research, studies based on CM with optical mapping may lead to new translational insights into better-guided ablation therapies.
Collapse
Affiliation(s)
- Jimena G. Siles-Paredes
- Graduate Program in Biotechnoscience, Federal University of ABC, São Paulo, Brazil
- HEartLab, Federal University of ABC, São Paulo, Brazil
- *Correspondence: Jimena G. Siles-Paredes,
| | | | - Flavio H. Fenton
- Georgia Institute of Technology, School of Physics, Atlanta, GA, United States
| | - Neal Bhatia
- Division of Cardiology, Section of Electrophysiology, Emory University Hospital, Atlanta, GA, United States
| | - Shahriar Iravanian
- Division of Cardiology, Section of Electrophysiology, Emory University Hospital, Atlanta, GA, United States
| | | | - Stefan Pollnow
- Karlsruhe Institute of Technology (KIT)/Institute of Biomedical Engineering, Karlsruhe, Germany
| | - Olaf Dössel
- Karlsruhe Institute of Technology (KIT)/Institute of Biomedical Engineering, Karlsruhe, Germany
| | - João Salinet
- Graduate Program in Biotechnoscience, Federal University of ABC, São Paulo, Brazil
- HEartLab, Federal University of ABC, São Paulo, Brazil
| | - Ilija Uzelac
- Georgia Institute of Technology, School of Physics, Atlanta, GA, United States
| |
Collapse
|
13
|
Nayir S, Lacour SP, Kucera JP. Active force generation contributes to the complexity of spontaneous activity and to the response to stretch of murine cardiomyocyte cultures. J Physiol 2022; 600:3287-3312. [PMID: 35679256 PMCID: PMC9541716 DOI: 10.1113/jp283083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Abstract Cardiomyocyte cultures exhibit spontaneous electrical and contractile activity, as in a natural cardiac pacemaker. In such preparations, beat rate variability exhibits features similar to those of heart rate variability in vivo. Mechanical deformations and forces feed back on the electrical properties of cardiomyocytes, but it is not fully elucidated how this mechano‐electrical interplay affects beating variability in such preparations. Using stretchable microelectrode arrays, we assessed the effects of the myosin inhibitor blebbistatin and the non‐selective stretch‐activated channel blocker streptomycin on beating variability and on the response of neonatal or fetal murine ventricular cell cultures against deformation. Spontaneous electrical activity was recorded without stretch and upon predefined deformation protocols (5% uniaxial and 2% equibiaxial strain, applied repeatedly for 1 min every 3 min). Without stretch, spontaneous activity originated from the edge of the preparations, and its site of origin switched frequently in a complex manner across the cultures. Blebbistatin did not change mean beat rate, but it decreased the spatial complexity of spontaneous activity. In contrast, streptomycin did not exert any manifest effects. During the deformation protocols, beat rate increased transiently upon stretch but, paradoxically, also upon release. Blebbistatin attenuated the response to stretch, whereas this response was not affected by streptomycin. Therefore, our data support the notion that in a spontaneously firing network of cardiomyocytes, active force generation, rather than stretch‐activated channels, is involved mechanistically in the complexity of the spatiotemporal patterns of spontaneous activity and in the stretch‐induced acceleration of beating.
![]() Key points Monolayer cultures of cardiac cells exhibit spontaneous electrical and contractile activity, as in a natural cardiac pacemaker. Beating variability in these preparations recapitulates the power‐law behaviour of heart rate variability in vivo. However, the effects of mechano‐electrical feedback on beating variability are not yet fully understood. Using stretchable microelectrode arrays, we examined the effects of the contraction uncoupler blebbistatin and the non‐specific stretch‐activated channel blocker streptomycin on beating variability and on stretch‐induced changes of beat rate. Without stretch, blebbistatin decreased the spatial complexity of beating variability, whereas streptomycin had no effects. Both stretch and release increased beat rate transiently; blebbistatin attenuated the increase of beat rate upon stretch, whereas streptomycin had no effects. Active force generation contributes to the complexity of spatiotemporal patterns of beating variability and to the increase of beat rate upon mechanical deformation. Our study contributes to the understanding of how mechano‐electrical feedback influences heart rate variability.
Collapse
Affiliation(s)
- Seyma Nayir
- Department of Physiology, University of Bern, Bern, Switzerland
| | | | - Jan P Kucera
- Department of Physiology, University of Bern, Bern, Switzerland
| |
Collapse
|
14
|
Zhang W, Li DS, Bui-Thanh T, Sacks MS. Simulation of the 3D Hyperelastic Behavior of Ventricular Myocardium using a Finite-Element Based Neural-Network Approach. COMPUTER METHODS IN APPLIED MECHANICS AND ENGINEERING 2022; 394:114871. [PMID: 35422534 PMCID: PMC9004630 DOI: 10.1016/j.cma.2022.114871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
High-fidelity cardiac models using attribute-rich finite element based models have been developed to a very mature stage. However, such finite-element based approaches remain time consuming, which have limited their clinical use. There remains a need for alternative methods for novel cardiac simulation methods of capable of high fidelity simulations in clinically relevant time frames. Surrogate models are one approach, which traditionally use a data-driven approach for training, requiring the generation of a sufficiently large number of simulation results as the training dataset. Alternatively, a physics-informed neural network can be trained by minimizing the PDE residuals or energy potentials. However, this approach does not provide for a general method to easily using existing finite element models. To address these challenges, we developed a hybrid approach that seamlessly bridged a neural network surrogate model with a differentiable finite element domain representation (NNFE). Given its importance in cardiac simulations, we applied this approach to simulations of the hyperelastic mechanical behavior of ventricular myocardium from recent 3D kinematic constitutive model (J Mech Behav Biomed Mater, 2020 doi: 10.1016/j.jmbbm.2019.103508). We utilized cuboidal domain and conducted numerical studies of individual myocardium specimens discretized by a finite element mesh and assigned with experimentally obtained myofiber architectures. Both parameterized Dirichlet and Neumann boundary conditions were studied. We developed a second-order Newton optimization method, instead of using stochastic gradient descent method, to train the neural network efficiently. The resulting trained neural network surrogate model demonstrated excellent agreement with the corresponding 'ground truth' finite element solutions over the entire physiological deformation range. More importantly, the NNFE approach provided a significantly decreased computational time for a range of finite element mesh sizes for online predictions. For example, as the finite element mesh sized increased from 2744 to 175615 elements the NNFE computational time increased from 0.1108 s to 0.1393 s, while the 'ground truth' FE model increased from 4.541 s to 719.9 s. These results suggests that NNFE run times can be significantly reduced compared with the traditional large-deformation based finite element solution methods. The trade off is to train the NNFE off-line within a range of anticipated physiological responses. However, training time would only have to be performed once before any number of application uses. Moreover, since the NNFE is an analytical function its computational performance will be amplified when the corresponding problem becomes more complex.
Collapse
Affiliation(s)
- Wenbo Zhang
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712, USA
| | - David S Li
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712, USA
| | - Tan Bui-Thanh
- Department of Aerospace Engineering and Engineering Mechanics, and Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712, USA
| | - Michael S Sacks
- James T. Willerson Center for Cardiovascular Modeling and Simulation, Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712, USA
- Department of Biomedical Engineering, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712, USA
- Department of Aerospace Engineering and Engineering Mechanics, and Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, 201 East 24th St, Stop C0200, Austin, TX 78712, USA
| |
Collapse
|
15
|
Camman M, Joanne P, Agbulut O, Hélary C. 3D models of dilated cardiomyopathy: Shaping the chemical, physical and topographical properties of biomaterials to mimic the cardiac extracellular matrix. Bioact Mater 2022; 7:275-291. [PMID: 34466733 PMCID: PMC8379361 DOI: 10.1016/j.bioactmat.2021.05.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of dilated cardiomyopathy (DCM), one major cause of heart failure, is characterized by the dilation of the heart but remains poorly understood because of the lack of adequate in vitro models. Current 2D models do not allow for the 3D organotypic organization of cardiomyocytes and do not reproduce the ECM perturbations. In this review, the different strategies to mimic the chemical, physical and topographical properties of the cardiac tissue affected by DCM are presented. The advantages and drawbacks of techniques generating anisotropy required for the cardiomyocytes alignment are discussed. In addition, the different methods creating macroporosity and favoring organotypic organization are compared. Besides, the advances in the induced pluripotent stem cells technology to generate cardiac cells from healthy or DCM patients will be described. Thanks to the biomaterial design, some features of the DCM extracellular matrix such as stiffness, porosity, topography or chemical changes can impact the cardiomyocytes function in vitro and increase their maturation. By mimicking the affected heart, both at the cellular and at the tissue level, 3D models will enable a better understanding of the pathology and favor the discovery of novel therapies.
