1
|
Sinitsyna A, Berezhnoy A, Semidetnov I, Naumov V, Sergeeva T, Bakumenko S, Slotvitsky M, Tsvelaya V, Agladze K. Advanced Cardiovascular Toxicity Screening: Integrating Human iPSC-Derived Cardiomyocytes with 2D In Silico Models. Cardiovasc Toxicol 2025; 25:790-804. [PMID: 40153244 DOI: 10.1007/s12012-025-09987-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
The pharmaceutical industry is evolving with the use of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) for in vitro cardiac safety screening. Traditional reliance on QT-interval prolongation as a main arrhythmogenicity marker is being challenged. In addition, the Comprehensive In Vitro Proarrhythmia Assay (CiPA) initiative recommends using computer modeling and in silico platforms as a more comprehensive approach for arrhythmogenicity testing in conjunction with hiPSC-CM in vitro screening. Our study presents an innovative platform that integrates in vitro hiPSC-CM propagation test with in silico models to assess the potential arrhythmogenic effect of drug-induced impact on ionic currents and electrophysiological intercellular coupling. Utilizing the electrophysiological and morphological characteristics of hiPSC-CM, we offer a thorough evaluation of potential drug-induced cardiac risks by computer modeling. We show, using the examples of lidocaine (100-300 μM) and Cyclophosphamide (639, 852 μM), that with the use of an integrative experimental and computer platform, it is possible to correctly display the clinical manifestations of side effects in advance.
Collapse
Affiliation(s)
- Anastasiya Sinitsyna
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
| | - Andrey Berezhnoy
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia, 129110
- ITMO University, Kronverksky Pr. 49, Bldg. A, St. Petersburg, Russia, 197101
| | - Ivan Semidetnov
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
| | - Vadim Naumov
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
| | - Tatyana Sergeeva
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia, 129110
- ITMO University, Kronverksky Pr. 49, Bldg. A, St. Petersburg, Russia, 197101
| | - Sergey Bakumenko
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia, 129110
- ITMO University, Kronverksky Pr. 49, Bldg. A, St. Petersburg, Russia, 197101
| | - Mikhail Slotvitsky
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701.
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia, 129110.
- ITMO University, Kronverksky Pr. 49, Bldg. A, St. Petersburg, Russia, 197101.
| | - Valeriya Tsvelaya
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701.
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia, 129110.
- ITMO University, Kronverksky Pr. 49, Bldg. A, St. Petersburg, Russia, 197101.
| | - Konstantin Agladze
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Dolgoprudny, Russia, 141701
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia, 129110
| |
Collapse
|
2
|
Kiseleva DG, Dzhabrailov VD, Aitova AA, Turchaninova EA, Tsvelaya VA, Kazakova MA, Plyusnina TY, Markin AM. Arrhythmogenic Potential of Myocardial Edema: The Interstitial Osmolality Induces Spiral Waves and Multiple Excitation Wavelets. Biomedicines 2024; 12:1770. [PMID: 39200234 PMCID: PMC11351629 DOI: 10.3390/biomedicines12081770] [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: 07/14/2024] [Revised: 08/01/2024] [Accepted: 08/03/2024] [Indexed: 09/02/2024] Open
Abstract
Myocardial edema is a common symptom of pathological processes in the heart, causing aggravation of cardiovascular diseases and leading to irreversible myocardial remodeling. Patient-based studies show that myocardial edema is associated with arrhythmias. Currently, there are no studies that have examined how edema may influence changes in calcium dynamics in the functional syncytium. We performed optical mapping of calcium dynamics on a monolayer of neonatal rat cardiomyocytes with Fluo-4. The osmolality of the solutions was adjusted using the NaCl content. The initial Tyrode solution contained 140 mM NaCl (1T) and the hypoosmotic solutions contained 105 (0.75T) and 70 mM NaCl (0.5T). This study demonstrated a sharp decrease in the calcium wave propagation speed with a decrease in the solution osmolality. The successive decrease in osmolality also showed a transition from a normal wavefront to spiral wave and multiple wavelets of excitation with wave break. Our study demonstrated that, in a cellular model, hypoosmolality and, as a consequence, myocardial edema, could potentially lead to fatal ventricular arrhythmias, which to our knowledge has not been studied before. At 0.75T spiral waves appeared, whereas multiple wavelets of excitation occurred in 0.5T, which had not been recorded previously in a two-dimensional monolayer under conditions of cell edema without changes in the pacing protocol.
