1
|
Kong C, Guo Z, Teng T, Yao Q, Yu J, Wang M, Ma Y, Wang P, Tang Q. Electroactive Nanomaterials for the Prevention and Treatment of Heart Failure: From Materials and Mechanisms to Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406206. [PMID: 39268781 DOI: 10.1002/smll.202406206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Heart failure (HF) represents a cardiovascular disease that significantly threatens global well-being and quality of life. Electroactive nanomaterials, characterized by their distinctive physical and chemical properties, emerge as promising candidates for HF prevention and management. This review comprehensively examines electroactive nanomaterials and their applications in HF intervention. It presents the definition, classification, and intrinsic characteristics of conductive, piezoelectric, and triboelectric nanomaterials, emphasizing their mechanical robustness, electrical conductivity, and piezoelectric coefficients. The review elucidates their applications and mechanisms: 1) early detection and diagnosis, employing nanomaterial-based sensors for real-time cardiac health monitoring; 2) cardiac tissue repair and regeneration, providing mechanical, chemical, and electrical stimuli for tissue restoration; 3) localized administration of bioactive biomolecules, genes, or pharmacotherapeutic agents, using nanomaterials as advanced drug delivery systems; and 4) electrical stimulation therapies, leveraging their properties for innovative pacemaker and neurostimulation technologies. Challenges in clinical translation, such as biocompatibility, stability, and scalability, are discussed, along with future prospects and potential innovations, including multifunctional and stimuli-responsive nanomaterials for precise HF therapies. This review encapsulates current research and future directions concerning the use of electroactive nanomaterials in HF prevention and management, highlighting their potential to innovating in cardiovascular medicine.
Collapse
Affiliation(s)
- Chunyan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Jiabin Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Yulan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| |
Collapse
|
2
|
Joshi J, Albers C, Smole N, Guo S, Smith SA. Human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) for modeling cardiac arrhythmias: strengths, challenges and potential solutions. Front Physiol 2024; 15:1475152. [PMID: 39328831 PMCID: PMC11424716 DOI: 10.3389/fphys.2024.1475152] [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: 08/02/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
Ion channels and cytoskeletal proteins in the cardiac dyad play a critical role in maintaining excitation-contraction (E-C) coupling and provide cardiac homeostasis. Functional changes in these dyad proteins, whether induced by genetic, epigenetic, metabolic, therapeutic, or environmental factors, can disrupt normal cardiac electrophysiology, leading to abnormal E-C coupling and arrhythmias. Animal models and heterologous cell cultures provide platforms to elucidate the pathogenesis of arrhythmias for basic cardiac research; however, these traditional systems do not truly reflect human cardiac electro-pathophysiology. Notably, patients with the same genetic variants of inherited channelopathies (ICC) often exhibit incomplete penetrance and variable expressivity which underscores the need to establish patient-specific disease models to comprehend the mechanistic pathways of arrhythmias and determine personalized therapies. Patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) inherit the genetic background of the patient and reflect the electrophysiological characteristics of the native cardiomyocytes. Thus, iPSC-CMs provide an innovative and translational pivotal platform in cardiac disease modeling and therapeutic screening. In this review, we will examine how patient-specific iPSC-CMs historically evolved to model arrhythmia syndromes in a dish, and their utility in understanding the role of specific ion channels and their functional characteristics in causing arrhythmias. We will also examine how CRISPR/Cas9 have enabled the establishment of patient-independent and variant-induced iPSC-CMs-based arrhythmia models. Next, we will examine the limitations of using human iPSC-CMs with respect to in vitro arrhythmia modeling that stems from variations in iPSCs or toxicity due to gene editing on iPSC or iPSC-CMs and explore how such hurdles are being addressed. Importantly, we will also discuss how novel 3D iPSC-CM models can better capture in vitro characteristics and how all-optical platforms provide non-invasive and high- throughput electrophysiological data that is useful for stratification of emerging arrhythmogenic variants and drug discovery. Finally, we will examine strategies to improve iPSC-CM maturity, including powerful gene editing and optogenetic tools that can introduce/modify specific ion channels in iPSC-CMs and tailor cellular and functional characteristics. We anticipate that an elegant synergy of iPSCs, novel gene editing, 3D- culture models, and all-optical platforms will offer a high-throughput template to faithfully recapitulate in vitro arrhythmogenic events necessary for personalized arrhythmia monitoring and drug screening process.
Collapse
Affiliation(s)
- Jyotsna Joshi
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Cora Albers
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Nathan Smole
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Shuliang Guo
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Sakima A Smith
- Department of Internal Medicine, Division of Cardiology, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
3
|
Wu HF, Hamilton C, Porritt H, Winbo A, Zeltner N. Modelling neurocardiac physiology and diseases using human pluripotent stem cells: current progress and future prospects. J Physiol 2024. [PMID: 39235952 DOI: 10.1113/jp286416] [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: 02/19/2024] [Accepted: 08/07/2024] [Indexed: 09/07/2024] Open
Abstract
Throughout our lifetime the heart executes cycles of contraction and relaxation to meet the body's ever-changing metabolic needs. This vital function is continuously regulated by the autonomic nervous system. Cardiovascular dysfunction and autonomic dysregulation are also closely associated; however, the degrees of cause and effect are not always readily discernible. Thus, to better understand cardiovascular disorders, it is crucial to develop model systems that can be used to study the neurocardiac interaction in healthy and diseased states. Human pluripotent stem cell (hiPSC) technology offers a unique human-based modelling system that allows for studies of disease effects on the cells of the heart and autonomic neurons as well as of their interaction. In this review, we summarize current understanding of the embryonic development of the autonomic, cardiac and neurocardiac systems, their regulation, as well as recent progress of in vitro modelling systems based on hiPSCs. We further discuss the advantages and limitations of hiPSC-based models in neurocardiac research.
Collapse
Affiliation(s)
- Hsueh-Fu Wu
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Charlotte Hamilton
- Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Harrison Porritt
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Department of Chemical and Materials Engineering, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
| | - Annika Winbo
- Department of Physiology, The University of Auckland, Auckland, New Zealand
- Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Nadja Zeltner
- Center for Molecular Medicine, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Department of Cellular Biology, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
4
|
Belzil A, Gélinas R, Comtois P. Development of a high-speed imaging system for real time evaluation and monitoring of cardiac engineered tissues. Front Bioeng Biotechnol 2024; 12:1403044. [PMID: 39188370 PMCID: PMC11345265 DOI: 10.3389/fbioe.2024.1403044] [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: 03/18/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Stem cell derived cardiac monolayers have high potential for tissue regeneration, in vitro drug testing and disease modeling. However, current differentiation protocols are still sub-optimal, resulting in cultures with variable yields and properties. We propose a high-speed lenseless imaging system, integrated with an electrical stimulation unit, to optimize the generation of these cultures. This tool relies on the variations of cellular patterns, during contraction, measured by digital imaging. The imaging system can monitor cardiac cell sheet function and structure, providing the necessary tools to quickly evaluate engineered monolayer. It can record high speed videos and capture high resolution images, from which tissue spatial organization and contractile characteristics can be obtained. Validation of the system was performed using cardiomyocytes derived from human induced pluripotent stem cell and neonatal rat cardiomyocytes. The imaging system allows the observation, acquisition and analysis of important data relating to contractile activity development of cardiac cells, making it a promising tool for optimization in cardiac tissue engineering.
Collapse
Affiliation(s)
- Antoine Belzil
- Research Centre, Montreal Heart Institute, Montréal, QC, Canada
- Department of Pharmacology and Physiology, Universite de Montreal, Montréal, QC, Canada
- Institute of Biomedical Engineering, Universite de Montreal, Montréal, QC, Canada
| | - Roselle Gélinas
- Laboratory of Genetics and Genomic Medicine of Inflammation, Montreal Heart Institute, Montréal, QC, Canada
- Department of Medicine, Universite de Montreal, Montréal, QC, Canada
| | - Philippe Comtois
- Department of Pharmacology and Physiology, Universite de Montreal, Montréal, QC, Canada
- Institute of Biomedical Engineering, Universite de Montreal, Montréal, QC, Canada
- School of Pharmaceutical Sciences, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
5
|
Cai WF, Jiang L, Liang J, Dutta S, Huang W, He X, Wu Z, Paul C, Gao X, Xu M, Kanisicak O, Zheng J, Wang Y. HAX1-Overexpression Augments Cardioprotective Efficacy of Stem Cell-Based Therapy Through Mediating Hippo-Yap Signaling. Stem Cell Rev Rep 2024; 20:1569-1586. [PMID: 38713406 PMCID: PMC11319392 DOI: 10.1007/s12015-024-10729-z] [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] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Although stem/progenitor cell therapy shows potential for myocardial infarction repair, enhancing the therapeutic efficacy could be achieved through additional genetic modifications. HCLS1-associated protein X-1 (HAX1) has been identified as a versatile modulator responsible for cardio-protective signaling, while its role in regulating stem cell survival and functionality remains unknown. In this study, we investigated whether HAX1 can augment the protective potential of Sca1+ cardiac stromal cells (CSCs) for myocardial injury. The overexpression of HAX1 significantly increased cell proliferation and conferred enhanced resistance to hypoxia-induced cell death in CSCs. Mechanistically, HAX1 can interact with Mst1 (a prominent conductor of Hippo signal transduction) and inhibit its kinase activity for protein phosphorylation. This inhibition led to enhanced nuclear translocation of Yes-associated protein (YAP) and activation of downstream therapeutic-related genes. Notably, HAX1 overexpression significantly increased the pro-angiogenic potential of CSCs, as demonstrated by elevated expression of vascular endothelial growth factors. Importantly, implantation of HAX1-overexpressing CSCs promoted neovascularization, protected against functional deterioration, and ameliorated cardiac fibrosis in ischemic mouse hearts. In conclusion, HAX1 emerges as a valuable and efficient inducer for enhancing the effectiveness of cardiac stem or progenitor cell therapeutics.
Collapse
Affiliation(s)
- Wen-Feng Cai
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267-0529, USA
| | - Wei Huang
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267-0529, USA
| | - Xingyu He
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Xiang Gao
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA
| | - Junmeng Zheng
- Department of Cardiovascular Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No.107 Yanjiang West Road, Guangzhou, 510120, Guangdong, China.
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0529, USA.
| |
Collapse
|
6
|
Inouye K, Yeganyan S, Kay K, Thankam FG. Programmed spontaneously beating cardiomyocytes in regenerative cardiology. Cytotherapy 2024; 26:790-796. [PMID: 38520412 DOI: 10.1016/j.jcyt.2024.03.004] [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: 12/20/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024]
Abstract
Stem cells have gained attention as a promising therapeutic approach for damaged myocardium, and there have been efforts to develop a protocol for regenerating cardiomyocytes (CMs). Certain cells have showed a greater aptitude for yielding beating CMs, such as induced pluripotent stem cells, embryonic stem cells, adipose-derived stromal vascular fraction cells and extended pluripotent stem cells. The approach for generating CMs from stem cells differs across studies, although there is evidence that Wnt signaling, chemical additives, electrical stimulation, co-culture, biomaterials and transcription factors triggers CM differentiation. Upregulation of Gata4, Mef2c and Tbx5 transcription factors has been correlated with successfully induced CMs, although Mef2c may potentially play a more prominent role in the generation of the beating phenotype, specifically. Regenerative research provides a possible candidate for cardiac repair; however, it is important to identify factors that influence their differentiation. Altogether, the spontaneously beating CMs would be monumental for regenerative research for cardiac repair.