Collapse
Affiliation(s)
- Marie Camman
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, Inserm ERL U1164, Biological Adaptation and Ageing, 7 quai St-Bernard (case 256), F-75005, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, UMR 7574, Laboratoire de Chimie de la Matière Condensée de Paris, 4 place Jussieu (case 174), F-75005, Paris, France
| |
Collapse
|
16
|
Graham C, Sethu P. Myocardial Fibrosis: Cell Signaling and In Vitro Modeling. CARDIOVASCULAR SIGNALING IN HEALTH AND DISEASE 2022:287-321. [DOI: 10.1007/978-3-031-08309-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
17
|
Shi Y, Sivarajan S, Xiang KM, Kostecki GM, Tung L, Crocker JC, Reich DH. Pervasive cytoquakes in the actomyosin cortex across cell types and substrate stiffness. Integr Biol (Camb) 2021; 13:246-257. [PMID: 34875067 DOI: 10.1093/intbio/zyab017] [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: 04/05/2021] [Revised: 10/25/2021] [Accepted: 11/02/2021] [Indexed: 11/14/2022]
Abstract
The actomyosin cytoskeleton enables cells to resist deformation, crawl, change their shape and sense their surroundings. Despite decades of study, how its molecular constituents can assemble together to form a network with the observed mechanics of cells remains poorly understood. Recently, it has been shown that the actomyosin cortex of quiescent cells can undergo frequent, abrupt reconfigurations and displacements, called cytoquakes. Notably, such fluctuations are not predicted by current physical models of actomyosin networks, and their prevalence across cell types and mechanical environments has not previously been studied. Using micropost array detectors, we have performed high-resolution measurements of the dynamic mechanical fluctuations of cells' actomyosin cortex and stress fiber networks. This reveals cortical dynamics dominated by cytoquakes-intermittent events with a fat-tailed distribution of displacements, sometimes spanning microposts separated by 4 μm, in all cell types studied. These included 3T3 fibroblasts, where cytoquakes persisted over substrate stiffnesses spanning the tissue-relevant range of 4.3 kPa-17 kPa, and primary neonatal rat cardiac fibroblasts and myofibroblasts, human embryonic kidney cells and human bone osteosarcoma epithelial (U2OS) cells, where cytoquakes were observed on substrates in the same stiffness range. Overall, these findings suggest that the cortex self-organizes into a marginally stable mechanical state whose physics may contribute to cell mechanical properties, active behavior and mechanosensing.
Collapse
Affiliation(s)
- Yu Shi
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Shankar Sivarajan
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Katherine M Xiang
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Geran M Kostecki
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - John C Crocker
- Department of Chemical and Biomolecular Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel H Reich
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
18
|
Han B, Trew ML, Zgierski-Johnston CM. Cardiac Conduction Velocity, Remodeling and Arrhythmogenesis. Cells 2021; 10:cells10112923. [PMID: 34831145 PMCID: PMC8616078 DOI: 10.3390/cells10112923] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/14/2021] [Accepted: 10/22/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac electrophysiological disorders, in particular arrhythmias, are a key cause of morbidity and mortality throughout the world. There are two basic requirements for arrhythmogenesis: an underlying substrate and a trigger. Altered conduction velocity (CV) provides a key substrate for arrhythmogenesis, with slowed CV increasing the probability of re-entrant arrhythmias by reducing the length scale over which re-entry can occur. In this review, we examine methods to measure cardiac CV in vivo and ex vivo, discuss underlying determinants of CV, and address how pathological variations alter CV, potentially increasing arrhythmogenic risk. Finally, we will highlight future directions both for methodologies to measure CV and for possible treatments to restore normal CV.
Collapse
Affiliation(s)
- Bo Han
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, 79110 Freiburg im Breisgau, Germany;
- Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, 79104 Freiburg im Breisgau, Germany
- Department of Cardiovascular Surgery, The Fourth People’s Hospital of Jinan, 250031 Jinan, China
| | - Mark L. Trew
- Auckland Bioengineering Institute, University of Auckland, Auckland 1010, New Zealand;
| | - Callum M. Zgierski-Johnston
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, 79110 Freiburg im Breisgau, Germany;
- Faculty of Medicine, University of Freiburg, 79110 Freiburg im Breisgau, Germany
- Correspondence:
| |
Collapse
|
19
|
Napiwocki B, Stempien A, Lang D, Kruepke R, Kim G, Zhang J, Eckhardt L, Glukhov A, Kamp T, Crone W. Micropattern platform promotes extracellular matrix remodeling by human PSC-derived cardiac fibroblasts and enhances contractility of co-cultured cardiomyocytes. Physiol Rep 2021; 9:e15045. [PMID: 34617673 PMCID: PMC8496154 DOI: 10.14814/phy2.15045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 08/20/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023] Open
Abstract
In native heart tissue, cardiac fibroblasts provide the structural framework of extracellular matrix (ECM) while also influencing the electrical and mechanical properties of cardiomyocytes. Recent advances in the field of stem cell differentiation have led to the availability of human pluripotent stem cell-derived cardiac fibroblasts (iPSC-CFs) in addition to cardiomyocytes (iPSC-CMs). Here we use a novel 2D in vitro micropatterned platform that provides control over ECM geometry and substrate stiffness. When cultured alone on soft micropatterned substrates, iPSC-CFs are confined to the micropatterned features and remodel the ECM into anisotropic fibers. Similar remodeling and ECM production occurs when cultured with iPSC-CMs in a co-culture model. In addition to modifications in the ECM, our results show that iPSC-CFs influence iPSC-CM function with accelerated Ca2+ transient rise-up time and greater contractile strains in the co-culture conditions compared to when iPSC-CMs are cultured alone. These combined observations highlight the important role cardiac fibroblasts play in vivo and the need for co-culture models like the one presented here to provide more representative in vitro cardiac constructs.
Collapse
Affiliation(s)
- B.N. Napiwocki
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Institute for DiscoveryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - A. Stempien
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Institute for DiscoveryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - D. Lang
- Department of MedicineDivision of Cardiovascular MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - R.A. Kruepke
- Engineering Mechanics ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - G. Kim
- Department of MedicineDivision of Cardiovascular MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - J. Zhang
- Department of MedicineDivision of Cardiovascular MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - L.L. Eckhardt
- Department of MedicineDivision of Cardiovascular MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - A.V. Glukhov
- Department of MedicineDivision of Cardiovascular MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - T.J. Kamp
- Department of MedicineDivision of Cardiovascular MedicineUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Cell and Regenerative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - W.C. Crone
- Department of Biomedical EngineeringUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Wisconsin Institute for DiscoveryUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Engineering Mechanics ProgramUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
- Department of Engineering PhysicsUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| |
Collapse
|
20
|
Liu F, Wu H, Yang X, Dong Y, Huang G, Genin GM, Lu TJ, Xu F. A new model of myofibroblast-cardiomyocyte interactions and their differences across species. Biophys J 2021; 120:3764-3775. [PMID: 34280368 DOI: 10.1016/j.bpj.2021.06.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/02/2021] [Accepted: 06/28/2021] [Indexed: 11/18/2022] Open
Abstract
Although coupling between cardiomyocytes and myofibroblasts is well known to affect the physiology and pathophysiology of cardiac tissues across species, relating these observations to humans is challenging because the effect of this coupling varies across species and because the sources of these effects are not known. To identify the sources of cross-species variation, we built upon previous mathematical models of myofibroblast electrophysiology and developed a mechanoelectrical model of cardiomyocyte-myofibroblast interactions as mediated by electrotonic coupling and transforming growth factor-β1. The model, as verified by experimental data from the literature, predicted that both electrotonic coupling and transforming growth factor-β1 interaction between myocytes and myofibroblast prolonged action potential in rat myocytes but shortened action potential in human myocytes. This variance could be explained by differences in the transient outward K+ current associated with differential Kv4.2 gene expression across species. Results are useful for efforts to extrapolate the results of animal models to the predicted effects in humans and point to potential therapeutic targets for fibrotic cardiomyopathy.