Collapse
Affiliation(s)
- Diana G. Kiseleva
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia;
- Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Vitalii D. Dzhabrailov
- ITMO University, 191002 Saint-Petersburg, Russia; (V.D.D.); (V.A.T.)
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Aleria A. Aitova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
| | - Elena A. Turchaninova
- ITMO University, 191002 Saint-Petersburg, Russia; (V.D.D.); (V.A.T.)
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Valeriya A. Tsvelaya
- ITMO University, 191002 Saint-Petersburg, Russia; (V.D.D.); (V.A.T.)
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Maria A. Kazakova
- Department of Biophysics, Faculty of Physics, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Tatiana Yu. Plyusnina
- Department of Biophysics, Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Alexander M. Markin
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, 119991 Moscow, Russia;
- Medical Institute, Peoples’ Friendship University of Russia named after Patrice Lumumba (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
3
|
Abrasheva VO, Kovalenko SG, Slotvitsky M, Romanova SА, Aitova AA, Frolova S, Tsvelaya V, Syunyaev RA. Human sodium current voltage-dependence at physiological temperature measured by coupling a patch-clamp experiment to a mathematical model. J Physiol 2024; 602:633-661. [PMID: 38345560 DOI: 10.1113/jp285162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/02/2024] [Indexed: 02/20/2024] Open
Abstract
Voltage-gated Na+ channels are crucial to action potential propagation in excitable tissues. Because of the high amplitude and rapid activation of the Na+ current, voltage-clamp measurements are very challenging and are usually performed at room temperature. In this study, we measured Na+ current voltage-dependence in stem cell-derived cardiomyocytes at physiological temperature. While the apparent activation and inactivation curves, measured as the dependence of current amplitude on voltage, fall within the range reported in previous studies, we identified a systematic error in our measurements. This error is caused by the deviation of the membrane potential from the command potential of the amplifier. We demonstrate that it is possible to account for this artifact using computer simulation of the patch-clamp experiment. We obtained surprising results through patch-clamp model optimization: a half-activation of -11.5 mV and a half-inactivation of -87 mV. Although the half-activation deviates from previous research, we demonstrate that this estimate reproduces the conduction velocity dependence on extracellular potassium concentration. KEY POINTS: Voltage-gated Na+ currents play a crucial role in excitable tissues including neurons, cardiac and skeletal muscle. Measurement of Na+ current is challenging because of its high amplitude and rapid kinetics, especially at physiological temperature. We have used the patch-clamp technique to measure human Na+ current voltage-dependence in human induced pluripotent stem cell-derived cardiomyocytes. The patch-clamp data were processed by optimization of the model accounting for voltage-clamp experiment artifacts, revealing a large difference between apparent parameters of Na+ current and the results of the optimization. We conclude that actual Na+ current activation is extremely depolarized in comparison to previous studies. The new Na+ current model provides a better understanding of action potential propagation; we demonstrate that it explains propagation in hyperkalaemic conditions.
Collapse
Affiliation(s)
| | - Sandaara G Kovalenko
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | - Mihail Slotvitsky
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | - Serafima А Romanova
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | - Aleria A Aitova
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | - Sheida Frolova
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
| | - Valeria Tsvelaya
- Moscow Institute of Physics and Technology, Moscow, Russia
- M. F. Vladimirsky Moscow Regional Research Clinical Institute, Moscow, Russia
- ITMO University, St Petersburg, Russia
| | | |
Collapse
|
4
|
Aitova A, Berezhnoy A, Tsvelaya V, Gusev O, Lyundup A, Efimov AE, Agapov I, Agladze K. Biomimetic Cardiac Tissue Models for In Vitro Arrhythmia Studies. Biomimetics (Basel) 2023; 8:487. [PMID: 37887618 PMCID: PMC10604593 DOI: 10.3390/biomimetics8060487] [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: 08/28/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Cardiac arrhythmias are a major cause of cardiovascular mortality worldwide. Many arrhythmias are caused by reentry, a phenomenon where excitation waves circulate in the heart. Optical mapping techniques have revealed the role of reentry in arrhythmia initiation and fibrillation transition, but the underlying biophysical mechanisms are still difficult to investigate in intact hearts. Tissue engineering models of cardiac tissue can mimic the structure and function of native cardiac tissue and enable interactive observation of reentry formation and wave propagation. This review will present various approaches to constructing cardiac tissue models for reentry studies, using the authors' work as examples. The review will highlight the evolution of tissue engineering designs based on different substrates, cell types, and structural parameters. A new approach using polymer materials and cellular reprogramming to create biomimetic cardiac tissues will be introduced. The review will also show how computational modeling of cardiac tissue can complement experimental data and how such models can be applied in the biomimetics of cardiac tissue.