Collapse
Affiliation(s)
- Keiko Inouye
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Stephanie Yeganyan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Kaelen Kay
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA
| | - Finosh G Thankam
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, USA.
| |
Collapse
|
7
|
Chen EZ, Kannan S, Murphy S, Farid M, Kwon C. Protocol for quantifying stem-cell-derived cardiomyocyte maturity using transcriptomic entropy score. STAR Protoc 2024; 5:103083. [PMID: 38781077 PMCID: PMC11145390 DOI: 10.1016/j.xpro.2024.103083] [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: 12/08/2023] [Revised: 03/15/2024] [Accepted: 05/01/2024] [Indexed: 05/25/2024] Open
Abstract
The inability to quantify cardiomyocyte (CM) maturation remains a significant barrier to evaluating the effects of ongoing efforts to produce adult-like CMs from pluripotent stem cells (PSCs). Here, we present a protocol to quantify stem-cell-derived CM maturity using a single-cell RNA sequencing-based metric "entropy score." We describe steps for generating an entropy score using customized R code. This tool can be used to quantify maturation levels of PSC-CMs and potentially other cell types. For complete details on the use and execution of this protocol, please refer to Kannan et al.1.
Collapse
Affiliation(s)
- Elaine Zhelan Chen
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Suraj Kannan
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Sean Murphy
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Michael Farid
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA; Department of Cell Biology, Johns Hopkins School of Medicine; Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins School of Medicine; Baltimore, MD, USA.
| |
Collapse
|
8
|
Bravo-Olín J, Martínez-Carreón SA, Francisco-Solano E, Lara AR, Beltran-Vargas NE. Analysis of the role of perfusion, mechanical, and electrical stimulation in bioreactors for cardiac tissue engineering. Bioprocess Biosyst Eng 2024; 47:767-839. [PMID: 38643271 DOI: 10.1007/s00449-024-03004-5] [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: 09/16/2023] [Accepted: 03/13/2024] [Indexed: 04/22/2024]
Abstract
Since cardiovascular diseases (CVDs) are globally one of the leading causes of death, of which myocardial infarction (MI) can cause irreversible damage and decrease survivors' quality of life, novel therapeutics are needed. Current approaches such as organ transplantation do not fully restore cardiac function or are limited. As a valuable strategy, tissue engineering seeks to obtain constructs that resemble myocardial tissue, vessels, and heart valves using cells, biomaterials as scaffolds, biochemical and physical stimuli. The latter can be induced using a bioreactor mimicking the heart's physiological environment. An extensive review of bioreactors providing perfusion, mechanical and electrical stimulation, as well as the combination of them is provided. An analysis of the stimulations' mechanisms and modes that best suit cardiac construct culture is developed. Finally, we provide insights into bioreactor configuration and culture assessment properties that need to be elucidated for its clinical translation.
Collapse
Affiliation(s)
- Jorge Bravo-Olín
- Biological Engineering Undergraduate Program, Division of Natural Science and Engineering, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México
| | - Sabina A Martínez-Carreón
- Biological Engineering Undergraduate Program, Division of Natural Science and Engineering, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México
| | - Emmanuel Francisco-Solano
- Natural Science and Engineering Graduate Program, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México
| | - Alvaro R Lara
- Department of Biological and Chemical Engineering, Aarhus University, Gustav Wieds Vej 10, 8000, Aarhus, Denmark
| | - Nohra E Beltran-Vargas
- Process and Technology Department, Division of Natural Science and Engineering, Universidad Autonoma Metropolitana-Cuajimalpa, Ciudad de Mexico C.P. 05348, México.
| |
Collapse
|
9
|
Martín D, Ruano D, Yúfera A, Daza P. Electrical pulse stimulation parameters modulate N2a neuronal differentiation. Cell Death Discov 2024; 10:49. [PMID: 38272891 PMCID: PMC10810886 DOI: 10.1038/s41420-024-01820-y] [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: 08/01/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Electrical pulse stimulation has been used to enhance the differentiation or proliferation of neuronal progenitor cells in tissue engineering and cancer treatment. Therefore, a comprehensive investigation of the effects caused by its parameters is crucial for improvements in those fields. We propose a study of pulse parameters, to allow the control of N2a cell line fate and behavior. We have focused on designing an experimental setup that allows for the knowledge and control over the environment and the stimulation signals applied. To map the effects of the stimulation on N2a cells, their morphology and the cellular and molecular reactions induced by the pulse stimulation have been analyzed. Immunofluorescence, rt-PCR and western blot analysis have been carried out for this purpose, as well as cell counting. Our results show that low-amplitude electrical pulse stimulation promotes proliferation of N2a cells, whilst amplitudes in the range 250 mV/mm-500 mV/mm induce differentiation. Amplitudes higher than 750 mV/mm produce cell damage at low frequencies. For high frequencies, large amplitudes are needed to cause cell death. An inverse relation has been found between cell density and pulse-induced neuronal differentiation. The best condition for neuronal differentiation was found to be 500 mV/mm at 100 Hz. These findings have been confirmed by up-regulation of the Neurod1 gene. Our preliminary study of the molecular effects of electrical pulse stimulation on N2a offers premonitory clues of the PI3K/Akt/GSK-3β pathway implications on the neuronal differentiation process through ES. In general, we have successfully mapped the sensitivity of N2a cells to electrical pulse stimulation parameters.
Collapse
Affiliation(s)
- Daniel Martín
- Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain.
- Instituto de Microelectrónica de Sevilla (IMSE), Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain.
| | - Diego Ruano
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain
| | - Alberto Yúfera
- Instituto de Microelectrónica de Sevilla (IMSE), Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain
- Departamento de Tecnología Electrónica, ETSII, Universidad de Sevilla, Sevilla, Spain
| | - Paula Daza
- Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
10
|
Vo QD, Saito Y, Nakamura K, Iida T, Yuasa S. Induced Pluripotent Stem Cell-Derived Cardiomyocytes Therapy for Ischemic Heart Disease in Animal Model: A Meta-Analysis. Int J Mol Sci 2024; 25:987. [PMID: 38256060 PMCID: PMC10815661 DOI: 10.3390/ijms25020987] [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: 12/08/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Ischemic heart disease (IHD) poses a significant challenge in cardiovascular health, with current treatments showing limited success. Induced pluripotent derived-cardiomyocyte (iPSC-CM) therapy within regenerative medicine offers potential for IHD patients, although its clinical impacts remain uncertain. This study utilizes meta-analysis to assess iPSC-CM outcomes in terms of efficacy and safety in IHD animal model studies. A meta-analysis encompassing PUBMED, ScienceDirect, Web of Science, and the Cochrane Library databases, from inception until October 2023, investigated iPSC therapy effects on cardiac function and safety outcomes. Among 51 eligible studies involving 1012 animals, despite substantial heterogeneity, the iPSC-CM transplantation improved left ventricular ejection fraction (LVEF) by 8.23% (95% CI, 7.15 to 9.32%; p < 0.001) compared to control groups. Additionally, cell-based treatment reduced the left ventricle fibrosis area and showed a tendency to reduce left ventricular end-systolic volume (LVESV) and end-diastolic volume (LVEDV). No significant differences emerged in mortality and arrhythmia risk between iPSC-CM treatment and control groups. In conclusion, this meta-analysis indicates iPSC-CM therapy's promise as a safe and beneficial intervention for enhancing heart function in IHD. However, due to observed heterogeneity, the efficacy of this treatment must be further explored through large randomized controlled trials based on rigorous research design.
Collapse
Affiliation(s)
- Quan Duy Vo
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Kazufumi Nakamura
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Toshihiro Iida
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| | - Shinsuke Yuasa
- Department of Cardiovascular Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, Japan; (Q.D.V.); (T.I.); (S.Y.)
| |
Collapse
|
11
|
Sharif NA. Electrical, Electromagnetic, Ultrasound Wave Therapies, and Electronic Implants for Neuronal Rejuvenation, Neuroprotection, Axonal Regeneration, and IOP Reduction. J Ocul Pharmacol Ther 2023; 39:477-498. [PMID: 36126293 DOI: 10.1089/jop.2022.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The peripheral nervous system (PNS) of mammals and nervous systems of lower organisms possess significant regenerative potential. In contrast, although neural plasticity can provide some compensation, the central nervous system (CNS) neurons and nerves of adult mammals generally fail to regenerate after an injury or damage. However, use of diverse electrical, electromagnetic and sonographic energy waves are illuminating novel ways to stimulate neuronal differentiation, proliferation, neurite growth, and axonal elongation/regeneration leading to various levels of functional recovery in animals and humans afflicted with disorders of the CNS, PNS, retina, and optic nerve. Tools such as acupuncture, electroacupuncture, electroshock therapy, electrical stimulation, transcranial magnetic stimulation, red light therapy, and low-intensity pulsed ultrasound therapy are demonstrating efficacy in treating many different maladies. These include wound healing, partial recovery from motor dysfunctions, recovery from ischemic/reperfusion insults and CNS and ocular remyelination, retinal ganglion cell (RGC) rejuvenation, and RGC axonal regeneration. Neural rejuvenation and axonal growth/regeneration processes involve activation or intensifying of the intrinsic bioelectric waves (action potentials) that exist in every neuronal circuit of the body. In addition, reparative factors released at the nerve terminals and via neuronal dendrites (transmitter substances), extracellular vesicles containing microRNAs and neurotrophins, and intercellular communication occurring via nanotubes aid in reestablishing lost or damaged connections between the traumatized tissues and the PNS and CNS. Many other beneficial effects of the aforementioned treatment paradigms are mediated via gene expression alterations such as downregulation of inflammatory and death-signal genes and upregulation of neuroprotective and cytoprotective genes. These varied techniques and technologies will be described and discussed covering cell-based and animal model-based studies. Data from clinical applications and linkage to human ocular diseases will also be discussed where relevant translational research has been reported.
Collapse
Affiliation(s)
- Najam A Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, California, USA
- Singapore Eye Research Institute (SERI), Singapore
- SingHealth Duke-NUS Ophthalmology and Visual Sciences Academic Clinical Programme, Duke-National University of Singapore Medical School, Singapore
- Department of Surgery and Cancer, Imperial College of Science and Technology, London, United Kingdom
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Sciences Center, Fort Worth, Texas, USA
- Department of Pharmacy Sciences, Creighton University, Omaha, Nebraska, USA
- Insitute of Ophthalmology, University College London (UCL), London, United Kingdom
| |
Collapse
|
12
|
Yang H, Yang Y, Kiskin FN, Shen M, Zhang JZ. Recent advances in regulating the proliferation or maturation of human-induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Res Ther 2023; 14:228. [PMID: 37649113 PMCID: PMC10469435 DOI: 10.1186/s13287-023-03470-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023] Open
Abstract
In the last decade, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM)-based cell therapy has drawn broad attention as a potential therapy for treating injured hearts. However, mass production of hiPSC-CMs remains challenging, limiting their translational potential in regenerative medicine. Therefore, multiple strategies including cell cycle regulators, small molecules, co-culture systems, and epigenetic modifiers have been used to improve the proliferation of hiPSC-CMs. On the other hand, the immaturity of these proliferative hiPSC-CMs could lead to lethal arrhythmias due to their limited ability to functionally couple with resident cardiomyocytes. To achieve functional maturity, numerous methods such as prolonged culture, biochemical or biophysical stimulation, in vivo transplantation, and 3D culture approaches have been employed. In this review, we summarize recent approaches used to promote hiPSC-CM proliferation, and thoroughly review recent advances in promoting hiPSC-CM maturation, which will serve as the foundation for large-scale production of mature hiPSC-CMs for future clinical applications.