Collapse
Affiliation(s)
- Fusheng Liu
- State Key Laboratory for Strength and Vibration of Mechanical Structures, Xi'an, P.R. China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Hou Wu
- State Key Laboratory for Strength and Vibration of Mechanical Structures, Xi'an, P.R. China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an, P.R. China
| | - Xiaoyu Yang
- State Key Laboratory for Strength and Vibration of Mechanical Structures, Xi'an, P.R. China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an, P.R. China
| | - Yuqin Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China
| | - Guoyou Huang
- Department of Engineering Mechanics, School of Civil Engineering, Wuhan University, Wuhan, P.R. China
| | - Guy M Genin
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China; Department of Mechanical Engineering & Materials Science, St. Louis, Missouri; NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis, Missouri
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control of Mechanical Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, P.R. China.
| | - Feng Xu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an, P.R. China; Bioinspired Engineering and Biomechanics Center, Xi'an Jiaotong University, Xi'an, P.R. China.
| |
Collapse
|
21
|
Boyle PM, Yu J, Klimas A, Williams JC, Trayanova NA, Entcheva E. OptoGap is an optogenetics-enabled assay for quantification of cell-cell coupling in multicellular cardiac tissue. Sci Rep 2021; 11:9310. [PMID: 33927252 PMCID: PMC8085001 DOI: 10.1038/s41598-021-88573-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/31/2021] [Indexed: 12/23/2022] Open
Abstract
Intercellular electrical coupling is an essential means of communication between cells. It is important to obtain quantitative knowledge of such coupling between cardiomyocytes and non-excitable cells when, for example, pathological electrical coupling between myofibroblasts and cardiomyocytes yields increased arrhythmia risk or during the integration of donor (e.g., cardiac progenitor) cells with native cardiomyocytes in cell-therapy approaches. Currently, there is no direct method for assessing heterocellular coupling within multicellular tissue. Here we demonstrate experimentally and computationally a new contactless assay for electrical coupling, OptoGap, based on selective illumination of inexcitable cells that express optogenetic actuators and optical sensing of the response of coupled excitable cells (e.g., cardiomyocytes) that are light-insensitive. Cell-cell coupling is quantified by the energy required to elicit an action potential via junctional current from the light-stimulated cell(s). The proposed technique is experimentally validated against the standard indirect approach, GapFRAP, using light-sensitive cardiac fibroblasts and non-transformed cardiomyocytes in a two-dimensional setting. Its potential applicability to the complex three-dimensional setting of the native heart is corroborated by computational modelling and proper calibration. Lastly, the sensitivity of OptoGap to intrinsic cell-scale excitability is robustly characterized via computational analysis.
Collapse
Affiliation(s)
- Patrick M Boyle
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Jinzhu Yu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Aleksandra Klimas
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
- Department of Biomedical Engineering, George Washington University, 800 22nd Street NW, Suite 5000, Washington, DC, 20052, USA
| | - John C Williams
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Natalia A Trayanova
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, MD, USA
- Alliance for Cardiovascular Diagnostic and Treatment Innovation, Johns Hopkins University, Baltimore, MD, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
- Department of Biomedical Engineering, George Washington University, 800 22nd Street NW, Suite 5000, Washington, DC, 20052, USA.
| |
Collapse
|
22
|
Jorba I, Mostert D, Hermans LH, van der Pol A, Kurniawan NA, Bouten CV. In Vitro Methods to Model Cardiac Mechanobiology in Health and Disease. Tissue Eng Part C Methods 2021; 27:139-151. [PMID: 33514281 PMCID: PMC7984657 DOI: 10.1089/ten.tec.2020.0342] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022] Open
Abstract
In vitro cardiac modeling has taken great strides in the past decade. While most cell and engineered tissue models have focused on cell and tissue contractile function as readouts, mechanobiological cues from the cell environment that affect this function, such as matrix stiffness or organization, are less well explored. In this study, we review two-dimensional (2D) and three-dimensional (3D) models of cardiac function that allow for systematic manipulation or precise control of mechanobiological cues under simulated (patho)physiological conditions while acquiring multiple readouts of cell and tissue function. We summarize the cell types used in these models and highlight the importance of linking 2D and 3D models to address the multiscale organization and mechanical behavior. Finally, we provide directions on how to advance in vitro modeling for cardiac mechanobiology using next generation hydrogels that mimic mechanical and structural environmental features at different length scales and diseased cell types, along with the development of new tissue fabrication and readout techniques. Impact statement Understanding the impact of mechanobiology in cardiac (patho)physiology is essential for developing effective tissue regeneration and drug discovery strategies and requires detailed cause-effect studies. The development of three-dimensional in vitro models allows for such studies with high experimental control, while integrating knowledge from complementary cell culture models and in vivo studies for this purpose. Complemented by the use of human-induced pluripotent stem cells, with or without predisposed genetic diseases, these in vitro models will offer promising outlooks to delineate the impact of mechanobiological cues on human cardiac (patho)physiology in a dish.
Collapse
Affiliation(s)
- Ignasi Jorba
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Dylan Mostert
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Leon H.L. Hermans
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Atze van der Pol
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| | - Carlijn V.C. Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven, The Netherlands
| |
Collapse
|
23
|
Kostecki GM, Shi Y, Chen CS, Reich DH, Entcheva E, Tung L. Optogenetic current in myofibroblasts acutely alters electrophysiology and conduction of co-cultured cardiomyocytes. Sci Rep 2021; 11:4430. [PMID: 33627695 PMCID: PMC7904933 DOI: 10.1038/s41598-021-83398-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/27/2021] [Indexed: 01/31/2023] Open
Abstract
Interactions between cardiac myofibroblasts and myocytes may slow conduction and generate spontaneous beating in fibrosis, increasing the chance of life-threatening arrhythmia. While co-culture studies have shown that myofibroblasts can affect cardiomyocyte electrophysiology in vitro, the extent of myofibroblast-myocyte electrical conductance in a syncytium is unknown. In this neonatal rat study, cardiac myofibroblasts were transduced with Channelrhodopsin-2, which allowed acute and selective increase of myofibroblast current, and plated on top of cardiomyocytes. Optical mapping revealed significantly decreased conduction velocity (- 27 ± 6%, p < 10-3), upstroke rate (- 13 ± 4%, p = 0.002), and action potential duration (- 14 ± 7%, p = 0.004) in co-cultures when 0.017 mW/mm2 light was applied, as well as focal spontaneous beating in 6/7 samples and a decreased cycle length (- 36 ± 18%, p = 0.002) at 0.057 mW/mm2 light. In silico modeling of the experiments reproduced the experimental findings and suggested the light levels used in experiments produced excess current similar in magnitude to endogenous myofibroblast current. Fitting the model to experimental data predicted a tissue-level electrical conductance across the 3-D interface between myofibroblasts and cardiomyocytes of ~ 5 nS/cardiomyocyte, and showed how increased myofibroblast-myocyte conductance, increased myofibroblast/myocyte capacitance ratio, and increased myofibroblast current, which occur in fibrosis, can work in tandem to produce pro-arrhythmic increases in conduction and spontaneous beating.
Collapse
Affiliation(s)
- Geran M Kostecki
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave., Baltimore, MD, 21205, USA
| | - Yu Shi
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Daniel H Reich
- Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Leslie Tung
- Department of Biomedical Engineering, Johns Hopkins University, 720 Rutland Ave., Baltimore, MD, 21205, USA.
| |
Collapse
|
24
|
Hall C, Gehmlich K, Denning C, Pavlovic D. Complex Relationship Between Cardiac Fibroblasts and Cardiomyocytes in Health and Disease. J Am Heart Assoc 2021; 10:e019338. [PMID: 33586463 PMCID: PMC8174279 DOI: 10.1161/jaha.120.019338] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiac fibroblasts are the primary cell type responsible for deposition of extracellular matrix in the heart, providing support to the contracting myocardium and contributing to a myriad of physiological signaling processes. Despite the importance of fibrosis in processes of wound healing, excessive fibroblast proliferation and activation can lead to pathological remodeling, driving heart failure and the onset of arrhythmias. Our understanding of the mechanisms driving the cardiac fibroblast activation and proliferation is expanding, and evidence for their direct and indirect effects on cardiac myocyte function is accumulating. In this review, we focus on the importance of the fibroblast-to-myofibroblast transition and the cross talk of cardiac fibroblasts with cardiac myocytes. We also consider the current use of models used to explore these questions.
Collapse
Affiliation(s)
- Caitlin Hall
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom.,Division of Cardiovascular Medicine Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford University of Oxford United Kingdom
| | - Chris Denning
- Biodiscovery Institute University of Nottingham United Kingdom
| | - Davor Pavlovic
- Institute of Cardiovascular Sciences University of Birmingham United Kingdom
| |
Collapse
|
25
|
Ho HC, Hsu YH, Jhang JF, Jiang YH, Kuo HC. Ultrastructural changes in the underactive bladder. Tzu Chi Med J 2020; 33:345-349. [PMID: 34760629 PMCID: PMC8532586 DOI: 10.4103/tcmj.tcmj_153_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/21/2020] [Accepted: 08/03/2020] [Indexed: 11/04/2022] Open
Abstract
Underactive bladder (UAB) is a symptom complex suggestive of detrusor underactivity (DU). Although it implies a primary dysfunction of the detrusor muscle, many other conditions such as advanced age, neurogenic factors, and bladder outlet obstruction also lead to UAB. The current understanding of the pathophysiology directly leading to UAB is limited. We believe that by identifying the morphological changes associated with UAB might shed light on this. Therefore, we searched literature with keywords of electron microscopy, ultrastructure, UAB, and DU to review current ultrastructural evidence concerning UAB.