Collapse
Affiliation(s)
- Aleria Aitova
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Andrey Berezhnoy
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Valeriya Tsvelaya
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Oleg Gusev
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420018 Kazan, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | | | - Anton E. Efimov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Igor Agapov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Konstantin Agladze
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
| |
Collapse
|
5
|
Djemai M, Cupelli M, Boutjdir M, Chahine M. Optical Mapping of Cardiomyocytes in Monolayer Derived from Induced Pluripotent Stem Cells. Cells 2023; 12:2168. [PMID: 37681899 PMCID: PMC10487143 DOI: 10.3390/cells12172168] [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: 07/18/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Optical mapping is a powerful imaging technique widely adopted to measure membrane potential changes and intracellular Ca2+ variations in excitable tissues using voltage-sensitive dyes and Ca2+ indicators, respectively. This powerful tool has rapidly become indispensable in the field of cardiac electrophysiology for studying depolarization wave propagation, estimating the conduction velocity of electrical impulses, and measuring Ca2+ dynamics in cardiac cells and tissues. In addition, mapping these electrophysiological parameters is important for understanding cardiac arrhythmia mechanisms. In this review, we delve into the fundamentals of cardiac optical mapping technology and its applications when applied to hiPSC-derived cardiomyocytes and discuss related advantages and challenges. We also provide a detailed description of the processing and analysis of optical mapping data, which is a crucial step in the study of cardiac diseases and arrhythmia mechanisms for extracting and comparing relevant electrophysiological parameters.
Collapse
Affiliation(s)
- Mohammed Djemai
- CERVO Brain Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC G1J 2G3, Canada
| | - Michael Cupelli
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, New York, NY 11203, USA
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, New York, NY 11209, USA
- Department of Medicine, Cell Biology and Pharmacology, State University of New York Downstate Health Sciences University, New York, NY 11203, USA
- Department of Medicine, NYU School of Medicine, New York, NY 10016, USA
| | - Mohamed Chahine
- CERVO Brain Research Center, Institut Universitaire en Santé Mentale de Québec, Quebec City, QC G1J 2G3, Canada
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
6
|
Chen Y, Huang P, Niu M, Tian C, Zhang T, Peng Z. Regeneration of T cells from human-induced pluripotent stem cells for CAR-T cell medicated immunotherapy. Front Bioeng Biotechnol 2023; 11:1159507. [PMID: 37274170 PMCID: PMC10233047 DOI: 10.3389/fbioe.2023.1159507] [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: 02/06/2023] [Accepted: 05/08/2023] [Indexed: 06/06/2023] Open
Abstract
Background: Chimeric antigen receptor (CAR) T cell treatment involves in vitro production of T cells from patient blood with synthetic receptors specific to a cancer antigen. They circumvent the major histocompatibility complex to recognize the tumor antigen, reducing hematologic malignancy remission rates by 80%. Considering the efficacy of CAR-T treatment, the present work aimed at generating functional clusters of differentiation (CD)8 + T cells from human induced pluripotent stem cells (hiPSC) and to generate hiPS-CAR-T cells with high antigen-specific cytotoxicity. Methods: The Alkaline phosphatase assay and MycoEasy rapid mycoplasma detection kit was implemented for detection of hiPSCs and mycoplasma, respectively. The CD34+ HSPCs were harvested in AggreWellTM 400 using a 37-micron reversible strainer. Likewise, the lymphoid progenitor and CD4+CD8+ DP T cells were also harvested. The Cell Counting Kit-8 (CCK-8) assay was used to mark cytotoxicity and ELISA was used to detect IFN-γ secretion. Further, flow cytometry and transwell chambers were used to assess cell cycle, and migration and invasion. Finally, the in vivo antitumor effects of the CAR-T cells were evaluated using experimental animals (mice). Results: Results revealed that a serum-free, feeder layer-free differentiation system significantly yielded hiPSC-based T cell immunotherapy with interleukin-2, interleukin-15, and activators at the differentiation stage to promote the maturation of these cells into human induced pluripotent stem (hiPS)-T cells. The infection of hiPSCs with the CD19 CAR lentivirus resulted in the production of the hiPSC-CAR-T cells. We validated the function of hiPS-CAR-T cells in vivo and in vitro experimentation which revealed no significant differences in cell morphology and function between hiPSC-derived hiPS-CAR-T cells and peripheral blood-derived CAR-T cells. Conclusion: This study developed a culture method that is efficient and clinically useful to make functional CD8+ T cells from hiPSC and to get hiPS-CAR-T cells with high antigen-specific cytotoxicity that are not very different from CAR T cells found in peripheral blood. As a result, our findings may open the way for the clinical use of hiPSC to create functional CD8+ T and hiPS-CAR-T cells cells for use in cell-based cancer therapy.