Collapse
Affiliation(s)
- Hao Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Yuan Yang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Fedir N Kiskin
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Mengcheng Shen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Joe Z Zhang
- Institute of Neurological and Psychiatric Disorders, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
13
|
Bui TA, Stafford N, Oceandy D. Genetic and Pharmacological YAP Activation Induces Proliferation and Improves Survival in Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Cells 2023; 12:2121. [PMID: 37681853 PMCID: PMC10487209 DOI: 10.3390/cells12172121] [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: 06/13/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Cardiomyocyte loss following myocardial infarction cannot be addressed with current clinical therapies. Cell therapy with induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) is a potential approach to replace cardiomyocyte loss. However, engraftment rates in pre-clinical studies have been low, highlighting a need to refine current iPSC-CM technology. In this study, we demonstrated that inducing Yes-associated protein (YAP) by genetic and pharmacological approaches resulted in increased iPSC-CM proliferation and reduced apoptosis in response to oxidative stress. Interestingly, iPSC-CM maturation was differently affected by each strategy, with genetic activation of YAP resulting in a more immature cardiomyocyte-like phenotype not witnessed upon pharmacological YAP activation. Overall, we conclude that YAP activation in iPSC-CMs enhances cell survival and proliferative capacity. Therefore, strategies targeting YAP, or its upstream regulator the Hippo signalling pathway, could potentially be used to improve the efficacy of iPSC-CM technology for use as a future regenerative therapy in myocardial infarction.
Collapse
Affiliation(s)
| | | | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK; (T.A.B.); (N.S.)
| |
Collapse
|
14
|
Zhuo D, Lei I, Li W, Liu L, Li L, Ni J, Liu Z, Fan G. The origin, progress, and application of cell-based cardiac regeneration therapy. J Cell Physiol 2023; 238:1732-1755. [PMID: 37334836 DOI: 10.1002/jcp.31060] [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: 03/17/2023] [Revised: 05/08/2023] [Accepted: 05/29/2023] [Indexed: 06/21/2023]
Abstract
Cardiovascular disease (CVD) has become a severe threat to human health, with morbidity and mortality increasing yearly and gradually becoming younger. When the disease progresses to the middle and late stages, the loss of a large number of cardiomyocytes is irreparable to the body itself, and clinical drug therapy and mechanical support therapy cannot reverse the development of the disease. To explore the source of regenerated myocardium in model animals with the ability of heart regeneration through lineage tracing and other methods, and develop a new alternative therapy for CVDs, namely cell therapy. It directly compensates for cardiomyocyte proliferation through adult stem cell differentiation or cell reprogramming, which indirectly promotes cardiomyocyte proliferation through non-cardiomyocyte paracrine, to play a role in heart repair and regeneration. This review comprehensively summarizes the origin of newly generated cardiomyocytes, the research progress of cardiac regeneration based on cell therapy, the opportunity and development of cardiac regeneration in the context of bioengineering, and the clinical application of cell therapy in ischemic diseases.
Collapse
Affiliation(s)
- Danping Zhuo
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ienglam Lei
- Department of Cardiac Surgery, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Wenjun Li
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Li Liu
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jingyu Ni
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihao Liu
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
15
|
Zhou X, Li G, Wu D, Liang H, Zhang W, Zeng L, Zhu Q, Lai P, Wen Z, Yang C, Pan Y. Recent advances of cellular stimulation with triboelectric nanogenerators. EXPLORATION (BEIJING, CHINA) 2023; 3:20220090. [PMID: 37933231 PMCID: PMC10624380 DOI: 10.1002/exp.20220090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 12/06/2022] [Indexed: 11/08/2023]
Abstract
Triboelectric nanogenerators (TENGs) are new energy collection devices that have the characteristics of high efficiency, low cost, miniaturization capability, and convenient manufacture. TENGs mainly utilize the triboelectric effect to obtain mechanical energy from organisms or the environment, and this mechanical energy is then converted into and output as electrical energy. Bioelectricity is a phenomenon that widely exists in various cellular processes, including cell proliferation, senescence, apoptosis, as well as adjacent cells' communication and coordination. Therefore, based on these features, TENGs can be applied in organisms to collect energy and output electrical stimulation to act on cells, changing their activities and thereby playing a role in regulating cellular function and interfering with cellular fate, which can further develop into new methods of health care and disease intervention. In this review, we first introduce the working principle of TENGs and their working modes, and then summarize the current research status of cellular function regulation and fate determination stimulated by TENGs, and also analyze their application prospects for changing various processes of cell activity. Finally, we discuss the opportunities and challenges of TENGs in the fields of life science and biomedical engineering, and propose a variety of possibilities for their potential development direction.
Collapse
Affiliation(s)
- Xingyu Zhou
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Gaocai Li
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Di Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Huaizhen Liang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Weifeng Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lingli Zeng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Qianqian Zhu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouChina
| | - Puxiang Lai
- Department of Biomedical EngineeringHong Kong Polytechnic UniversityHong KongChina
| | - Zhen Wen
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouChina
| | - Cao Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong‐Hong Kong Joint Laboratory for RNA MedicineMedical Research Center, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
16
|
Licata JP, Schwab KH, Har-El YE, Gerstenhaber JA, Lelkes PI. Bioreactor Technologies for Enhanced Organoid Culture. Int J Mol Sci 2023; 24:11427. [PMID: 37511186 PMCID: PMC10380004 DOI: 10.3390/ijms241411427] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
An organoid is a 3D organization of cells that can recapitulate some of the structure and function of native tissue. Recent work has seen organoids gain prominence as a valuable model for studying tissue development, drug discovery, and potential clinical applications. The requirements for the successful culture of organoids in vitro differ significantly from those of traditional monolayer cell cultures. The generation and maturation of high-fidelity organoids entails developing and optimizing environmental conditions to provide the optimal cues for growth and 3D maturation, such as oxygenation, mechanical and fluidic activation, nutrition gradients, etc. To this end, we discuss the four main categories of bioreactors used for organoid culture: stirred bioreactors (SBR), microfluidic bioreactors (MFB), rotating wall vessels (RWV), and electrically stimulating (ES) bioreactors. We aim to lay out the state-of-the-art of both commercial and in-house developed bioreactor systems, their benefits to the culture of organoids derived from various cells and tissues, and the limitations of bioreactor technology, including sterilization, accessibility, and suitability and ease of use for long-term culture. Finally, we discuss future directions for improvements to existing bioreactor technology and how they may be used to enhance organoid culture for specific applications.
Collapse
Affiliation(s)
- Joseph P Licata
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Kyle H Schwab
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yah-El Har-El
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Jonathan A Gerstenhaber
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
17
|
Vuorenpää H, Björninen M, Välimäki H, Ahola A, Kroon M, Honkamäki L, Koivumäki JT, Pekkanen-Mattila M. Building blocks of microphysiological system to model physiology and pathophysiology of human heart. Front Physiol 2023; 14:1213959. [PMID: 37485060 PMCID: PMC10358860 DOI: 10.3389/fphys.2023.1213959] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
Microphysiological systems (MPS) are drawing increasing interest from academia and from biomedical industry due to their improved capability to capture human physiology. MPS offer an advanced in vitro platform that can be used to study human organ and tissue level functions in health and in diseased states more accurately than traditional single cell cultures or even animal models. Key features in MPS include microenvironmental control and monitoring as well as high biological complexity of the target tissue. To reach these qualities, cross-disciplinary collaboration from multiple fields of science is required to build MPS. Here, we review different areas of expertise and describe essential building blocks of heart MPS including relevant cardiac cell types, supporting matrix, mechanical stimulation, functional measurements, and computational modelling. The review presents current methods in cardiac MPS and provides insights for future MPS development with improved recapitulation of human physiology.
Collapse
Affiliation(s)
- Hanna Vuorenpää
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Miina Björninen
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hannu Välimäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti Ahola
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mart Kroon
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Honkamäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jussi T. Koivumäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
18
|
Haridhasapavalan KK, Borthakur A, Thummer RP. Direct Cardiac Reprogramming: Current Status and Future Prospects. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1436:1-18. [PMID: 36662416 DOI: 10.1007/5584_2022_760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Advances in cellular reprogramming articulated the path for direct cardiac lineage conversion, bypassing the pluripotent state. Direct cardiac reprogramming attracts major attention because of the low or nil regenerative ability of cardiomyocytes, resulting in permanent cell loss in various heart diseases. In the field of cardiology, balancing this loss of cardiomyocytes was highly challenging, even in the modern medical world. Soon after the discovery of cell reprogramming, direct cardiac reprogramming also became a promising alternative for heart regeneration. This review mainly focused on the various direct cardiac reprogramming approaches (integrative and non-integrative) for the derivation of induced autologous cardiomyocytes. It also explains the advancements in cardiac reprogramming over the decade with the pros and cons of each approach. Further, the review highlights the importance of clinically relevant (non-integrative) approaches and their challenges for the prospective applications for personalized medicine. Apart from direct cardiac reprogramming, it also discusses the other strategies for generating cardiomyocytes from different sources. The understanding of these strategies could pave the way for the efficient generation of integration-free functional autologous cardiomyocytes through direct cardiac reprogramming for various biomedical applications.
Collapse
Affiliation(s)
- Krishna Kumar Haridhasapavalan
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Atreyee Borthakur
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
19
|
Zhu K, Bao X, Wang Y, Lu T, Zhang L. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocyte modelling of cardiovascular diseases for natural compound discovery. Biomed Pharmacother 2023; 157:113970. [PMID: 36371854 DOI: 10.1016/j.biopha.2022.113970] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022] Open
Abstract
Cardiovascular disease (CVD) remains the leading cause of death worldwide. Natural compounds extracted from medicinal plants characterized by diverse biological activities and low toxicity or side effects, are increasingly taking center stage in the search for new drugs. Currently, preclinical evaluation of natural products relies mainly on the use of immortalized cell lines of human origin or animal models. Increasing evidence indicates that cardiomyopathy models based on immortalized cell lines do not recapitulate pathogenic phenotypes accurately and a substantial physiological discrepancy between animals and humans casts doubt on the clinical relevance of animal models for these studies. The newly developed human induced pluripotent stem cell (hiPSC) technology in combination with highly-efficient cardiomyocyte differentiation methods provides an ideal tool for modeling human cardiomyopathies in vitro. Screening of drugs, especially screening of natural products, based on these models has been widely used and has shown that evaluation in such models can recapitulate important aspects of the physiological properties of drugs. The purpose of this review is to provide information on the latest developments in this area of research and to help researchers perform screening of natural products using the hiPSC-CM platform.