Collapse
Affiliation(s)
- Han-Chen Ho
- Department of Anatomy, Tzu Chi University, Hualien, Taiwan
| | - Yung-Hsiang Hsu
- Department of Pathology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and Tzu Chi University, Hualien, Taiwan
| | - Jia-Fong Jhang
- Department of Urology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and Tzu Chi University, Hualien, Taiwan
| | - Yuan-Hong Jiang
- Department of Urology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and Tzu Chi University, Hualien, Taiwan
| | - Hann-Chorng Kuo
- Department of Urology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, and Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
26
|
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: 0.2] [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
|
27
|
Giacomelli E, Meraviglia V, Campostrini G, Cochrane A, Cao X, van Helden RWJ, Krotenberg Garcia A, Mircea M, Kostidis S, Davis RP, van Meer BJ, Jost CR, Koster AJ, Mei H, Míguez DG, Mulder AA, Ledesma-Terrón M, Pompilio G, Sala L, Salvatori DCF, Slieker RC, Sommariva E, de Vries AAF, Giera M, Semrau S, Tertoolen LGJ, Orlova VV, Bellin M, Mummery CL. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease. Cell Stem Cell 2020; 26:862-879.e11. [PMID: 32459996 PMCID: PMC7284308 DOI: 10.1016/j.stem.2020.05.004] [Citation(s) in RCA: 361] [Impact Index Per Article: 72.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/05/2020] [Accepted: 05/01/2020] [Indexed: 12/12/2022]
Abstract
Cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) are functionally immature, but this is improved by incorporation into engineered tissues or forced contraction. Here, we showed that tri-cellular combinations of hiPSC-derived CMs, cardiac fibroblasts (CFs), and cardiac endothelial cells also enhance maturation in easily constructed, scaffold-free, three-dimensional microtissues (MTs). hiPSC-CMs in MTs with CFs showed improved sarcomeric structures with T-tubules, enhanced contractility, and mitochondrial respiration and were electrophysiologically more mature than MTs without CFs. Interactions mediating maturation included coupling between hiPSC-CMs and CFs through connexin 43 (CX43) gap junctions and increased intracellular cyclic AMP (cAMP). Scaled production of thousands of hiPSC-MTs was highly reproducible across lines and differentiated cell batches. MTs containing healthy-control hiPSC-CMs but hiPSC-CFs from patients with arrhythmogenic cardiomyopathy strikingly recapitulated features of the disease. Our MT model is thus a simple and versatile platform for modeling multicellular cardiac diseases that will facilitate industry and academic engagement in high-throughput molecular screening. Cardiac fibroblasts and endothelial cells induce hiPSC-cardiomyocyte maturation CX43 gap junctions form between cardiac fibroblasts and cardiomyocytes cAMP-pathway activation contributes to hiPSC-cardiomyocyte maturation Patient-derived hiPSC-cardiac fibroblasts cause arrhythmia in microtissues
Collapse
Affiliation(s)
- Elisa Giacomelli
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Giulia Campostrini
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Amy Cochrane
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Xu Cao
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Ruben W J van Helden
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Ana Krotenberg Garcia
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Maria Mircea
- Leiden Institute of Physics, Leiden University, 2333 Leiden, the Netherlands
| | - Sarantos Kostidis
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Richard P Davis
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Berend J van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Carolina R Jost
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Abraham J Koster
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - David G Míguez
- Centro de Biologia Molecular Severo Ochoa, Departamento de Física de la Materia Condensada, Instituto Nicolas Cabrera and Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Aat A Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Mario Ledesma-Terrón
- Centro de Biologia Molecular Severo Ochoa, Departamento de Física de la Materia Condensada, Instituto Nicolas Cabrera and Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Giulio Pompilio
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milan, Italy
| | - Luca Sala
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Daniela C F Salvatori
- Central Laboratory Animal Facility, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Epidemiology and Biostatistics, Amsterdam Public Health Institute, VU University Medical Center, 1007 Amsterdam, the Netherlands
| | - Elena Sommariva
- Vascular Biology and Regenerative Medicine Unit, Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy
| | - Antoine A F de Vries
- Department of Cardiology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Stefan Semrau
- Leiden Institute of Physics, Leiden University, 2333 Leiden, the Netherlands
| | - Leon G J Tertoolen
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands
| | - Valeria V Orlova
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands.
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Biology, University of Padua, 35121 Padua, Italy; Veneto Institute of Molecular Medicine, 35129 Padua, Italy.
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333 Leiden, the Netherlands; Department of Applied Stem Cell Technologies, University of Twente, 7500 Enschede, the Netherlands.
| |
Collapse
|
28
|
Kappadan V, Telele S, Uzelac I, Fenton F, Parlitz U, Luther S, Christoph J. High-Resolution Optical Measurement of Cardiac Restitution, Contraction, and Fibrillation Dynamics in Beating vs. Blebbistatin-Uncoupled Isolated Rabbit Hearts. Front Physiol 2020; 11:464. [PMID: 32528304 PMCID: PMC7264405 DOI: 10.3389/fphys.2020.00464] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/16/2020] [Indexed: 11/13/2022] Open
Abstract
Optical mapping is a high-resolution fluorescence imaging technique, that uses voltage- or calcium-sensitive dyes to visualize electrical excitation waves on the heart surface. However, optical mapping is very susceptible to the motion of cardiac tissue, which results in so-called motion artifacts in the fluorescence signal. To avoid motion artifacts, contractions of the heart muscle are typically suppressed using pharmacological excitation-contraction uncoupling agents, such as Blebbistatin. The use of pharmacological agents, however, may influence cardiac electrophysiology. Recently, it has been shown that numerical motion tracking can significantly reduce motion-related artifacts in optical mapping, enabling the simultaneous optical measurement of cardiac electrophysiology and mechanics. Here, we combine ratiometric optical mapping with numerical motion tracking to further enhance the robustness and accuracy of these measurements. We evaluate the method's performance by imaging and comparing cardiac restitution and ventricular fibrillation (VF) dynamics in contracting, non-working vs. Blebbistatin-arrested Langendorff-perfused rabbit hearts (N = 10). We found action potential durations (APD) to be, on average, 25 ± 5% shorter in contracting hearts compared to hearts uncoupled with Blebbistatin. The relative shortening of the APD was found to be larger at higher frequencies. VF was found to be significantly accelerated in contracting hearts, i.e., 9 ± 2Hz with Blebbistatin and 15 ± 4Hz without Blebbistatin, and maintained a broader frequency spectrum. In contracting hearts, the average number of phase singularities was NPS = 11 ± 4 compared to NPS = 6 ± 3 with Blebbistatin during VF on the anterior ventricular surface. VF inducibility was reduced with Blebbistatin. We found the effect of Blebbistatin to be concentration-dependent and reversible by washout. Aside from the electrophysiological characterization, we also measured and analyzed cardiac motion. Our findings may have implications for the interpretation of optical mapping data, and highlight that physiological conditions, such as oxygenation and metabolic demand, must be carefully considered in ex vivo imaging experiments.
Collapse
Affiliation(s)
- Vineesh Kappadan
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Saba Telele
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Partnersite Göttingen, Göttingen, Germany
| | - Ilija Uzelac
- School of Physics, Georgia Institute of Technology, Atlanta, GA, United States
| | - Flavio Fenton
- School of Physics, Georgia Institute of Technology, Atlanta, GA, United States
| | - Ulrich Parlitz
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Partnersite Göttingen, Göttingen, Germany.,Institute for the Dynamics of Complex Systems, University of Göttingen, Göttingen, Germany
| | - Stefan Luther
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Partnersite Göttingen, Göttingen, Germany.,Department of Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Jan Christoph
- Research Group Biomedical Physics, Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK e.V.), Partnersite Göttingen, Göttingen, Germany.,Department of Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
29
|
Quinn TA, Kohl P. Cardiac Mechano-Electric Coupling: Acute Effects of Mechanical Stimulation on Heart Rate and Rhythm. Physiol Rev 2020; 101:37-92. [PMID: 32380895 DOI: 10.1152/physrev.00036.2019] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The heart is vital for biological function in almost all chordates, including humans. It beats continually throughout our life, supplying the body with oxygen and nutrients while removing waste products. If it stops, so does life. The heartbeat involves precise coordination of the activity of billions of individual cells, as well as their swift and well-coordinated adaption to changes in physiological demand. Much of the vital control of cardiac function occurs at the level of individual cardiac muscle cells, including acute beat-by-beat feedback from the local mechanical environment to electrical activity (as opposed to longer term changes in gene expression and functional or structural remodeling). This process is known as mechano-electric coupling (MEC). In the current review, we present evidence for, and implications of, MEC in health and disease in human; summarize our understanding of MEC effects gained from whole animal, organ, tissue, and cell studies; identify potential molecular mediators of MEC responses; and demonstrate the power of computational modeling in developing a more comprehensive understanding of ‟what makes the heart tick.ˮ.