Collapse
|
7
|
Cellular electrophysiological effects of botulinum toxin A on neonatal rat cardiomyocytes and on cardiomyocytes derived from human-induced pluripotent stem cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:513-524. [PMID: 36399184 DOI: 10.1007/s00210-022-02332-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/07/2022] [Indexed: 11/19/2022]
Abstract
Botulinum toxin A is a well-known neurotransmitter inhibitor with a wide range of applications in modern medicine. Recently, botulinum toxin A preparations have been used in clinical trials to suppress cardiac arrhythmias, especially in the postoperative period. Its antiarrhythmic action is associated with inhibition of the nervous system of the heart, but its direct effect on heart tissue remains unclear. Accordingly, we investigate the effect of botulinum toxin A on isolated cardiac cells and on layers of cardiac cells capable of conducting excitation. Cardiomyocytes of neonatal rat pups and human cardiomyocytes obtained through cell reprogramming were used. A patch-clamp study showed that botulinum toxin A inhibited fast sodium currents and L-type calcium currents in a dose-dependent manner, with no apparent effect on potassium currents. Optical mapping showed that in the presence of botulinum toxin A, the propagation of the excitation wave in the layer of cardiac cells slows down sharply, conduction at high concentrations becomes chaotic, but reentry waves do not form. The combination of botulinum toxin A with a preparation of chitosan showed a stronger inhibitory effect by an order of magnitude. Further, the inhibitory effect of botulinum toxin A is not permanent and disappears after 12 days of cell culture in a botulinum toxin A-free medium. The main conclusion of the work is that the antiarrhythmic effect of botulinum toxin A found in clinical studies is associated not only with depression of the nervous system but also with a direct effect on heart tissue. Moreover, the combination of botulinum toxin A and chitosan reduces the effective dose of botulinum toxin A.
Collapse
|
8
|
Slotvitsky M, Berezhnoy A, Scherbina S, Rimskaya B, Tsvelaya V, Balashov V, Efimov AE, Agapov I, Agladze K. Polymer Kernels as Compact Carriers for Suspended Cardiomyocytes. MICROMACHINES 2022; 14:51. [PMID: 36677111 PMCID: PMC9865253 DOI: 10.3390/mi14010051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
Induced pluripotent stem cells (iPSCs) constitute a potential source of patient-specific human cardiomyocytes for a cardiac cell replacement therapy via intramyocardial injections, providing a major benefit over other cell sources in terms of immune rejection. However, intramyocardial injection of the cardiomyocytes has substantial challenges related to cell survival and electrophysiological coupling with recipient tissue. Current methods of manipulating cell suspensions do not allow one to control the processes of adhesion of injected cells to the tissue and electrophysiological coupling with surrounding cells. In this article, we documented the possibility of influencing these processes using polymer kernels: biocompatible fiber fragments of subcellular size that can be adsorbed to a cell, thereby creating the minimum necessary adhesion foci to shape the cell and provide support for the organization of the cytoskeleton and the contractile apparatus prior to adhesion to the recipient tissue. Using optical excitation markers, the restoration of the excitability of cardiomyocytes in suspension upon adsorption of polymer kernels was shown. It increased the likelihood of the formation of a stable electrophysiological coupling in vitro. The obtained results may be considered as a proof of concept that the stochastic engraftment process of injected suspension cells can be controlled by smart biomaterials.