Collapse
Affiliation(s)
- Keyang Zhu
- Zhejiang Key Laboratory of Pathophysiology, School of Public Health, School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Xiaoming Bao
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China; Department of Global Health, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, PR China
| | - Yingchao Wang
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Ting Lu
- Clinical Research Center of The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.
| | - Ling Zhang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
20
|
He X, Liang J, Paul C, Huang W, Dutta S, Wang Y. Advances in Cellular Reprogramming-Based Approaches for Heart Regenerative Repair. Cells 2022; 11:3914. [PMID: 36497171 PMCID: PMC9740402 DOI: 10.3390/cells11233914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/18/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Continuous loss of cardiomyocytes (CMs) is one of the fundamental characteristics of many heart diseases, which eventually can lead to heart failure. Due to the limited proliferation ability of human adult CMs, treatment efficacy has been limited in terms of fully repairing damaged hearts. It has been shown that cell lineage conversion can be achieved by using cell reprogramming approaches, including human induced pluripotent stem cells (hiPSCs), providing a promising therapeutic for regenerative heart medicine. Recent studies using advanced cellular reprogramming-based techniques have also contributed some new strategies for regenerative heart repair. In this review, hiPSC-derived cell therapeutic methods are introduced, and the clinical setting challenges (maturation, engraftment, immune response, scalability, and tumorigenicity), with potential solutions, are discussed. Inspired by the iPSC reprogramming, the approaches of direct cell lineage conversion are merging, such as induced cardiomyocyte-like cells (iCMs) and induced cardiac progenitor cells (iCPCs) derived from fibroblasts, without induction of pluripotency. The studies of cellular and molecular pathways also reveal that epigenetic resetting is the essential mechanism of reprogramming and lineage conversion. Therefore, CRISPR techniques that can be repurposed for genomic or epigenetic editing become attractive approaches for cellular reprogramming. In addition, viral and non-viral delivery strategies that are utilized to achieve CM reprogramming will be introduced, and the therapeutic effects of iCMs or iCPCs on myocardial infarction will be compared. After the improvement of reprogramming efficiency by developing new techniques, reprogrammed iCPCs or iCMs will provide an alternative to hiPSC-based approaches for regenerative heart therapies, heart disease modeling, and new drug screening.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jialiang Liang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Christian Paul
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Wei Huang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Yigang Wang
- Department of Pathology & Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
21
|
Wang Y, Xue Y, Guo HD. Intervention effects of traditional Chinese medicine on stem cell therapy of myocardial infarction. Front Pharmacol 2022; 13:1013740. [PMID: 36330092 PMCID: PMC9622800 DOI: 10.3389/fphar.2022.1013740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular diseases are the leading cause of global mortality, in which myocardial infarction accounts for 46% of total deaths. Although good progress has been achieved in medication and interventional techniques, a proven method to repair the damaged myocardium has not yet been determined. Stem cell therapy for damaged myocardial repair has evolved into a promising treatment for ischemic heart disease. However, low retention and poor survival of the injected stem cells are the major obstacles to achieving the intended therapeutic effects. Chinese botanical and other natural drug substances are a rich source of effective treatment for various diseases. As such, numerous studies have revealed the role of Chinese medicine in stem cell therapy for myocardial infarction treatment, including promoting proliferation, survival, migration, angiogenesis, and differentiation of stem cells. Here, we discuss the potential and limitations of stem cell therapy, as well as the regulatory mechanism of Chinese medicines underlying stem cell therapy. We focus on the evidence from pre-clinical trials and clinical practices, and based on traditional Chinese medicine theories, we further summarize the mechanisms of Chinese medicine treatment in stem cell therapy by the commonly used prescriptions. Despite the pre-clinical evidence showing that traditional Chinese medicine is helpful in stem cell therapy, there are still some limitations of traditional Chinese medicine therapy. We also systematically assess the detailed experimental design and reliability of included pharmacological research in our review. Strictly controlled animal models with multi-perspective pharmacokinetic profiles and high-grade clinical evidence with multi-disciplinary efforts are highly demanded in the future.
Collapse
Affiliation(s)
- Yu Wang
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuezhen Xue
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hai-dong Guo
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Anatomy, School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
Martín D, Bocio-Nuñez J, Scagliusi SF, Pérez P, Huertas G, Yúfera A, Giner M, Daza P. DC electrical stimulation enhances proliferation and differentiation on N2a and MC3T3 cell lines. J Biol Eng 2022; 16:27. [PMID: 36229846 PMCID: PMC9563743 DOI: 10.1186/s13036-022-00306-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Background Electrical stimulation is a novel tool to promote the differentiation and proliferation of precursor cells. In this work we have studied the effects of direct current (DC) electrical stimulation on neuroblastoma (N2a) and osteoblast (MC3T3) cell lines as a model for nervous and bone tissue regeneration, respectively. We have developed the electronics and encapsulation of a proposed stimulation system and designed a setup and protocol to stimulate cell cultures. Methods Cell cultures were subjected to several assays to assess the effects of electrical stimulation on them. N2a cells were analyzed using microscope images and an inmunofluorescence assay, differentiated cells were counted and neurites were measured. MC3T3 cells were subjected to an AlamarBlue assay for viability, ALP activity was measured, and a real time PCR was carried out. Results Our results show that electrically stimulated cells had more tendency to differentiate in both cell lines when compared to non-stimulated cultures, paired with a promotion of neurite growth and polarization in N2a cells and an increase in proliferation in MC3T3 cell line. Conclusions These results prove the effectiveness of electrical stimulation as a tool for tissue engineering and regenerative medicine, both for neural and bone injuries. Bone progenitor cells submitted to electrical stimulation have a higher tendency to differentiate and proliferate, filling the gaps present in injuries. On the other hand, neuronal progenitor cells differentiate, and their neurites can be polarized to follow the electric field applied.
Collapse
Affiliation(s)
- Daniel Martín
- Electronics Technology Department, Universidad de Sevilla, Seville, Spain. .,Microelectronics Institute of Seville, Universidad de Sevilla, Seville, Spain.
| | - J Bocio-Nuñez
- Bone Metabolism Unit, UGC Medicina Interna, HUV Macarena, Seville, Spain
| | - Santiago F Scagliusi
- Electronics Technology Department, Universidad de Sevilla, Seville, Spain.,Microelectronics Institute of Seville, Universidad de Sevilla, Seville, Spain
| | - Pablo Pérez
- Electronics Technology Department, Universidad de Sevilla, Seville, Spain.,Microelectronics Institute of Seville, Universidad de Sevilla, Seville, Spain
| | - Gloria Huertas
- Microelectronics Institute of Seville, Universidad de Sevilla, Seville, Spain.,Electronics and Electromagnetism Department, Universidad de Sevilla, Seville, Spain
| | - Alberto Yúfera
- Electronics Technology Department, Universidad de Sevilla, Seville, Spain.,Microelectronics Institute of Seville, Universidad de Sevilla, Seville, Spain
| | - Mercè Giner
- Departamento de Citologia e Histologia Normal y Patologica, Universidad de Sevilla, Seville, Spain
| | - Paula Daza
- Cell Biology Department, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
23
|
Leowattana W, Leowattana T, Leowattana P. Human-induced pluripotent stem cell-atrial-specific cardiomyocytes and atrial fibrillation. World J Clin Cases 2022; 10:9588-9601. [PMID: 36186184 PMCID: PMC9516943 DOI: 10.12998/wjcc.v10.i27.9588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/22/2022] [Accepted: 08/16/2022] [Indexed: 02/05/2023] Open
Abstract
Patient-specific human-induced pluripotent stem cell-derived atrial cardiomyocytes (hiPSC-aCMs) may be produced, genome-edited, and differentiated into multiple cell types for regenerative medicine, disease modeling, drug testing, toxicity screening, and three-dimensional tissue fabrication. There is presently no complete model of atrial fibrillation (AF) available for studying human pharmacological responses and evaluating the toxicity of potential medication candidates. It has been demonstrated that hiPSC-aCMs can replicate the electrophysiological disease phenotype and genotype of AF. The hiPSC-aCMs, however, are immature and do not reflect the maturity of aCMs in the native myocardium. Numerous laboratories utilize a variety of methodologies and procedures to improve and promote aCM maturation, including electrical stimulation, culture duration, biophysical signals, and changes in metabolic variables. This review covers the current methods being explored for use in the maturation of patient-specific hiPSC-aCMs and their application towards a personalized approach to the pharmacologic therapy of AF.
Collapse
Affiliation(s)
- Wattana Leowattana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Tawithep Leowattana
- Department of Medicine, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
| | - Pathomthep Leowattana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
24
|
Basara G, Bahcecioglu G, Ozcebe SG, Ellis BW, Ronan G, Zorlutuna P. Myocardial infarction from a tissue engineering and regenerative medicine point of view: A comprehensive review on models and treatments. BIOPHYSICS REVIEWS 2022; 3:031305. [PMID: 36091931 PMCID: PMC9447372 DOI: 10.1063/5.0093399] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 05/12/2023]
Abstract
In the modern world, myocardial infarction is one of the most common cardiovascular diseases, which are responsible for around 18 million deaths every year or almost 32% of all deaths. Due to the detrimental effects of COVID-19 on the cardiovascular system, this rate is expected to increase in the coming years. Although there has been some progress in myocardial infarction treatment, translating pre-clinical findings to the clinic remains a major challenge. One reason for this is the lack of reliable and human representative healthy and fibrotic cardiac tissue models that can be used to understand the fundamentals of ischemic/reperfusion injury caused by myocardial infarction and to test new drugs and therapeutic strategies. In this review, we first present an overview of the anatomy of the heart and the pathophysiology of myocardial infarction, and then discuss the recent developments on pre-clinical infarct models, focusing mainly on the engineered three-dimensional cardiac ischemic/reperfusion injury and fibrosis models developed using different engineering methods such as organoids, microfluidic devices, and bioprinted constructs. We also present the benefits and limitations of emerging and promising regenerative therapy treatments for myocardial infarction such as cell therapies, extracellular vesicles, and cardiac patches. This review aims to overview recent advances in three-dimensional engineered infarct models and current regenerative therapeutic options, which can be used as a guide for developing new models and treatment strategies.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - S. Gulberk Ozcebe
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - George Ronan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Pinar Zorlutuna
- Present address: 143 Multidisciplinary Research Building, University of Notre Dame, Notre Dame, IN 46556. Author to whom correspondence should be addressed:. Tel.: +1 574 631 8543. Fax: +1 574 631 8341
| |
Collapse
|
25
|
Wang Y, Yu M, Hao K, Lei W, Tang M, Hu S. Cardiomyocyte Maturation-the Road is not Obstructed. Stem Cell Rev Rep 2022; 18:2966-2981. [PMID: 35788883 DOI: 10.1007/s12015-022-10407-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 12/29/2022]
Abstract
Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) represent one of the most promising ways to treat cardiovascular diseases. High-purity cardiomyocytes (CM) from different cell sources could be obtained at present. However, the immature nature of these cardiomyocytes hinders its further clinical application. From immature to mature state, it involves structural, functional, and metabolic changes in cardiomyocytes. Generally, two types of culturing (2D and 3D) systems have been reported to induce cardiomyocyte maturation. 2D culture mainly achieves the maturation of cardiomyocytes through long-term culture, co-culture, supplementation of small molecule compounds, and the application of biophysical cues. The combined use of biomaterial's surface topography and biophysical cues also facilitates the maturation of cardiomyocytes. Cardiomyocyte maturation is a complex process involving many signaling pathways, and current methods fail to fully reproduce this process. Therefore, analyzing the signaling pathway network related to the maturation and producing hPSC-CMs with adult-like phenotype is a challenge. In this review, we summarized the structural and functional differences between hPSC-CMs and mature cardiomyocytes, and introduced various methods to induce cardiomyocyte maturation.