Collapse
Affiliation(s)
- T Alexander Quinn
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Peter Kohl
- Department of Physiology and Biophysics and School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada; Institute for Experimental Cardiovascular Medicine, University Heart Centre Freiburg/Bad Krozingen, Medical Faculty of the University of Freiburg, Freiburg, Germany; and CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
30
|
Davidson MD, Burdick JA, Wells RG. Engineered Biomaterial Platforms to Study Fibrosis. Adv Healthc Mater 2020; 9:e1901682. [PMID: 32181987 PMCID: PMC7274888 DOI: 10.1002/adhm.201901682] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/13/2022]
Abstract
Many pathologic conditions lead to the development of tissue scarring and fibrosis, which are characterized by the accumulation of abnormal extracellular matrix (ECM) and changes in tissue mechanical properties. Cells within fibrotic tissues are exposed to dynamic microenvironments that may promote or prolong fibrosis, which makes it difficult to treat. Biomaterials have proved indispensable to better understand how cells sense their extracellular environment and are now being employed to study fibrosis in many tissues. As mechanical testing of tissues becomes more routine and biomaterial tools become more advanced, the impact of biophysical factors in fibrosis are beginning to be understood. Herein, fibrosis from a materials perspective is reviewed, including the role and mechanical properties of ECM components, the spatiotemporal mechanical changes that occur during fibrosis, current biomaterial systems to study fibrosis, and emerging biomaterial systems and tools that can further the understanding of fibrosis initiation and progression. This review concludes by highlighting considerations in promoting wide-spread use of biomaterials for fibrosis investigations and by suggesting future in vivo studies that it is hoped will inspire the development of even more advanced biomaterial systems.
Collapse
Affiliation(s)
- Matthew D Davidson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rebecca G Wells
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
31
|
Connexin43 expression in bone marrow derived cells contributes to the electrophysiological properties of cardiac scar tissue. Sci Rep 2020; 10:2617. [PMID: 32054938 PMCID: PMC7018966 DOI: 10.1038/s41598-020-59449-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 01/29/2020] [Indexed: 11/26/2022] Open
Abstract
Cardiac pathologies associated with arrhythmic activity are often accompanied by inflammation. The contribution of inflammatory cells to the electrophysiological properties of injured myocardium is unknown. Myocardial scar cell types and intercellular contacts were analyzed using a three-dimensional reconstruction from serial blockface scanning electron microscopy data. Three distinct cell populations were identified: inflammatory, fibroblastic and endocardial cells. While individual fibroblastic cells interface with a greater number of cells, inflammatory cells have the largest contact area suggesting a role in establishing intercellular electrical connections in scar tissue. Optical mapping was used to study the electrophysiological properties of scars in fetal liver chimeric mice generated using connexin43 knockout donors (bmpKO). Voltage changes were elicited in response to applied current pulses. Isopotential maps showed a steeper pattern of decay with distance from the electrode in scars compared with uninjured regions, suggesting reduced electrical coupling. The tissue decay constant, defined as the distance voltage reaches 37% of the amplitude at the edge of the scar, was 0.48 ± 0.04 mm (n = 11) in the scar of the bmpCTL group and decreased 37.5% in the bmpKO group (n = 10). Together these data demonstrate inflammatory cells significantly contribute to scar electrophysiology through coupling mediated at least partially by connexin43 expression.
Collapse
|
32
|
Scholz B, Schulte JS, Hamer S, Himmler K, Pluteanu F, Seidl MD, Stein J, Wardelmann E, Hammer E, Völker U, Müller FU. HDAC (Histone Deacetylase) Inhibitor Valproic Acid Attenuates Atrial Remodeling and Delays the Onset of Atrial Fibrillation in Mice. Circ Arrhythm Electrophysiol 2019; 12:e007071. [PMID: 30879335 PMCID: PMC6426346 DOI: 10.1161/circep.118.007071] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Background: A structural, electrical and metabolic atrial remodeling is central in the development of atrial fibrillation (AF) contributing to its initiation and perpetuation. In the heart, HDACs (histone deacetylases) control remodeling associated processes like hypertrophy, fibrosis, and energy metabolism. Here, we analyzed, whether the HDAC class I/IIa inhibitor valproic acid (VPA) is able to attenuate atrial remodeling in CREM-IbΔC-X (cAMP responsive element modulator isoform IbΔC-X) transgenic mice, a mouse model of extensive atrial remodeling with age-dependent progression from spontaneous atrial ectopy to paroxysmal and finally long-lasting AF. Methods: VPA was administered for 7 or 25 weeks to transgenic and control mice. Atria were analyzed macroscopically and using widefield and electron microscopy. Action potentials were recorded from atrial cardiomyocytes using patch-clamp technique. ECG recordings documented the onset of AF. A proteome analysis with consecutive pathway mapping identified VPA-mediated proteomic changes and related pathways. Results: VPA attenuated many components of atrial remodeling that are present in transgenic mice, animal AF models, and human AF. VPA significantly (P<0.05) reduced atrial dilatation, cardiomyocyte enlargement, atrial fibrosis, and the disorganization of myocyte’s ultrastructure. It significantly reduced the occurrence of atrial thrombi, reversed action potential alterations, and finally delayed the onset of AF by 4 to 8 weeks. Increased histone H4-acetylation in atria from VPA-treated transgenic mice verified effective in vivo HDAC inhibition. Cardiomyocyte-specific genetic inactivation of HDAC2 in transgenic mice attenuated the ultrastructural disorganization of myocytes comparable to VPA. Finally, VPA restrained dysregulation of proteins in transgenic mice that are involved in a multitude of AF relevant pathways like oxidative phosphorylation or RhoA (Ras homolog gene family, member A) signaling and disease functions like cardiac fibrosis and apoptosis of muscle cells. Conclusions: Our results suggest that VPA, clinically available, well-tolerated, and prescribed to many patients for years, has the therapeutic potential to delay the development of atrial remodeling and the onset of AF in patients at risk.
Collapse
Affiliation(s)
- Beatrix Scholz
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Jan Sebastian Schulte
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Sabine Hamer
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Kirsten Himmler
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Florentina Pluteanu
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Matthias Dodo Seidl
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Juliane Stein
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Germany (E.W.)