Collapse
Affiliation(s)
- Mikhail Slotvitsky
- Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Andrey Berezhnoy
- Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Serafima Scherbina
- Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
| | - Beatrisa Rimskaya
- Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
| | - Valerya Tsvelaya
- Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Victor Balashov
- Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
| | - Anton E. Efimov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, Schukinskaya St., 1, 123182 Moscow, Russia
| | - Igor Agapov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, Schukinskaya St., 1, 123182 Moscow, Russia
| | - Konstantin Agladze
- Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| |
Collapse
|
9
|
Adami R, Bottai D. NSC Physiological Features in Spinal Muscular Atrophy: SMN Deficiency Effects on Neurogenesis. Int J Mol Sci 2022; 23:ijms232315209. [PMID: 36499528 PMCID: PMC9736802 DOI: 10.3390/ijms232315209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/20/2022] [Accepted: 11/30/2022] [Indexed: 12/08/2022] Open
Abstract
While the U.S. Food and Drug Administration and the European Medicines Evaluation Agency have recently approved new drugs to treat spinal muscular atrophy 1 (SMA1) in young patients, they are mostly ineffective in older patients since many motor neurons have already been lost. Therefore, understanding nervous system (NS) physiology in SMA patients is essential. Consequently, studying neural stem cells (NSCs) from SMA patients is of significant interest in searching for new treatment targets that will enable researchers to identify new pharmacological approaches. However, studying NSCs in these patients is challenging since their isolation damages the NS, making it impossible with living patients. Nevertheless, it is possible to study NSCs from animal models or create them by differentiating induced pluripotent stem cells obtained from SMA patient peripheral tissues. On the other hand, therapeutic interventions such as NSCs transplantation could ameliorate SMA condition. This review summarizes current knowledge on the physiological properties of NSCs from animals and human cellular models with an SMA background converging on the molecular and neuronal circuit formation alterations of SMA fetuses and is not focused on the treatment of SMA. By understanding how SMA alters NSC physiology, we can identify new and promising interventions that could help support affected patients.
Collapse
|
10
|
Maretina MA, Valetdinova KR, Tsyganova NA, Egorova AA, Ovechkina VS, Schiöth HB, Zakian SM, Baranov VS, Kiselev AV. Identification of specific gene methylation patterns during motor neuron differentiation from spinal muscular atrophy patient-derived iPSC. Gene 2022; 811:146109. [PMID: 34871761 DOI: 10.1016/j.gene.2021.146109] [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: 08/12/2020] [Revised: 05/08/2021] [Accepted: 11/16/2021] [Indexed: 11/04/2022]
Abstract
Spinal muscular atrophy is a progressive motor neuron disorder caused by deletions or point mutations in the SMN1 gene. It is not known why motor neurons are particularly sensitive to a decrease in SMN protein levels and what factors besides SMN2 underlie the high clinical heterogeneity of the disease. Here we studied the methylation patterns of genes on sequential stages of motor neuron differentiation from induced pluripotent stem cells derived from the patients with SMA type I and II. The genes involved in the regulation of pluripotency, neural differentiation as well as those associated with spinal muscular atrophy development were included. The results show that the PAX6, HB9, CHAT, ARHGAP22, and SMN2 genes are differently methylated in cells derived from SMA patients compared to the cells of healthy individuals. This study clarifies the specificities of the disease pathogenesis and extends the knowledge of pathways involved in the SMA progression.