Collapse
Affiliation(s)
- Yaning Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Miao Yu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Kaili Hao
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Wei Lei
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China
| | - Mingliang Tang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, 215000, China.
| |
Collapse
|
26
|
Afjeh-Dana E, Naserzadeh P, Moradi E, Hosseini N, Seifalian AM, Ashtari B. Stem Cell Differentiation into Cardiomyocytes: Current Methods and Emerging Approaches. Stem Cell Rev Rep 2022; 18:2566-2592. [PMID: 35508757 DOI: 10.1007/s12015-021-10280-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2021] [Indexed: 12/26/2022]
Abstract
Cardiovascular diseases (CVDs) are globally known to be important causes of mortality and disabilities. Common treatment strategies for CVDs, such as pharmacological therapeutics impose serious challenges due to the failure of treatments for myocardial necrosis. By contrast, stem cells (SCs) based therapies are seen to be promising approaches to CVDs treatment. In such approaches, cardiomyocytes are differentiated from SCs. To fulfill SCs complete potential, the method should be appointed to generate cardiomyocytes with more mature structure and well-functioning operations. For heart repairing applications, a greatly scalable and medical-grade cardiomyocyte generation must be used. Nonetheless, there are some challenges such as immune rejection, arrhythmogenesis, tumorigenesis, and graft cell death potential. Herein, we discuss the types of potential SCs, and commonly used methods including embryoid bodies related techniques, co-culture, mechanical stimulation, and electrical stimulation and their applications, advantages and limitations in this field. An estimated 17.9 million people died from CVDs in 2019, representing 32 % of all global deaths. Of these deaths, 85 % were due to heart attack and stroke.
Collapse
Affiliation(s)
- Elham Afjeh-Dana
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Parvaneh Naserzadeh
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Moradi
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Nasrin Hosseini
- Neuroscience Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| | - Alexander Marcus Seifalian
- Nanotechnology & Regenerative Medicine Commercialisation Centre (NanoRegMed Ltd), London BioScience Innovation Centre, London, UK
| | - Behnaz Ashtari
- Radiation Biology Research Centre, Iran University of Medical Sciences, Tehran, Iran. .,Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran. .,Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Iberite F, Gruppioni E, Ricotti L. Skeletal muscle differentiation of human iPSCs meets bioengineering strategies: perspectives and challenges. NPJ Regen Med 2022; 7:23. [PMID: 35393412 PMCID: PMC8991236 DOI: 10.1038/s41536-022-00216-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 03/01/2022] [Indexed: 12/31/2022] Open
Abstract
Although skeletal muscle repairs itself following small injuries, genetic diseases or severe damages may hamper its ability to do so. Induced pluripotent stem cells (iPSCs) can generate myogenic progenitors, but their use in combination with bioengineering strategies to modulate their phenotype has not been sufficiently investigated. This review highlights the potential of this combination aimed at pushing the boundaries of skeletal muscle tissue engineering. First, the overall organization and the key steps in the myogenic process occurring in vivo are described. Second, transgenic and non-transgenic approaches for the myogenic induction of human iPSCs are compared. Third, technologies to provide cells with biophysical stimuli, biomaterial cues, and biofabrication strategies are discussed in terms of recreating a biomimetic environment and thus helping to engineer a myogenic phenotype. The embryonic development process and the pro-myogenic role of the muscle-resident cell populations in co-cultures are also described, highlighting the possible clinical applications of iPSCs in the skeletal muscle tissue engineering field.
Collapse
Affiliation(s)
- Federica Iberite
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy. .,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.
| | - Emanuele Gruppioni
- Centro Protesi INAIL, Istituto Nazionale per l'Assicurazione contro gli Infortuni sul Lavoro, 40054, Vigorso di Budrio (BO), Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy.,Department of Excellence in Robotics & AI, Scuola Superiore Sant'Anna, 56127, Pisa (PI), Italy
| |
Collapse
|
28
|
Li Y, Wei L, Lan L, Gao Y, Zhang Q, Dawit H, Mao J, Guo L, Shen L, Wang L. Conductive biomaterials for cardiac repair: A review. Acta Biomater 2022; 139:157-178. [PMID: 33887448 DOI: 10.1016/j.actbio.2021.04.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/26/2021] [Accepted: 04/10/2021] [Indexed: 12/18/2022]
Abstract
Myocardial infarction (MI) is one of the fatal diseases in humans. Its incidence is constantly increasing annually all over the world. The problem is accompanied by the limited regenerative capacity of cardiomyocytes, yielding fibrous scar tissue formation. The propagation of electrical impulses in such tissue is severely hampered, negatively influencing the normal heart pumping function. Thus, reconstruction of the internal cardiac electrical connection is currently a major concern of myocardial repair. Conductive biomaterials with or without cell loading were extensively investigated to address this problem. This article introduces a detailed overview of the recent progress in conductive biomaterials and fabrication methods of conductive scaffolds for cardiac repair. After that, the advances in myocardial tissue construction in vitro by the restoration of intercellular communication and simulation of the dynamic electrophysiological environment are systematically reviewed. Furthermore, the latest trend in the study of cardiac repair in vivo using various conductive patches is summarized. Finally, we discuss the achievements and shortcomings of the existing conductive biomaterials and the properties of an ideal conductive patch for myocardial repair. We hope this review will help readers understand the importance and usefulness of conductive biomaterials in cardiac repair and inspire researchers to design and develop new conductive patches to meet the clinical requirements. STATEMENT OF SIGNIFICANCE: After myocardial infarction, the infarcted myocardial area is gradually replaced by heterogeneous fibrous tissue with inferior conduction properties, resulting in arrhythmia and heart remodeling. Conductive biomaterials have been extensively adopted to solve the problem. Summarizing the relevant literature, this review presents an overview of the types and fabrication methods of conductive biomaterials, and focally discusses the recent advances in myocardial tissue construction in vitro and myocardial repair in vivo, which is rarely covered in previous reviews. As well, the deficiencies of the existing conductive patches and their construction strategies for myocardial repair are discussed as well as the improving directions. Confidently, the readers of this review would appreciate advantages and current limitations of conductive biomaterials/patches in cardiac repair.
Collapse
Affiliation(s)
- Yimeng Li
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Leqian Wei
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Lizhen Lan
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Yaya Gao
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Qian Zhang
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Hewan Dawit
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| | - Jifu Mao
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China.
| | - Lamei Guo
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China
| | - Li Shen
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| | - Lu Wang
- Key Laboratory of Textile Science & Technology of Ministry of Education and College of Textiles, Donghua University, Shanghai, 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai, 201620, China
| |
Collapse
|
29
|
Šafaříková E, Ehlich J, Stříteský S, Vala M, Weiter M, Pacherník J, Kubala L, Víteček J. Conductive Polymer PEDOT:PSS-Based Platform for Embryonic Stem-Cell Differentiation. Int J Mol Sci 2022; 23:ijms23031107. [PMID: 35163031 PMCID: PMC8835127 DOI: 10.3390/ijms23031107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/13/2022] [Accepted: 01/17/2022] [Indexed: 01/12/2023] Open
Abstract
Organic semiconductors are constantly gaining interest in regenerative medicine. Their tunable physico-chemical properties, including electrical conductivity, are very promising for the control of stem-cell differentiation. However, their use for combined material-based and electrical stimulation remains largely underexplored. Therefore, we carried out a study on whether a platform based on the conductive polymer poly(3,4-ethylenedioxythiophene):polystyrene sulfonate (PEDOT:PSS) can be beneficial to the differentiation of mouse embryonic stem cells (mESCs). The platform was prepared using the layout of a standard 24-well cell-culture plate. Polyethylene naphthalate foil served as the substrate for the preparation of interdigitated gold electrodes by physical vapor deposition. The PEDOT:PSS pattern was fabricated by precise screen printing over the gold electrodes. The PEDOT:PSS platform was able to produce higher electrical current with the pulsed-direct-current (DC) electrostimulation mode (1 Hz, 200 mV/mm, 100 ms pulse duration) compared to plain gold electrodes. There was a dominant capacitive component. In proof-of-concept experiments, mESCs were able to respond to such electrostimulation by membrane depolarization and elevation of cytosolic calcium. Further, the PEDOT:PSS platform was able to upregulate cardiomyogenesis and potentially inhibit early neurogenesis per se with minor contribution of electrostimulation. Hence, the present work highlights the large potential of PEDOT:PSS in regenerative medicine.
Collapse
Affiliation(s)
- Eva Šafaříková
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; (E.Š.); (L.K.)
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic;
| | - Jiří Ehlich
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Stanislav Stříteský
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Martin Vala
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Martin Weiter
- Faculty of Chemistry, Brno University of Technology, Purkyňova 118, 612 00 Brno, Czech Republic; (J.E.); (S.S.); (M.V.); (M.W.)
| | - Jiří Pacherník
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic;
| | - Lukáš Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; (E.Š.); (L.K.)
- Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic;
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 53, 656 91 Brno, Czech Republic
| | - Jan Víteček
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, 612 65 Brno, Czech Republic; (E.Š.); (L.K.)
- Correspondence: ; Tel./Fax: +420-541-517104; Fax: +420-541-517104
| |
Collapse
|
30
|
Emerging trends and prospects of electroconductive bioinks for cell-laden and functional 3D bioprinting. Biodes Manuf 2022. [DOI: 10.1007/s42242-021-00169-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
31
|
Jiang L, Liang J, Huang W, Ma J, Park KH, Wu Z, Chen P, Zhu H, Ma JJ, Cai W, Paul C, Niu L, Fan GC, Wang HS, Kanisicak O, Xu M, Wang Y. CRISPR activation of endogenous genes reprograms fibroblasts into cardiovascular progenitor cells for myocardial infarction therapy. Mol Ther 2022; 30:54-74. [PMID: 34678511 PMCID: PMC8753567 DOI: 10.1016/j.ymthe.2021.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/27/2021] [Accepted: 10/18/2021] [Indexed: 01/07/2023] Open
Abstract
Fibroblasts can be reprogrammed into cardiovascular progenitor cells (CPCs) using transgenic approaches, although the underlying mechanism remains unclear. We determined whether activation of endogenous genes such as Gata4, Nkx2.5, and Tbx5 can rapidly establish autoregulatory loops and initiate CPC generation in adult extracardiac fibroblasts using a CRISPR activation system. The induced fibroblasts (>80%) showed phenotypic changes as indicated by an Nkx2.5 cardiac enhancer reporter. The progenitor characteristics were confirmed by colony formation and expression of cardiovascular genes. Cardiac sphere induction segregated the early and late reprogrammed cells that can generate functional cardiomyocytes and vascular cells in vitro. Therefore, they were termed CRISPR-induced CPCs (ciCPCs). Transcriptomic analysis showed that cell cycle and heart development pathways were important to accelerate CPC formation during the early reprogramming stage. The CRISPR system opened the silenced chromatin locus, thereby allowing transcriptional factors to access their own promoters and eventually forming a positive feedback loop. The regenerative potential of ciCPCs was assessed after implantation in mouse myocardial infarction models. The engrafted ciCPCs differentiated into cardiovascular cells in vivo but also significantly improved contractile function and scar formation. In conclusion, multiplex gene activation was sufficient to drive CPC reprogramming, providing a new cell source for regenerative therapeutics.