| | - Elke Hammer
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Uwe Völker
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| |
Collapse
|
33
|
Bae H, Kim T, Lim I. Effects of nitric oxide on apoptosis and voltage-gated calcium channels in human cardiac myofibroblasts. Clin Exp Pharmacol Physiol 2019; 47:16-26. [PMID: 31519057 DOI: 10.1111/1440-1681.13178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/30/2019] [Accepted: 09/10/2019] [Indexed: 02/05/2023]
Abstract
We characterised the voltage-gated Ca2+ channels (VGCCs) in human cardiac fibroblasts (HCFs) and myofibroblasts (HCMFs) and investigated the effects of nitric oxide (NO) on apoptosis and on these channels. Western blotting and immunofluorescence analyses show that α-smooth muscle actin (a myofibroblast marker) was markedly expressed in passage (P) 12-15 but not in P4 HCF cells, whereas calponin (a fibroblast marker) was expressed only in P4 cells. CaV 1.2 (L-type) and CaV 3.3 (T-type) of VGCCs were highly expressed in P12-15 cells, but only weak CaV 2.3 (R-type) expression was identified in P4 cells using reverse transcription-polymerase chain reaction analysis. S-Nitroso-N-acetylpenicillamine (SNAP, an NO donor) decreased cell viability of HCMFs in a dose-dependent manner and induced apoptotic changes, and nifedipine (an L-type Ca2+ channel blocker) prevented apoptosis as shown with immunofluorescence staining and flow cytometry. Whole-cell mode patch-clamp recordings demonstrate the presence of L-type Ca2+ (IC a,L ) and T-type Ca2+ (IC a,T ) currents in HCMFs. SNAP inhibited IC a,L of HCMFs, but pre-treatment with ODQ (a guanylate cyclase inhibitor) or KT5823 (a PKG inhibitor) prevented it. Pre-treating cells with KT5720 (a PKA inhibitor) or SQ22536 (an adenylate cyclase inhibitor) blocked SNAP-induced inhibition of IC a,L . 8-Bromo-cyclic GMP or 8-bromo-cyclic AMP also inhibited IC a,L . However, pre-treatment with N-ethylmaleimide (a thiol-alkylating reagent) did not block the SNAP effect, nor did DL-dithiothreitol (a reducing agent) reverse it. These data suggest that high concentrations of NO injure HCMFs and inhibit IC a,L through the PKG and PKA signalling pathways but not through the S-nitrosylation pathway.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
34
|
Wang L, Ripplinger CM. Putting the pieces together using in vivo optical mapping. Cardiovasc Res 2019; 115:1574-1575. [PMID: 30924872 DOI: 10.1093/cvr/cvz089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Lianguo Wang
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA 95616, USA
| | - Crystal M Ripplinger
- Department of Pharmacology, UC Davis School of Medicine, 2419B Tupper Hall, One Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
35
|
Schultz F, Swiatlowska P, Alvarez-Laviada A, Sanchez-Alonso JL, Song Q, de Vries AAF, Pijnappels DA, Ongstad E, Braga VMM, Entcheva E, Gourdie RG, Miragoli M, Gorelik J. Cardiomyocyte-myofibroblast contact dynamism is modulated by connexin-43. FASEB J 2019; 33:10453-10468. [PMID: 31253057 PMCID: PMC6704460 DOI: 10.1096/fj.201802740rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Healthy cardiomyocytes are electrically coupled at the intercalated discs by gap junctions. In infarcted hearts, adverse gap-junctional remodeling occurs in the border zone, where cardiomyocytes are chemically and electrically influenced by myofibroblasts. The physical movement of these contacts remains unquantified. Using scanning ion conductance microscopy, we show that intercellular contacts between cardiomyocytes and myofibroblasts are highly dynamic, mainly owing to the edge dynamics (lamellipodia) of the myofibroblasts. Decreasing the amount of functional connexin-43 (Cx43) at the membrane through Cx43 silencing, suppression of Cx43 trafficking, or hypoxia-induced Cx43 internalization attenuates heterocellular contact dynamism. However, we found decreased dynamism and stabilized membrane contacts when cellular coupling was strengthened using 4-phenylbutyrate (4PB). Fluorescent-dye transfer between cells showed that the extent of functional coupling between the 2 cell types correlated with contact dynamism. Intercellular calcein transfer from myofibroblasts to cardiomyocytes is reduced after myofibroblast-specific Cx43 down-regulation. Conversely, 4PB-treated myofibroblasts increased their functional coupling to cardiomyocytes. Consistent with lamellipodia-mediated contacts, latrunculin-B decreases dynamism, lowers physical communication between heterocellular pairs, and reduces Cx43 intensity in contact regions. Our data show that heterocellular cardiomyocyte-myofibroblast contacts exhibit high dynamism. Therefore, Cx43 is a potential target for prevention of aberrant cardiomyocyte coupling and myofibroblast proliferation in the infarct border zone.-Schultz, F., Swiatlowska, P., Alvarez-Laviada, A., Sanchez-Alonso, J. L., Song, Q., de Vries, A. A. F., Pijnappels, D. A., Ongstad, E., Braga, V. M. M., Entcheva, E., Gourdie, R. G., Miragoli, M., Gorelik, J. Cardiomyocyte-myofibroblast contact dynamism is modulated by connexin-43.
Collapse
Affiliation(s)
- Francisca Schultz
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pamela Swiatlowska
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | | | - Qianqian Song
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Daniël A. Pijnappels
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emily Ongstad
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, Virginia, USA
| | - Vania M. M. Braga
- Department of Respiratory Sciences, Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Emilia Entcheva
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Robert G. Gourdie
- Center for Heart and Regenerative Medicine, Virginia Tech Carilion Research Institute, Roanoke, Virginia, USA
| | - Michele Miragoli
- Humanitas Clinical and Research Center, Milan, Italy;,Department of Medicine and Surgery, University of Parma, Parma, Italy,Correspondence: Department of Medicine and Surgery, University of Parma, Via Gramsci 14, 43124 Parma, Italy. E-mail:
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, London, United Kingdom;,Correspondence: National Heart and Lung Institute, 4th Floor, Imperial Centre for Translational and Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Rd., London W12 0NN, United Kingdom. E-mail:
| |
Collapse
|
36
|
Rubart M, Tao W, Lu XL, Conway SJ, Reuter SP, Lin SF, Soonpaa MH. Electrical coupling between ventricular myocytes and myofibroblasts in the infarcted mouse heart. Cardiovasc Res 2019; 114:389-400. [PMID: 29016731 DOI: 10.1093/cvr/cvx163] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/10/2017] [Indexed: 12/20/2022] Open
Abstract
Aims Recent studies have demonstrated electrotonic coupling between scar tissue and the surrounding myocardium in cryoinjured hearts. However, the electrical dynamics occurring at the myocyte-nonmyocyte interface in the fibrotic heart remain undefined. Here, we sought to develop an assay to interrogate the nonmyocyte cell type contributing to heterocellular coupling and to characterize, on a cellular scale, its voltage response in the infarct border zone of living hearts. Methods and results We used two-photon laser scanning microscopy in conjunction with a voltage-sensitive dye to record transmembrane voltage changes simultaneously from cardiomyocytes and adjoined nonmyocytes in Langendorff-perfused mouse hearts with healing myocardial infarction. Transgenic mice with cardiomyocyte-restricted expression of a green fluorescent reporter protein underwent permanent coronary artery ligation and their hearts were subjected to voltage imaging 7-10 days later. Reporter-negative cells, i.e. nonmyocytes, in the infarct border zone exhibited depolarizing transients at a 1:1 coupling ratio with action potentials recorded simultaneously from adjacent, reporter-positive ventricular myocytes. The electrotonic responses in the nonmyocytes exhibited slower rates of de- and repolarization compared to the action potential waveform of juxtaposed myocytes. Voltage imaging in infarcted hearts expressing a fluorescent reporter specifically in myofibroblasts revealed that the latter were electrically coupled to border zone myocytes. Their voltage transient properties were indistinguishable from those of nonmyocytes in hearts with cardiomyocyte-restricted reporter expression. The density of connexin43 expression at myofibroblast-cardiomyocyte junctions was ∼5% of that in the intercalated disc regions of paired ventricular myocytes in the remote, uninjured myocardium, whereas the ratio of connexin45 to connexin43 expression levels at heterocellular contacts was ∼1%. Conclusion Myofibroblasts contribute to the population of electrically coupled nonmyocytes in the infarct border zone. The slower kinetics of myofibroblast voltage responses may reflect low electrical conductivity across heterocellular junctions, in accordance with the paucity of connexin expression at myofibroblast-cardiomyocyte contacts.
Collapse
Affiliation(s)
- Michael Rubart
- Wells Centre for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202, USA
| | - Wen Tao
- Wells Centre for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202, USA
| | - Xiao-Long Lu
- Wells Centre for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202, USA
| | - Simon J Conway
- Wells Centre for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202, USA
| | - Sean P Reuter
- Wells Centre for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202, USA
| | - Shien-Fong Lin
- Institute of Biomedical Engineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Mark H Soonpaa
- Wells Centre for Pediatric Research, Indiana University School of Medicine, 1044 West Walnut Street, Indianapolis, IN 46202, USA.,Krannert Institute of Cardiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
37
|
Cavallini F, Tarantola M. ECIS based wounding and reorganization of cardiomyocytes and fibroblasts in co-cultures. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2019; 144:116-127. [DOI: 10.1016/j.pbiomolbio.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/22/2018] [Accepted: 06/26/2018] [Indexed: 12/11/2022]
|
38
|
Sayegh MN, Fernandez N, Cho HC. Strength-duration relationship as a tool to prioritize cardiac tissue properties that govern electrical excitability. Am J Physiol Heart Circ Physiol 2019; 317:H13-H25. [PMID: 30925072 PMCID: PMC7199233 DOI: 10.1152/ajpheart.00161.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Engineered cardiac tissue and cardiomyocyte cell cultures offer wide opportunities for improved therapeutic intervention and laboratory heart models. Electrical field excitation is a common intervention in the production of engineered tissue and the investigation of the electrical properties of in vitro cell cultures. In this work, we use strength-duration relationships to investigate systematically factors influencing electrical excitability of two- (2D) and three-dimensional (3D) neonatal rat ventricular myocyte cultures. We find that the strength of the voltage pulse is negatively correlated with the threshold duration, as predicted by the Lapicque-Hill equation, and show that higher pacing frequencies require higher thresholds to capture paced cultures. We also study the impact of properties intrinsic to the 2D and 3D cultures on strength-duration relationships. We show that a smaller culture dimension, perpendicular anisotropic culture orientation with respect to electrical field, higher proportion of added fibroblasts, and TBX18-induced pacemaker reprogramming independently result in higher stimulation thresholds. These properties reflect the characteristics of the well-insulated endogenous pacemaking tissue in the heart (sinoatrial node) and should guide the engineering of biological pacemakers for improved outcomes. NEW & NOTEWORTHY Gaps exist in the availability of in vitro functional assessment tools that can emulate the integration of regenerative cells and tissues to the host myocardium. We use strength-duration relationships of electrically stimulated two- and three-dimensional myocardial constructs to study the effects of pacing frequency, culture dimensions, anisotropic cell alignment, fibroblast content, and pacemaker phenotype on electrical excitability. Our study delivers electrical strength-duration as a quantifiable parameter to evaluate design parameters of engineered cardiac tissue constructs.