Collapse
Affiliation(s)
- M A Maretina
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia
| | - K R Valetdinova
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novsibirsk, Russia
| | - N A Tsyganova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia
| | - A A Egorova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia
| | - V S Ovechkina
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novsibirsk, Russia; Novosibirsk State University, 630090 Novosibirsk, Russia
| | - H B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, S-75124 Uppsala, Sweden; Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - S M Zakian
- The Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novsibirsk, Russia; Meshalkin National Medical Research Center, Ministry of Healthcare of the Russian Federation, 630055 Novosibirsk, Russia
| | - V S Baranov
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia
| | - A V Kiselev
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 Saint-Petersburg, Russia.
| |
Collapse
|
11
|
Podgurskaya AD, Slotvitsky MM, Tsvelaya VA, Frolova SR, Romanova SG, Balashov VA, Agladze KI. Cyclophosphamide arrhythmogenicitytesting using human-induced pluripotent stem cell-derived cardiomyocytes. Sci Rep 2021; 11:2336. [PMID: 33504826 PMCID: PMC7841168 DOI: 10.1038/s41598-020-79085-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 12/04/2020] [Indexed: 11/09/2022] Open
Abstract
Cyclophosphamide (CP) is an anticancer drug, an alkylating agent. Cardiotoxicity of CP is associated with one of its metabolites, acrolein, and clinical cardiotoxicity manifestations are described for cases of taking CP in high doses. Nevertheless, modern arrhythmogenicity prediction assays in vitro include evaluation of beat rhythm and rate as well as suppression of cardiac late markers after acute exposure to CP, but not its metabolites. The mechanism of CP side effects when taken at low doses (i.e., < 100 mg/kg), especially at the cellular level, remains unclear. In this study conduction properties and cytoskeleton structure of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) obtained from a healthy donor under CP were evaluated. Arrhythmogenicity testing including characterization of 3 values: conduction velocity, maximum capture rate (MCR) measurements and number of occasions of re-entry on a standard linear obstacle was conducted and revealed MCR decrease of 25% ± 7% under CP. Also, conductivity area reduced by 34 ± 15%. No effect of CP on voltage-gated ion channels was found. Conduction changes (MCR and conductivity area decrease) are caused by exposure time-dependent alpha-actinin disruption detected both in hiPSC-CMs and neonatal ventricular cardiomyocytes in vitro. Deviation from the external stimulus frequency and appearance of non-conductive areas in cardiac tissue under CP is potentially arrhythmogenic and could develop arrhythmic effects in vivo.
Collapse
Affiliation(s)
- A D Podgurskaya
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - M M Slotvitsky
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - V A Tsvelaya
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - S R Frolova
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - S G Romanova
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - V A Balashov
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701, Russian Federation
| | - K I Agladze
- Moscow Institute of Physics and Technology (National Research University), Dolgoprudny, Moscow Region, 141701, Russian Federation.
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Moscow, 129110, Russian Federation.
| |
Collapse
|
12
|
Arrhythmia Mechanisms in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. J Cardiovasc Pharmacol 2020; 77:300-316. [PMID: 33323698 DOI: 10.1097/fjc.0000000000000972] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/08/2020] [Indexed: 12/30/2022]
Abstract
ABSTRACT Despite major efforts by clinicians and researchers, cardiac arrhythmia remains a leading cause of morbidity and mortality in the world. Experimental work has relied on combining high-throughput strategies with standard molecular and electrophysiological studies, which are, to a great extent, based on the use of animal models. Because this poses major challenges for translation, the progress in the development of novel antiarrhythmic agents and clinical care has been mostly disappointing. Recently, the advent of human induced pluripotent stem cell-derived cardiomyocytes has opened new avenues for both basic cardiac research and drug discovery; now, there is an unlimited source of cardiomyocytes of human origin, both from healthy individuals and patients with cardiac diseases. Understanding arrhythmic mechanisms is one of the main use cases of human induced pluripotent stem cell-derived cardiomyocytes, in addition to pharmacological cardiotoxicity and efficacy testing, in vitro disease modeling, developing patient-specific models and personalized drugs, and regenerative medicine. Here, we review the advances that the human induced pluripotent stem cell-derived-based modeling systems have brought so far regarding the understanding of both arrhythmogenic triggers and substrates, while also briefly speculating about the possibilities in the future.