Collapse
Affiliation(s)
- Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Ki Ho Park
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Peng Chen
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jian-Jie Ma
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Wenfeng Cai
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Liang Niu
- Division of Biostatistics and Bioinformatics, Department of Environmental Health, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Hong-Sheng Wang
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA.
| |
Collapse
|
32
|
Otarola G, Hu JC, Athanasiou KA. INTRACELLULAR CALCIUM AND SODIUM MODULATION OF SELF-ASSEMBLED NEOCARTILAGE USING COSTAL CHONDROCYTES. Tissue Eng Part A 2021; 28:595-605. [PMID: 34877888 DOI: 10.1089/ten.tea.2021.0169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ion signaling via Ca2+ and Na+ plays a key role in mechanotransduction and encourages a chondrogenic phenotype and tissue maturation. Here, we propose that the pleiotropic effects of Ca2+ and Na+ modulation can be used to induce maturation and improvement of neocartilage derived from re-differentiated expanded chondrocytes from minipig rib cartilage. Three ion modulators were employed: 1) 4α-phorbol-12,13-didecanoate (4-αPDD), an agonist of the Ca2+-permeable transient receptor potential vanilloid 4 (TRPV4), 2) ouabain, an inhibitor of the Na+/K+ pump, and 3) ionomycin, a Ca2+ ionophore. These ion modulators were used individually or in combination. While no beneficial effects were observed when using combinations of the ion modulators, single treatment of constructs with the three ion modulators resulted in multiple effects in structure-function relationships. The most significant findings were related to ionomycin. Treatment of neocartilage with ionomycin produced 61% and 115% increases in glycosaminoglycan and pyridinoline crosslink content, respectively, compared to the control. Moreover, treatment with this Ca2+ ionophore resulted in a 45% increase of the aggregate modulus, and a 63% increase in the tensile Young's modulus, resulting in aggregate and Young's moduli of 567 kPa and 8.43 MPa, respectively. These results support the use of ion modulation to develop biomimetic neocartilage using expanded re-differentiated costal chondrocytes.
Collapse
Affiliation(s)
- Gaston Otarola
- University of California, Irvine, BME, Irvine, California, United States;
| | - Jerry C Hu
- University of California, Irvine, BME, Irvine, California, United States;
| | | |
Collapse
|
33
|
Yuan M, Li X, Liu J, Zheng Y, Cheng L, Tang N, Zhang R, Xu S, Fu X, Haick H, Xu Y. Fully Integrated Self-Powered Electrical Stimulation Cell Culture Dish for Noncontact High-Efficiency Plasmid Transfection. ACS APPLIED MATERIALS & INTERFACES 2021; 13:54762-54769. [PMID: 34757708 DOI: 10.1021/acsami.1c16748] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Plasmid DNA transfection of mammalian cells is widely used in biomedical research and genetic drug delivery, but low transfection efficiency, especially in the context of the primary cells, limits its application. To improve the efficiency of plasmid transfection, a fully integrated self-powered electrical stimulation cell culture dish (SESD) has been developed to provide self-powered electrical stimulation (ES) of adherent cells, significantly improving the efficiency of plasmid transfection into mammalian cells and cell survival by the standard lipofectamine transfection method. Mechanistically, ES can safely increase the intracellular calcium concentration by opening calcium-ion channels, leading to a higher efficiency of plasmid transfection. Therefore, SESD has the potential to become an effective platform for high-efficiency plasmid DNA transfection in biomedical research and drug delivery.
Collapse
Affiliation(s)
- Miaomiao Yuan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Xiaoying Li
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Jingfeng Liu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Youbin Zheng
- Department of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Li Cheng
- School of Materials and Energy, Lanzhou University, Lanzhou 730000, China
| | - Ning Tang
- Department of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Rongjun Zhang
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Shuxiang Xu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
| | - Xuemei Fu
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Hossam Haick
- Department of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Yang Xu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| |
Collapse
|
34
|
Vitus V, Ibrahim F, Wan Kamarul Zaman WS. Modelling of Stem Cells Microenvironment Using Carbon-Based Scaffold for Tissue Engineering Application-A Review. Polymers (Basel) 2021; 13:4058. [PMID: 34883564 PMCID: PMC8658938 DOI: 10.3390/polym13234058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022] Open
Abstract
A scaffold is a crucial biological substitute designed to aid the treatment of damaged tissue caused by trauma and disease. Various scaffolds are developed with different materials, known as biomaterials, and have shown to be a potential tool to facilitate in vitro cell growth, proliferation, and differentiation. Among the materials studied, carbon materials are potential biomaterials that can be used to develop scaffolds for cell growth. Recently, many researchers have attempted to build a scaffold following the origin of the tissue cell by mimicking the pattern of their extracellular matrix (ECM). In addition, extensive studies were performed on the various parameters that could influence cell behaviour. Previous studies have shown that various factors should be considered in scaffold production, including the porosity, pore size, topography, mechanical properties, wettability, and electroconductivity, which are essential in facilitating cellular response on the scaffold. These interferential factors will help determine the appropriate architecture of the carbon-based scaffold, influencing stem cell (SC) response. Hence, this paper reviews the potential of carbon as a biomaterial for scaffold development. This paper also discusses several crucial factors that can influence the feasibility of the carbon-based scaffold architecture in supporting the efficacy and viability of SCs.
Collapse
Affiliation(s)
- Vieralynda Vitus
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (V.V.); (F.I.)
- Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Fatimah Ibrahim
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (V.V.); (F.I.)
- Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Centre for Printable Electronics, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Wan Safwani Wan Kamarul Zaman
- Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (V.V.); (F.I.)
- Centre for Innovation in Medical Engineering (CIME), Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| |
Collapse
|
35
|
Kong Y, Duan J, Liu F, Han L, Li G, Sun C, Sang Y, Wang S, Yi F, Liu H. Regulation of stem cell fate using nanostructure-mediated physical signals. Chem Soc Rev 2021; 50:12828-12872. [PMID: 34661592 DOI: 10.1039/d1cs00572c] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
One of the major issues in tissue engineering is regulation of stem cell differentiation toward specific lineages. Unlike biological and chemical signals, physical signals with adjustable properties can be applied to stem cells in a timely and localized manner, thus making them a hot topic for research in the fields of biomaterials, tissue engineering, and cell biology. According to the signals sensed by cells, physical signals used for regulating stem cell fate can be classified into six categories: mechanical, light, thermal, electrical, acoustic, and magnetic. In most cases, external macroscopic physical fields cannot be used to modulate stem cell fate, as only the localized physical signals accepted by the surface receptors can regulate stem cell differentiation via nanoscale fibrin polysaccharide fibers. However, surface receptors related to certain kinds of physical signals are still unknown. Recently, significant progress has been made in the development of functional materials for energy conversion. Consequently, localized physical fields can be produced by absorbing energy from an external physical field and subsequently releasing another type of localized energy through functional nanostructures. Based on the above concepts, we propose a methodology that can be utilized for stem cell engineering and for the regulation of stem cell fate via nanostructure-mediated physical signals. In this review, the combined effect of various approaches and mechanisms of physical signals provides a perspective on stem cell fate promotion by nanostructure-mediated physical signals. We expect that this review will aid the development of remote-controlled and wireless platforms to physically guide stem cell differentiation both in vitro and in vivo, using optimized stimulation parameters and mechanistic investigations while driving the progress of research in the fields of materials science, cell biology, and clinical research.
Collapse
Affiliation(s)
- Ying Kong
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Jiazhi Duan
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Feng Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266200, China.
| | - Gang Li
- Neurological Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Chunhui Sun
- Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, China
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Shuhua Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China.
| | - Fan Yi
- The Key Laboratory of Infection and Immunity of Shandong Province, Department of Pharmacology, School of Basic Medical Science, Shandong University, Jinan, 250012, China.
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China. .,Collaborative Innovation Center of Technology and Equipment for Biological Diagnosis and Therapy in Universities of Shandong, Institute for Advanced Interdisciplinary Research (iAIR), University of Jinan, Jinan, 250022, China
| |
Collapse
|
36
|
Serna JA, Rueda-Gensini L, Céspedes-Valenzuela DN, Cifuentes J, Cruz JC, Muñoz-Camargo C. Recent Advances on Stimuli-Responsive Hydrogels Based on Tissue-Derived ECMs and Their Components: Towards Improving Functionality for Tissue Engineering and Controlled Drug Delivery. Polymers (Basel) 2021; 13:3263. [PMID: 34641079 PMCID: PMC8512780 DOI: 10.3390/polym13193263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
Due to their highly hydrophilic nature and compositional versatility, hydrogels have assumed a protagonic role in the development of physiologically relevant tissues for several biomedical applications, such as in vivo tissue replacement or regeneration and in vitro disease modeling. By forming interconnected polymeric networks, hydrogels can be loaded with therapeutic agents, small molecules, or cells to deliver them locally to specific tissues or act as scaffolds for hosting cellular development. Hydrogels derived from decellularized extracellular matrices (dECMs), in particular, have gained significant attention in the fields of tissue engineering and regenerative medicine due to their inherently high biomimetic capabilities and endowment of a wide variety of bioactive cues capable of directing cellular behavior. However, these hydrogels often exhibit poor mechanical stability, and their biological properties alone are not enough to direct the development of tissue constructs with functional phenotypes. This review highlights the different ways in which external stimuli (e.g., light, thermal, mechanical, electric, magnetic, and acoustic) have been employed to improve the performance of dECM-based hydrogels for tissue engineering and regenerative medicine applications. Specifically, we outline how these stimuli have been implemented to improve their mechanical stability, tune their microarchitectural characteristics, facilitate tissue morphogenesis and enable precise control of drug release profiles. The strategic coupling of the bioactive features of dECM-based hydrogels with these stimulation schemes grants considerable advances in the development of functional hydrogels for a wide variety of applications within these fields.
Collapse
Affiliation(s)
| | | | | | | | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (J.A.S.); (L.R.-G.); (D.N.C.-V.); (J.C.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (J.A.S.); (L.R.-G.); (D.N.C.-V.); (J.C.)
| |
Collapse
|
37
|
Liu Z, Wan X, Wang ZL, Li L. Electroactive Biomaterials and Systems for Cell Fate Determination and Tissue Regeneration: Design and Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007429. [PMID: 34117803 DOI: 10.1002/adma.202007429] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/19/2020] [Indexed: 06/12/2023]
Abstract
During natural tissue regeneration, tissue microenvironment and stem cell niche including cell-cell interaction, soluble factors, and extracellular matrix (ECM) provide a train of biochemical and biophysical cues for modulation of cell behaviors and tissue functions. Design of functional biomaterials to mimic the tissue/cell microenvironment have great potentials for tissue regeneration applications. Recently, electroactive biomaterials have drawn increasing attentions not only as scaffolds for cell adhesion and structural support, but also as modulators to regulate cell/tissue behaviors and function, especially for electrically excitable cells and tissues. More importantly, electrostimulation can further modulate a myriad of biological processes, from cell cycle, migration, proliferation and differentiation to neural conduction, muscle contraction, embryogenesis, and tissue regeneration. In this review, endogenous bioelectricity and piezoelectricity are introduced. Then, design rationale of electroactive biomaterials is discussed for imitating dynamic cell microenvironment, as well as their mediated electrostimulation and the applying pathways. Recent advances in electroactive biomaterials are systematically overviewed for modulation of stem cell fate and tissue regeneration, mainly including nerve regeneration, bone tissue engineering, and cardiac tissue engineering. Finally, the significance for simulating the native tissue microenvironment is emphasized and the open challenges and future perspectives of electroactive biomaterials are concluded.