Collapse
Affiliation(s)
- Michael N. Sayegh
- 1Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia,2Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| | - Natasha Fernandez
- 1Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Hee Cheol Cho
- 1Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia,2Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia
| |
Collapse
|
39
|
Dilaveris P, Antoniou CK, Manolakou P, Tsiamis E, Gatzoulis K, Tousoulis D. Biomarkers Associated with Atrial Fibrosis and Remodeling. Curr Med Chem 2019; 26:780-802. [PMID: 28925871 DOI: 10.2174/0929867324666170918122502] [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: 07/15/2016] [Revised: 12/16/2016] [Accepted: 12/23/2016] [Indexed: 12/22/2022]
Abstract
Atrial fibrillation is the most common rhythm disturbance encountered in clinical practice. Although often considered as solely arrhythmic in nature, current evidence has established that atrial myopathy constitutes both the substrate and the outcome of atrial fibrillation, thus initiating a vicious, self-perpetuating cycle. This myopathy is triggered by stress-induced (including pressure/volume overload, inflammation, oxidative stress) responses of atrial tissue, which in the long term become maladaptive, and combine elements of both structural, especially fibrosis, and electrical remodeling, with contemporary approaches yielding potentially useful biomarkers of these processes. Biomarker value becomes greater given the fact that they can both predict atrial fibrillation occurrence and treatment outcome. This mini-review will focus on the biomarkers of atrial remodeling (both electrical and structural) and fibrosis that have been validated in human studies, including biochemical, histological and imaging approaches.
Collapse
Affiliation(s)
- Polychronis Dilaveris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Panagiota Manolakou
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Tsiamis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Gatzoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Tousoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
40
|
Molecular Characteristics of Underactive Bladder. CURRENT BLADDER DYSFUNCTION REPORTS 2019. [DOI: 10.1007/s11884-019-00512-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
41
|
Hayoun‐Neeman D, Korover N, Etzion S, Ofir R, Lichtenstein RG, Cohen S. Exploring peptide‐functionalized alginate scaffolds for engineering cardiac tissue from human embryonic stem cell‐derived cardiomyocytes in serum‐free medium. POLYM ADVAN TECHNOL 2019. [DOI: 10.1002/pat.4602] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Dana Hayoun‐Neeman
- Avram and Stella Goren‐Goldstein Department of Biotechnology EngineeringBen‐Gurion University of the Negev Beer Sheva Israel
| | - Nataly Korover
- Avram and Stella Goren‐Goldstein Department of Biotechnology EngineeringBen‐Gurion University of the Negev Beer Sheva Israel
| | - Sharon Etzion
- Regenerative Medicine and Stem Cell (RMSC) Research Center
| | - Rivka Ofir
- Regenerative Medicine and Stem Cell (RMSC) Research Center
| | - Rachel G. Lichtenstein
- Avram and Stella Goren‐Goldstein Department of Biotechnology EngineeringBen‐Gurion University of the Negev Beer Sheva Israel
- Regenerative Medicine and Stem Cell (RMSC) Research Center
| | - Smadar Cohen
- Avram and Stella Goren‐Goldstein Department of Biotechnology EngineeringBen‐Gurion University of the Negev Beer Sheva Israel
- Regenerative Medicine and Stem Cell (RMSC) Research Center
- The Else Katz Institute for Nanoscale Science and TechnologyBen‐Gurion University of the Negev Beer‐Sheva 84105 Israel
| |
Collapse
|
42
|
Liang J, Huang W, Jiang L, Paul C, Li X, Wang Y. Concise Review: Reduction of Adverse Cardiac Scarring Facilitates Pluripotent Stem Cell-Based Therapy for Myocardial Infarction. Stem Cells 2019; 37:844-854. [PMID: 30913336 PMCID: PMC6599570 DOI: 10.1002/stem.3009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/27/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
Pluripotent stem cells (PSCs) are an attractive, reliable source for generating functional cardiomyocytes for regeneration of infarcted heart. However, inefficient cell engraftment into host tissue remains a notable challenge to therapeutic success due to mechanical damage or relatively inhospitable microenvironment. Evidence has shown that excessively formed scar tissues around cell delivery sites present as mechanical and biological barriers that inhibit migration and engraftment of implanted cells. In this review, we focus on the functional responses of stem cells and cardiomyocytes during the process of cardiac fibrosis and scar formation. Survival, migration, contraction, and coupling function of implanted cells may be affected by matrix remodeling, inflammatory factors, altered tissue stiffness, and presence of electroactive myofibroblasts in the fibrotic microenvironment. Although paracrine factors from implanted cells can improve cardiac fibrosis, the transient effect is insufficient for complete repair of an infarcted heart. Furthermore, investigation of interactions between implanted cells and fibroblasts including myofibroblasts helps the identification of new targets to optimize the host substrate environment for facilitating cell engraftment and functional integration. Several antifibrotic approaches, including the use of pharmacological agents, gene therapies, microRNAs, and modified biomaterials, can prevent progression of heart failure and have been developed as adjunct therapies for stem cell-based regeneration. Investigation and optimization of new biomaterials is also required to enhance cell engraftment of engineered cardiac tissue and move PSCs from a laboratory setting into translational medicine.
Collapse
Affiliation(s)
- Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Xiangnan Li
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
43
|
Savoji H, Mohammadi MH, Rafatian N, Toroghi MK, Wang EY, Zhao Y, Korolj A, Ahadian S, Radisic M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019; 198:3-26. [PMID: 30343824 PMCID: PMC6397087 DOI: 10.1016/j.biomaterials.2018.09.036] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/11/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Although investment in drug discovery and development has been sky-rocketing, the number of approved drugs has been declining. Cardiovascular toxicity due to therapeutic drug use claims the highest incidence and severity of adverse drug reactions in late-stage clinical development. Therefore, to address this issue, new, additional, replacement and combinatorial approaches are needed to fill the gap in effective drug discovery and screening. The motivation for developing accurate, predictive models is twofold: first, to study and discover new treatments for cardiac pathologies which are leading in worldwide morbidity and mortality rates; and second, to screen for adverse drug reactions on the heart, a primary risk in drug development. In addition to in vivo animal models, in vitro and in silico models have been recently proposed to mimic the physiological conditions of heart and vasculature. Here, we describe current in vitro, in vivo, and in silico platforms for modelling healthy and pathological cardiac tissues and their advantages and disadvantages for drug screening and discovery applications. We review the pathophysiology and the underlying pathways of different cardiac diseases, as well as the new tools being developed to facilitate their study. We finally suggest a roadmap for employing these non-animal platforms in assessing drug cardiotoxicity and safety.