Collapse
|
13
|
Volova LT, Pugachev EI, Rossinskaya VV, Boltovskaya VV, Dolgushkin DA, Ossina N. Rheumatoid Arthritis: Applicability of Ready-to-Use Human Cartilaginous Cells for Screening of Compounds with TNF-Alpha Inhibitory Activity. Biomolecules 2020; 10:biom10111563. [PMID: 33212930 PMCID: PMC7698400 DOI: 10.3390/biom10111563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/29/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022] Open
Abstract
In the context of modern drug discovery, there is an obvious advantage to designing phenotypic bioassays based on human disease-relevant cells that express disease-relevant markers. The specific aim of the study was to develop a convenient and reliable method for screening compounds with Tumor Necrosis Factor-alpha (TNF-α) inhibitory activity. This assay was developed using cryopreserved ready-to-use cartilage-derived cells isolated from juvenile donors diagnosed with polydactyly. It has been demonstrated that all donor (10 donors) cells were able to respond to TNF-α treatment by increased secretion of pro-inflammatory cytokine IL-6 into subcultural medium. Inhibition of TNF-α using commercially available TNF-α inhibitor etanercept resulted in a dose-dependent decrease in IL-6 production which was measured by Enzyme-Linked Immunosorbent Assay (ELISA). TNF-α dependent IL-6 production was detected in the cells after both their prolonged cultivation in vitro (≥20 passages) and cryopreservation. This phenotypic bioassay based on ready-to-use primary human cells was developed for detection of novel TNF-α inhibitory compounds and profiling of biosimilar drugs.
Collapse
|
14
|
Muscular Thin Films for Label-Free Mapping of Excitation Propagation in Cardiac Tissue. Ann Biomed Eng 2020; 48:2425-2437. [PMID: 32314299 DOI: 10.1007/s10439-020-02513-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/11/2020] [Indexed: 01/10/2023]
Abstract
Muscular thin films (MTFs), have already found a variety of applications in cardiac tissue engineering and in building of lab-on-a-chip systems. Here we present a novel approach to label-free mapping of excitation waves in the cardiomyocyte cell cultures with the use of MTFs. Neonatal rat ventricular cardiomyocytes were cultured on polydimethylsiloxane (PDMS) thin films and observed by means of off-axis illumination. Inflexions of the membrane created by the contraction of cardiomyocytes led to formation of patterns of bright and dark areas on the surface of the membrane. These patterns were recorded and analyzed for the monitoring of the contraction propagation. The method was compared with a standard optical mapping technique based on the use of a Ca2+-sensitive fluorescent dye. A good consistency of the results obtained by these two methods was demonstrated. The proposed method is non-toxic and might be of particular interest for the purpose of continuous monitoring in test systems based on human induced pluripotent stem cells.
Collapse
|
15
|
van Gorp PRR, Trines SA, Pijnappels DA, de Vries AAF. Multicellular In vitro Models of Cardiac Arrhythmias: Focus on Atrial Fibrillation. Front Cardiovasc Med 2020; 7:43. [PMID: 32296716 PMCID: PMC7138102 DOI: 10.3389/fcvm.2020.00043] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/06/2020] [Indexed: 12/13/2022] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia in clinical practice with a large socioeconomic impact due to its associated morbidity, mortality, reduction in quality of life and health care costs. Currently, antiarrhythmic drug therapy is the first line of treatment for most symptomatic AF patients, despite its limited efficacy, the risk of inducing potentially life-threating ventricular tachyarrhythmias as well as other side effects. Alternative, in-hospital treatment modalities consisting of electrical cardioversion and invasive catheter ablation improve patients' symptoms, but often have to be repeated and are still associated with serious complications and only suitable for specific subgroups of AF patients. The development and progression of AF generally results from the interplay of multiple disease pathways and is accompanied by structural and functional (e.g., electrical) tissue remodeling. Rational development of novel treatment modalities for AF, with its many different etiologies, requires a comprehensive insight into the complex pathophysiological mechanisms. Monolayers of atrial cells represent a simplified surrogate of atrial tissue well-suited to investigate atrial arrhythmia mechanisms, since they can easily be used in a standardized, systematic and controllable manner to study the role of specific pathways and processes in the genesis, perpetuation and termination of atrial arrhythmias. In this review, we provide an overview of the currently available two- and three-dimensional multicellular in vitro systems for investigating the initiation, maintenance and termination of atrial arrhythmias and AF. This encompasses cultures of primary (animal-derived) atrial cardiomyocytes (CMs), pluripotent stem cell-derived atrial-like CMs and (conditionally) immortalized atrial CMs. The strengths and weaknesses of each of these model systems for studying atrial arrhythmias will be discussed as well as their implications for future studies.
Collapse
Affiliation(s)
| | | | | | - Antoine A. F. de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|