Collapse
Affiliation(s)
- Zhirong Liu
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Xingyi Wan
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Zhong Lin Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, 30332-0245, USA
| | - Linlin Li
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing, 100083, P. R. China
- School of Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
38
|
Litowczenko J, Woźniak-Budych MJ, Staszak K, Wieszczycka K, Jurga S, Tylkowski B. Milestones and current achievements in development of multifunctional bioscaffolds for medical application. Bioact Mater 2021; 6:2412-2438. [PMID: 33553825 PMCID: PMC7847813 DOI: 10.1016/j.bioactmat.2021.01.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/23/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue engineering (TE) is a rapidly growing interdisciplinary field, which aims to restore or improve lost tissue function. Despite that TE was introduced more than 20 years ago, innovative and more sophisticated trends and technologies point to new challenges and development. Current challenges involve the demand for multifunctional bioscaffolds which can stimulate tissue regrowth by biochemical curves, biomimetic patterns, active agents and proper cell types. For those purposes especially promising are carefully chosen primary cells or stem cells due to its high proliferative and differentiation potential. This review summarized a variety of recently reported advanced bioscaffolds which present new functions by combining polymers, nanomaterials, bioactive agents and cells depending on its desired application. In particular necessity of study biomaterial-cell interactions with in vitro cell culture models, and studies using animals with in vivo systems were discuss to permit the analysis of full material biocompatibility. Although these bioscaffolds have shown a significant therapeutic effect in nervous, cardiovascular and muscle, tissue engineering, there are still many remaining unsolved challenges for scaffolds improvement.
Collapse
Affiliation(s)
- Jagoda Litowczenko
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Marta J. Woźniak-Budych
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Katarzyna Staszak
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, Poznan, Poland
| | - Stefan Jurga
- NanoBioMedical Centre, Adam Mickiewicz University in Poznan, Wszechnicy Piastowskiej 3, Poznan, Poland
| | - Bartosz Tylkowski
- Eurecat, Centre Tecnològic de Catalunya, Chemical Technologies Unit, Marcel·lí Domingo s/n, Tarragona, 43007, Spain
| |
Collapse
|
39
|
Modeling Cardiomyopathies in a Dish: State-of-the-Art and Novel Perspectives on hiPSC-Derived Cardiomyocytes Maturation. BIOLOGY 2021; 10:biology10080730. [PMID: 34439963 PMCID: PMC8389603 DOI: 10.3390/biology10080730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/23/2022]
Abstract
The stem cell technology and the induced pluripotent stem cells (iPSCs) production represent an excellent alternative tool to study cardiomyopathies, which overcome the limitations associated with primary cardiomyocytes (CMs) access and manipulation. CMs from human iPSCs (hiPSC-CMs) are genetically identical to patient primary cells of origin, with the main electrophysiological and mechanical features of CMs. The key issue to be solved is to achieve a degree of structural and functional maturity typical of adult CMs. In this perspective, we will focus on the main differences between fetal-like hiPSC-CMs and adult CMs. A viewpoint is given on the different approaches used to improve hiPSC-CMs maturity, spanning from long-term culture to complex engineered heart tissue. Further, we outline limitations and future developments needed in cardiomyopathy disease modeling.
Collapse
|
40
|
Bilge S, Ergene E, Talak E, Gokyer S, Donar YO, Sınağ A, Yilgor Huri P. Recycled algae-based carbon materials as electroconductive 3D printed skeletal muscle tissue engineering scaffolds. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2021; 32:73. [PMID: 34152502 PMCID: PMC8217022 DOI: 10.1007/s10856-021-06534-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/28/2021] [Indexed: 05/03/2023]
Abstract
Skeletal muscle is an electrically and mechanically active tissue that contains highly oriented, densely packed myofibrils. The tissue has self-regeneration capacity upon injury, which is limited in the cases of volumetric muscle loss. Several regenerative therapies have been developed in order to enhance this capacity, as well as to structurally and mechanically support the defect site during regeneration. Among them, biomimetic approaches that recapitulate the native microenvironment of the tissue in terms of parallel-aligned structure and biophysical signals were shown to be effective. In this study, we have developed 3D printed aligned and electrically active scaffolds in which the electrical conductivity was provided by carbonaceous material (CM) derived from algae-based biomass. The synthesis of this conductive and functional CM consisted of eco-friendly synthesis procedure such as pre-carbonization and multi-walled carbon nanotube (MWCNT) catalysis. CM obtained from biomass via hydrothermal carbonization (CM-03) and its ash form (CM-03K) were doped within poly(ɛ-caprolactone) (PCL) matrix and 3D printed to form scaffolds with aligned fibers for structural biomimicry. Scaffolds were seeded with C2C12 mouse myoblasts and subjected to electrical stimulation during the in vitro culture. Enhanced myotube formation was observed in electroactive groups compared to their non-conductive counterparts and it was observed that myotube formation and myotube maturity were significantly increased for CM-03 group after electrical stimulation. The results have therefore showed that the CM obtained from macroalgae biomass is a promising novel source for the production of the electrically conductive scaffolds for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
- Selva Bilge
- Department of Chemistry, Ankara University Faculty of Science, Ankara, Turkey
| | - Emre Ergene
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
- Ankara University Biotechnology Institute, Ankara, Turkey
| | - Ebru Talak
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
| | - Seyda Gokyer
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey
| | - Yusuf Osman Donar
- Department of Chemistry, Ankara University Faculty of Science, Ankara, Turkey
| | - Ali Sınağ
- Department of Chemistry, Ankara University Faculty of Science, Ankara, Turkey.
| | - Pinar Yilgor Huri
- Department of Biomedical Engineering, Ankara University Faculty of Engineering, Ankara, Turkey.
| |
Collapse
|
41
|
Maturation strategies and limitations of induced pluripotent stem cell-derived cardiomyocytes. Biosci Rep 2021; 41:226678. [PMID: 33057659 PMCID: PMC8209171 DOI: 10.1042/bsr20200833] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/06/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) have the ability to differentiate into cardiomyocytes (CMs). They are not only widely used in cardiac pharmacology screening, human heart disease modeling, and cell transplantation-based treatments, but also the most promising source of CMs for experimental and clinical applications. However, their use is largely restricted by the immature phenotype of structure and function, which is similar to embryonic or fetal CMs and has certain differences from adult CMs. In order to overcome this critical issue, many studies have explored and revealed new strategies to induce the maturity of iPSC-CMs. Therefore, this article aims to review recent induction methods of mature iPSC-CMs, related mechanisms, and limitations.
Collapse
|
42
|
Gharanei M, Shafaattalab S, Sangha S, Gunawan M, Laksman Z, Hove-Madsen L, Tibbits GF. Atrial-specific hiPSC-derived cardiomyocytes in drug discovery and disease modeling. Methods 2021; 203:364-377. [PMID: 34144175 DOI: 10.1016/j.ymeth.2021.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/08/2021] [Accepted: 06/12/2021] [Indexed: 12/19/2022] Open
Abstract
The discovery and application of human-induced pluripotent stem cells (hiPSCs) have been instrumental in the investigation of the pathophysiology of cardiovascular diseases. Patient-specific hiPSCs can now be generated, genome-edited, and subsequently differentiated into various cell types and used for regenerative medicine, disease modeling, drug testing, toxicity screening, and 3D tissue generation. Modulation of the retinoic acid signaling pathway has been shown to direct cardiomyocyte differentiation towards an atrial lineage. A variety of studies have successfully differentiated patient-specific atrial cardiac myocytes (hiPSC-aCM) and atrial engineered heart tissue (aEHT) that express atrial specific genes (e.g., sarcolipin and ANP) and exhibit atrial electrophysiological and contractility profiles. Identification of protocols to differentiate atrial cells from patients with atrial fibrillation and other inherited diseases or creating disease models using genetic mutation studies has shed light on the mechanisms of atrial-specific diseases and identified the efficacy of atrial-selective pharmacological compounds. hiPSC-aCMs and aEHTs can be used in drug discovery and drug screening studies to investigate the efficacy of atrial selective drugs on atrial fibrillation models. Furthermore, hiPSC-aCMs can be effective tools in studying the mechanism, pathophysiology and treatment options of atrial fibrillation and its genetic underpinnings. The main limitation of using hiPSC-CMs is their immature phenotype compared to adult CMs. A wide range of approaches and protocols are used by various laboratories to optimize and enhance CM maturation, including electrical stimulation, culture time, biophysical cues and changes in metabolic factors.
Collapse
Affiliation(s)
- Mayel Gharanei
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Sanam Shafaattalab
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Sarabjit Sangha
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Marvin Gunawan
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Zachary Laksman
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Leif Hove-Madsen
- Cardiac Rhythm and Contraction Group, IIBB-CSIC, CIBERCV, IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona 08025, Spain
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Departments of Biomedical Physiology and Kinesiology and Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
43
|
Ly OT, Brown GE, Han YD, Darbar D, Khetani SR. Bioengineering approaches to mature induced pluripotent stem cell-derived atrial cardiomyocytes to model atrial fibrillation. Exp Biol Med (Maywood) 2021; 246:1816-1828. [PMID: 33899540 DOI: 10.1177/15353702211009146] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) serve as a robust platform to model several human arrhythmia syndromes including atrial fibrillation (AF). However, the structural, molecular, functional, and electrophysiological parameters of patient-specific iPSC-derived atrial cardiomyocytes (iPSC-aCMs) do not fully recapitulate the mature phenotype of their human adult counterparts. The use of physiologically inspired microenvironmental cues, such as postnatal factors, metabolic conditioning, extracellular matrix (ECM) modulation, electrical and mechanical stimulation, co-culture with non-parenchymal cells, and 3D culture techniques can help mimic natural atrial development and induce a more mature adult phenotype in iPSC-aCMs. Such advances will not only elucidate the underlying pathophysiological mechanisms of AF, but also identify and assess novel mechanism-based therapies towards supporting a more 'personalized' (i.e. patient-specific) approach to pharmacologic therapy of AF.
Collapse
Affiliation(s)
- Olivia T Ly
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Yong Duk Han
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.,Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA.,Department of Medicine, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
44
|
James EC, Tomaskovic-Crook E, Crook JM. Bioengineering Clinically Relevant Cardiomyocytes and Cardiac Tissues from Pluripotent Stem Cells. Int J Mol Sci 2021; 22:ijms22063005. [PMID: 33809429 PMCID: PMC8001925 DOI: 10.3390/ijms22063005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/23/2022] Open
Abstract
The regenerative capacity of cardiomyocytes is insufficient to functionally recover damaged tissue, and as such, ischaemic heart disease forms the largest proportion of cardiovascular associated deaths. Human-induced pluripotent stem cells (hiPSCs) have enormous potential for developing patient specific cardiomyocytes for modelling heart disease, patient-based cardiac toxicity testing and potentially replacement therapy. However, traditional protocols for hiPSC-derived cardiomyocytes yield mixed populations of atrial, ventricular and nodal-like cells with immature cardiac properties. New insights gleaned from embryonic heart development have progressed the precise production of subtype-specific hiPSC-derived cardiomyocytes; however, their physiological immaturity severely limits their utility as model systems and their use for drug screening and cell therapy. The long-entrenched challenges in this field are being addressed by innovative bioengingeering technologies that incorporate biophysical, biochemical and more recently biomimetic electrical cues, with the latter having the potential to be used to both direct hiPSC differentiation and augment maturation and the function of derived cardiomyocytes and cardiac tissues by mimicking endogenous electric fields.