Collapse
Affiliation(s)
- Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Naimeh Rafatian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Masood Khaksar Toroghi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada
| | - Yimu Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Samad Ahadian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada.
| |
Collapse
|
44
|
AnilKumar S, Allen SC, Tasnim N, Akter T, Park S, Kumar A, Chattopadhyay M, Ito Y, Suggs LJ, Joddar B. The applicability of furfuryl-gelatin as a novel bioink for tissue engineering applications. J Biomed Mater Res B Appl Biomater 2019; 107:314-323. [PMID: 29656592 PMCID: PMC6188846 DOI: 10.1002/jbm.b.34123] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/22/2018] [Accepted: 03/14/2018] [Indexed: 01/21/2023]
Abstract
Three-dimensional bioprinting is an innovative technique in tissue engineering, to create layer-by-layer structures, required for mimicking body tissues. However, synthetic bioinks do not generally possess high printability and biocompatibility at the same time. So, there is an urgent need for naturally derived bioinks that can exhibit such optimized properties. We used furfuryl-gelatin as a novel, visible-light crosslinkable bioink for fabricating cell-laden structures with high viability. Hyaluronic acid was added as a viscosity enhancer and either Rose Bengal or Riboflavin was used as a visible-light crosslinker. Crosslinking was done by exposing the printed structure for 2.5 min to visible light and confirmed using Fourier transform infrared spectroscopy and rheometry. Scanning electron microscopy revealed a highly porous networked structure. Three different cell types were successfully bioprinted within these constructs. Mouse mesenchymal stem cells printed within monolayer and bilayer sheets showed viability, network formation and proliferation (∼5.33 times) within 72 h of culture. C2C12 and STO cells were used to print a double layered structure, which showed evidence of the viability of both cells and heterocellular clusters within the construct. This furfuryl-gelatin based bioink can be used for tissue engineering of complex tissues and help in understanding how cellular crosstalk happens in vivo during normal or diseased pathology. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 107B: 314-323, 2019.
Collapse
Affiliation(s)
- Shweta AnilKumar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas, 79968
| | - Shane C Allen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, 78712
| | - Nishat Tasnim
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas, 79968
| | - Tahmina Akter
- Department of Chemistry, University of Texas at El Paso, El Paso, Texas, 79968
| | - Shinhye Park
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
| | - Alok Kumar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas, 79968
| | - Munmun Chattopadhyay
- Department of Biomedical Sciences, Center of Emphasis in Diabetes and Metabolism, Texas Tech University Health Sciences Center, El Paso, Texas, 79905
| | - Yoshihiro Ito
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
- Emergent Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, Wako, Saitama, Japan
| | - Laura J Suggs
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, Texas, 78712
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), Department of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas, 79968
- Nano Medical Engineering Laboratory, RIKEN, Wako, Saitama, Japan
- Border Biomedical Research Center (BBRC), University of Texas at El Paso, El Paso, Texas, 79968
| |
Collapse
|
45
|
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: 1.9] [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
|
46
|
Perbellini F, Watson SA, Bardi I, Terracciano CM. Heterocellularity and Cellular Cross-Talk in the Cardiovascular System. Front Cardiovasc Med 2018; 5:143. [PMID: 30443550 PMCID: PMC6221907 DOI: 10.3389/fcvm.2018.00143] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/25/2018] [Indexed: 01/08/2023] Open
Abstract
Cellular specialization and interactions with other cell types are the essence of complex multicellular life. The orchestrated function of different cell populations in the heart, in combination with a complex network of intercellular circuits of communication, is essential to maintain a healthy heart and its disruption gives rise to pathological conditions. Over the past few years, the development of new biological research tools has facilitated more accurate identification of the cardiac cell populations and their specific roles. This review aims to provide an overview on the significance and contributions of the various cellular components: cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, pericytes, and inflammatory cells. It also aims to describe their role in cardiac development, physiology and pathology with a particular focus on the importance of heterocellularity and cellular interaction between these different cell types.
Collapse
Affiliation(s)
- Filippo Perbellini
- Division of Cardiovascular Sciences, Myocardial Function, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | | | - Cesare M. Terracciano
- Division of Cardiovascular Sciences, Myocardial Function, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
47
|
Kim P, Chu N, Davis J, Kim DH. Mechanoregulation of Myofibroblast Fate and Cardiac Fibrosis. ADVANCED BIOSYSTEMS 2018; 2:1700172. [PMID: 31406913 PMCID: PMC6690497 DOI: 10.1002/adbi.201700172] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
During myocardial infarction, myocytes die and are replaced by a specialized fibrotic extracellular matrix, otherwise known as scarring. Fibrotic scarring presents a tremendous hemodynamic burden on the heart, as it creates a stiff substrate, which resists diastolic filling. Fibrotic mechanisms result in permanent scarring which often leads to hypertrophy, arrhythmias, and a rapid progression to failure. Despite the deep understanding of fibrosis in other tissues, acquired through previous investigations, the mechanisms of cardiac fibrosis remain unclear. Recent studies suggest that biochemical cues as well as mechanical cues regulate cells in myocardium. However, the steps in myofibroblast transdifferentiation, as well as the molecular mechanisms of such transdifferentiation in vivo, are poorly understood. This review is focused on defining myofibroblast physiology, scar mechanics, and examining current findings of myofibroblast regulation by mechanical stress, stiffness, and topography for understanding fibrotic disease dynamics.
Collapse
Affiliation(s)
- Peter Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Nick Chu
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Jennifer Davis
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| | - Deok-Ho Kim
- Department of Bioengineering, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
48
|
Johnston CM, Krafft AJ, Russe MF, Rog-Zielinska EA. A new look at the heart-novel imaging techniques. Herzschrittmacherther Elektrophysiol 2017; 29:14-23. [PMID: 29242981 DOI: 10.1007/s00399-017-0546-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/24/2017] [Indexed: 01/20/2023]
Abstract
The development and successful implementation of cutting-edge imaging technologies to visualise cardiac anatomy and function is a key component of effective diagnostic efforts in cardiology. Here, we describe a number of recent exciting advances in the field of cardiology spanning from macro- to micro- to nano-scales of observation, including magnetic resonance imaging, computed tomography, optical mapping, photoacoustic imaging, and electron tomography. The methodologies discussed are currently making the transition from scientific research to routine clinical use, albeit at different paces. We discuss the most likely trajectory of this transition into clinical research and standard diagnostics, and highlight the key challenges and opportunities associated with each of the methodologies.
Collapse
Affiliation(s)
- C M Johnston
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center - University of Freiburg, and Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - A J Krafft
- Department of Radiology, Medical Physics, Medical Center - Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - M F Russe
- Department of Radiology, Medical Center - Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - E A Rog-Zielinska
- Institute for Experimental Cardiovascular Medicine, University Heart Center, Medical Center - University of Freiburg, and Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
49
|
Worke LJ, Barthold JE, Seelbinder B, Novak T, Main RP, Harbin SL, Neu CP. Densification of Type I Collagen Matrices as a Model for Cardiac Fibrosis. Adv Healthc Mater 2017; 6. [PMID: 28881428 DOI: 10.1002/adhm.201700114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 06/10/2017] [Indexed: 12/17/2022]
Abstract
Cardiac fibrosis is a disease state characterized by excessive collagenous matrix accumulation within the myocardium that can lead to ventricular dilation and systolic failure. Current treatment options are severely lacking due in part to the poor understanding of the complexity of molecular pathways involved in cardiac fibrosis. To close this gap, in vitro model systems that recapitulate the defining features of the fibrotic cellular environment are in need. Type I collagen, a major cardiac extracellular matrix protein and the defining component of fibrotic depositions, is an attractive choice for a fibrosis model, but demonstrates poor mechanical strength due to solubility limits. However, plastic compression of collagen matrices is shown to significantly increase its mechanical properties. Here, confined compression of oligomeric, type I collagen matrices is utilized to resemble defining hallmarks seen in fibrotic tissue such as increased collagen content, fibril thickness, and bulk compressive modulus. Cardiomyocytes seeded on compressed matrices show a strong beating abrogation as observed in cardiac fibrosis. Gene expression analysis of selected fibrosis markers indicates fibrotic activation and cardiomyocyte maturation with regard to the existing literature. With these results, a promising first step toward a facile heart-on-chip model is presented to study cardiac fibrosis.
Collapse
Affiliation(s)
- Logan J. Worke
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
| | - Jeanne E. Barthold
- Department of Mechanical Engineering; University of Colorado Boulder; Boulder CO USA 80309
| | - Benjamin Seelbinder
- Department of Mechanical Engineering; University of Colorado Boulder; Boulder CO USA 80309
| | - Tyler Novak
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
| | - Russell P. Main
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
- Department of Basic Medical Sciences; Purdue University; West Lafayette IN USA 47906
| | - Sherry L. Harbin
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
- Department of Basic Medical Sciences; Purdue University; West Lafayette IN USA 47906
| | - Corey P. Neu
- Weldon School of Biomedical Engineering; Purdue University; West Lafayette IN USA 47906
- Department of Mechanical Engineering; University of Colorado Boulder; Boulder CO USA 80309
| |
Collapse
|
50
|
Adding dimension to cellular mechanotransduction: Advances in biomedical engineering of multiaxial cell-stretch systems and their application to cardiovascular biomechanics and mechano-signaling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017. [DOI: 10.1016/j.pbiomolbio.2017.06.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|