Collapse
Affiliation(s)
- Emma Claire James
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
| | - Eva Tomaskovic-Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| | - Jeremy Micah Crook
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, Wollongong 2500, Australia;
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2500, Australia
- Department of Surgery, St Vincent’s Hospital, The University of Melbourne, Fitzroy 3065, Australia
- Correspondence: (E.T.-C.); (J.M.C.)
| |
Collapse
|
45
|
Crestani T, Steichen C, Neri E, Rodrigues M, Fonseca-Alaniz MH, Ormrod B, Holt MR, Pandey P, Harding S, Ehler E, Krieger JE. Electrical stimulation applied during differentiation drives the hiPSC-CMs towards a mature cardiac conduction-like cells. Biochem Biophys Res Commun 2020; 533:376-382. [PMID: 32962862 DOI: 10.1016/j.bbrc.2020.09.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) resemble fetal cardiomyocytes and electrical stimulation (ES) has been explored to mature the differentiated cells. Here, we hypothesize that ES applied at the beginning of the differentiation process, triggers both differentiation of the hiPSC-CMs into a specialized conduction system (CS) phenotype and cell maturation. We applied ES for 15 days starting on day 0 of the differentiation process and found an increased expression of transcription factors and proteins associated with the development and function of CS including Irx3, Nkx2.5 and contactin 2, Hcn4 and Scn5a, respectively. We also found activation of intercalated disc proteins (Nrap and β-catenin). We detected ES-induced CM maturation as indicated by increased Tnni1 and Tnni3 expression. Confocal micrographs showed a shift towards expression of the gap junction protein connexin 40 in ES hiPSC-CM compared to the more dominant expression of connexin 43 in controls. Finally, analysis of functional parameters revealed that ES hiPSC-CMs exhibited faster action potential (AP) depolarization, longer intracellular Ca2+ transients, and slower AP duration at 90% of repolarization, resembling fast conducting fibers. Altogether, we provided evidence that ES during the differentiation of hiPSC to cardiomyocytes lead to development of cardiac conduction-like cells with more mature cytoarchitecture. Thus, hiPSC-CMs exposed to ES during differentiation can be instrumental to develop CS cells for cardiac disease modelling, screening individual drugs on a precison medicine type platform and support the development of novel therapeutics for arrhythmias.
Collapse
Affiliation(s)
- Thayane Crestani
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Clara Steichen
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Elida Neri
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Mariliza Rodrigues
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
| | | | - Beth Ormrod
- School of Cardiovascular Medicine and Sciences, BHF Research Excellence Centre, King's College London, UK; Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences, King's College London), UK
| | - Mark R Holt
- School of Cardiovascular Medicine and Sciences, BHF Research Excellence Centre, King's College London, UK; Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences, King's College London), UK
| | - Pragati Pandey
- National Heart and Lung Institute, Imperial College London, UK
| | - Sian Harding
- National Heart and Lung Institute, Imperial College London, UK
| | - Elisabeth Ehler
- School of Cardiovascular Medicine and Sciences, BHF Research Excellence Centre, King's College London, UK; Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences, King's College London), UK
| | - Jose E Krieger
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil.
| |
Collapse
|
46
|
Preconditioned and Genetically Modified Stem Cells for Myocardial Infarction Treatment. Int J Mol Sci 2020; 21:ijms21197301. [PMID: 33023264 PMCID: PMC7582407 DOI: 10.3390/ijms21197301] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/25/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023] Open
Abstract
Ischemic heart disease and myocardial infarction remain leading causes of mortality worldwide. Existing myocardial infarction treatments are incapable of fully repairing and regenerating the infarcted myocardium. Stem cell transplantation therapy has demonstrated promising results in improving heart function following myocardial infarction. However, poor cell survival and low engraftment at the harsh and hostile environment at the site of infarction limit the regeneration potential of stem cells. Preconditioning with various physical and chemical factors, as well as genetic modification and cellular reprogramming, are strategies that could potentially optimize stem cell transplantation therapy for clinical application. In this review, we discuss the most up-to-date findings related to utilizing preconditioned stem cells for myocardial infarction treatment, focusing mainly on preconditioning with hypoxia, growth factors, drugs, and biological agents. Furthermore, genetic manipulations on stem cells, such as the overexpression of specific proteins, regulation of microRNAs, and cellular reprogramming to improve their efficiency in myocardial infarction treatment, are discussed as well.
Collapse
|
47
|
Cacciapuoti M, Johnson B, Kapdia A, Powell S, Gallicano GI. The Role of Neuregulin and Stem Cells as Therapy Post-Myocardial Infarction. Stem Cells Dev 2020; 29:1266-1274. [PMID: 32731805 DOI: 10.1089/scd.2020.0099] [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] [Indexed: 12/18/2022] Open
Abstract
Coronary artery disease, including myocardial infarction (MI), is a leading cause of morbidity and mortality in the United States. Due to the limited self-renewal capacity of cardiac tissue, MIs can lead to progressive heart disease with a lasting impact on health and quality of life. The recent discovery of cardiac stem cells has incited research into their potential therapeutic applications for patients suffering from cardiovascular disease. Studies have demonstrated the ability of stem cells to both generate cardiac tissues in vitro and aid in the recovery of cardiovascular function in vivo in animal models. However, the long-term efficacy of stem cells as regenerative therapy is still unknown. Exploration of alternative therapies is underway, including the use of cardiac growth factor neuregulin-1 (NRG-1). Research has demonstrated that NRG-1 not only has direct effects on cardiomyocytes (CM) but also acts within the tissues supporting the CM. Transplantation of NRG-1 into ischemic cardiac tissue mitigates the progression of heart failure and can reverse cardiac remodeling. Recent publications have sought to study the combined use of these agents, and while the results are promising, they do warrant further research. This review aims to consider these therapies separately as well as in combination.
Collapse
Affiliation(s)
- Maria Cacciapuoti
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Bria Johnson
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Anjani Kapdia
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - Sarah Powell
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| | - G Ian Gallicano
- Georgetown University School of Medicine, Washington, District of Columbia, USA
| |
Collapse
|
48
|
Moon SH, Cho YW, Shim HE, Choi JH, Jung CH, Hwang IT, Kang SW. Electrically stimulable indium tin oxide plate for long-term in vitro cardiomyocyte culture. Biomater Res 2020; 24:10. [PMID: 32514370 PMCID: PMC7251917 DOI: 10.1186/s40824-020-00189-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/10/2020] [Indexed: 12/16/2022] Open
Abstract
Background We investigated whether electrical stimulation via indium tin oxide (ITO) could enhance the in vitro culture of neonatal rat ventricular myocytes (NRVMs), which are important in vitro models for studying the mechanisms underlying many aspects of cardiology. Methods Cardiomyocytes were obtained from 1-day-old neonatal rat heart ventricles. To evaluate function of NRVMs cultured on ITO with electrical stimulation, the cell viability, change of cell morphology, immunochemistry using cardiac-specific antibodies, and gene expression were tested. Results Defined sarcomeric structure, cell enlargement, and increased distribution of NRVMs appeared in the presence of electrical stimulation. These characteristics were absent in NRVMs cultured under standard culture conditions. In addition, the expression levels of cardiomyocyte-specific and ion channel markers were higher in NRVMs seeded on ITO-coated dishes than in the control group at 14 days after seeding. ITO-coated dishes could effectively provide electrical cues to support the in vitro culture of NRVMs. Conclusions These results provide supporting evidence that electrical stimulation via ITO can be effectively used to maintain culture and enhance function of cardiomyocytes in vitro.
Collapse
Affiliation(s)
- Sung-Hwan Moon
- Department of Medical Science, School of Medicine, Konkuk University, Seoul, South Korea
| | - Young-Woo Cho
- Drug Safety and Toxicity Evaluation Team, New Drug Development Center, Osong Medical Innovation Foundation, Cheongju-Si, Chungbuk South Korea
| | - Hye-Eun Shim
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon, South Korea
| | - Jae-Hak Choi
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon, South Korea
| | - Chan-Hee Jung
- Research Division for Industry and Environment, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeonbuk, South Korea
| | - In-Tae Hwang
- Research Division for Industry and Environment, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeonbuk, South Korea
| | - Sun-Woong Kang
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology, Daejeon, South Korea.,Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, South Korea
| |
Collapse
|
49
|
Analyzing Impetus of Regenerative Cellular Therapeutics in Myocardial Infarction. J Clin Med 2020; 9:jcm9051277. [PMID: 32354170 PMCID: PMC7287592 DOI: 10.3390/jcm9051277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
Both vasculature and myocardium in the heart are excessively damaged following myocardial infarction (MI), hence therapeutic strategies for treating MI hearts should concurrently aim for true cardiac repair by introducing new cardiomyocytes to replace lost or injured ones. Of them, mesenchymal stem cells (MSCs) have long been considered a promising candidate for cell-based therapy due to their unspecialized, proliferative differentiation potential to specific cell lineage and, most importantly, their capacity of secreting beneficial paracrine factors which further promote neovascularization, angiogenesis, and cell survival. As a consequence, the differentiated MSCs could multiply and replace the damaged tissues to and turn into tissue- or organ-specific cells with specialized functions. These cells are also known to release potent anti-fibrotic factors including matrix metalloproteinases, which inhibit the proliferation of cardiac fibroblasts, thereby attenuating fibrosis. To achieve the highest possible therapeutic efficacy of stem cells, the other interventions, including hydrogels, electrical stimulations, or platelet-derived biomaterials, have been supplemented, which have resulted in a narrow to broad range of outcomes. Therefore, this article comprehensively analyzed the progress made in stem cells and combinatorial therapies to rescue infarcted myocardium.
Collapse
|
50
|
Solazzo M, O'Brien FJ, Nicolosi V, Monaghan MG. The rationale and emergence of electroconductive biomaterial scaffolds in cardiac tissue engineering. APL Bioeng 2019; 3:041501. [PMID: 31650097 PMCID: PMC6795503 DOI: 10.1063/1.5116579] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
The human heart possesses minimal regenerative potential, which can often lead to chronic heart failure following myocardial infarction. Despite the successes of assistive support devices and pharmacological therapies, only a whole heart transplantation can sufficiently address heart failure. Engineered scaffolds, implantable patches, and injectable hydrogels are among the most promising solutions to restore cardiac function and coax regeneration; however, current biomaterials have yet to achieve ideal tissue regeneration and adequate integration due a mismatch of material physicochemical properties. Conductive fillers such as graphene, carbon nanotubes, metallic nanoparticles, and MXenes and conjugated polymers such as polyaniline, polypyrrole, and poly(3,4-ethylendioxythiophene) can possibly achieve optimal electrical conductivities for cardiac applications with appropriate suitability for tissue engineering approaches. Many studies have focused on the use of these materials in multiple fields, with promising effects on the regeneration of electrically active biological tissues such as orthopedic, neural, and cardiac tissue. In this review, we critically discuss the role of heart electrophysiology and the rationale toward the use of electroconductive biomaterials for cardiac tissue engineering. We present the emerging applications of these smart materials to create supportive platforms and discuss the crucial role that electrical stimulation has been shown to exert in maturation of cardiac progenitor cells.
Collapse
|