1
|
Tsai IT, Sun CK. Stem Cell Therapy against Ischemic Heart Disease. Int J Mol Sci 2024; 25:3778. [PMID: 38612587 PMCID: PMC11011361 DOI: 10.3390/ijms25073778] [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: 02/07/2024] [Revised: 03/12/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Ischemic heart disease, which is one of the top killers worldwide, encompasses a series of heart problems stemming from a compromised coronary blood supply to the myocardium. The severity of the disease ranges from an unstable manifestation of ischemic symptoms, such as unstable angina, to myocardial death, that is, the immediate life-threatening condition of myocardial infarction. Even though patients may survive myocardial infarction, the resulting ischemia-reperfusion injury triggers a cascade of inflammatory reactions and oxidative stress that poses a significant threat to myocardial function following successful revascularization. Moreover, despite evidence suggesting the presence of cardiac stem cells, the fact that cardiomyocytes are terminally differentiated and cannot significantly regenerate after injury accounts for the subsequent progression to ischemic cardiomyopathy and ischemic heart failure, despite the current advancements in cardiac medicine. In the last two decades, researchers have realized the possibility of utilizing stem cell plasticity for therapeutic purposes. Indeed, stem cells of different origin, such as bone-marrow- and adipose-derived mesenchymal stem cells, circulation-derived progenitor cells, and induced pluripotent stem cells, have all been shown to play therapeutic roles in ischemic heart disease. In addition, the discovery of stem-cell-associated paracrine effects has triggered intense investigations into the actions of exosomes. Notwithstanding the seemingly promising outcomes from both experimental and clinical studies regarding the therapeutic use of stem cells against ischemic heart disease, positive results from fraud or false data interpretation need to be taken into consideration. The current review is aimed at overviewing the therapeutic application of stem cells in different categories of ischemic heart disease, including relevant experimental and clinical outcomes, as well as the proposed mechanisms underpinning such observations.
Collapse
Affiliation(s)
- I-Ting Tsai
- Department of Emergency Medicine, E-Da Hospital, I-Shou University, Kaohsiung City 82445, Taiwan;
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Cheuk-Kwan Sun
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
- Department of Emergency Medicine, E-Da Dachang Hospital, I-Shou University, Kaohsiung City 80794, Taiwan
| |
Collapse
|
2
|
Bernava G, Iop L. Advances in the design, generation, and application of tissue-engineered myocardial equivalents. Front Bioeng Biotechnol 2023; 11:1247572. [PMID: 37811368 PMCID: PMC10559975 DOI: 10.3389/fbioe.2023.1247572] [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: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the limited regenerative ability of cardiomyocytes, the disabling irreversible condition of myocardial failure can only be treated with conservative and temporary therapeutic approaches, not able to repair the damage directly, or with organ transplantation. Among the regenerative strategies, intramyocardial cell injection or intravascular cell infusion should attenuate damage to the myocardium and reduce the risk of heart failure. However, these cell delivery-based therapies suffer from significant drawbacks and have a low success rate. Indeed, cardiac tissue engineering efforts are directed to repair, replace, and regenerate native myocardial tissue function. In a regenerative strategy, biomaterials and biomimetic stimuli play a key role in promoting cell adhesion, proliferation, differentiation, and neo-tissue formation. Thus, appropriate biochemical and biophysical cues should be combined with scaffolds emulating extracellular matrix in order to support cell growth and prompt favorable cardiac microenvironment and tissue regeneration. In this review, we provide an overview of recent developments that occurred in the biomimetic design and fabrication of cardiac scaffolds and patches. Furthermore, we sift in vitro and in situ strategies in several preclinical and clinical applications. Finally, we evaluate the possible use of bioengineered cardiac tissue equivalents as in vitro models for disease studies and drug tests.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Padua Medical School, University of Padua, Padua, Italy
| |
Collapse
|
3
|
Kamga Kapchoup MV, Hescheler J, Nguemo F. In vitro effect of hydroxychloroquine on pluripotent stem cells and their cardiomyocytes derivatives. Front Pharmacol 2023; 14:1128382. [PMID: 37502208 PMCID: PMC10369049 DOI: 10.3389/fphar.2023.1128382] [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: 02/17/2023] [Accepted: 06/19/2023] [Indexed: 07/29/2023] Open
Abstract
Introduction: Hydroxychloroquine (HDQ) is an antimalarial drug that has also shown its effectiveness in autoimmune diseases. Despite having side effects such as retinopathy, neuromyopathy and controversial cardiac toxicity, HDQ has been presented and now intensively studied for the treatment and prevention of coronavirus disease 2019 (COVID-19). Recent works revealed both beneficial and toxic effects during HDQ treatment. The cardiotoxic profile of HDQ remains unclear and identifying risk factors is challenging. Methods: Here, we used well-established cell-cultured to study the cytotoxic effect of HDQ, mouse induced pluripotent stem cells (miPSC) and their cardiomyocytes (CMs) derivatives were exposed to different concentrations of HDQ. Cell colony morphology was assessed by microscopy whereas cell viability was measured by flow cytometry and impedance-based methods. The effect of HDQ on beating activity of mouse and human induced pluripotent stem cell-derived CMs (miPSC-CMs and hiPSC-CMs, respectively) and mouse embryonic stem cell-derived CMs (mESC-CMs) were captured by the xCELLigence RTCA and microelectrode array (MEA) systems. Results and discussion: Our results revealed that 20 µM of HDQ promotes proliferation of stem cells used suggesting that if appropriately monitored, HDQ may have a cardioprotective effect and may also represent a possible candidate for tissue repair. In addition, the field potential signals revealed that higher doses of this medication caused bradycardia that could be reversed with a higher concentration of ß-adrenergic agonist, Isoproterenol (Iso). On the contrary, HDQ caused an increase in the beating rate of hiPSC-CMs, which was further helped upon application of Isoproterenol (Iso) suggesting that HDQ and Iso may also work synergistically. These results indicate that HDQ is potentially toxic at high concentrations and can modulate the beating activity of cardiomyocytes. Moreover, HDQ could have a synergistic inotropic effect with isoproterenol on cardiac cells.
Collapse
|
4
|
Roshanravan N, Ghaffari S, Bastani S, Pahlavan S, Asghari S, Doustvandi MA, Jalilzadeh- Razin S, Dastouri M. Human cardiac organoids: A recent revolution in disease modeling and regenerative medicine. J Cardiovasc Thorac Res 2023; 15:68-72. [PMID: 37654821 PMCID: PMC10466470 DOI: 10.34172/jcvtr.2023.31830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/10/2023] [Indexed: 09/02/2023] Open
Abstract
Three-dimensional (3D) myocardial tissues for studying human heart biology, physiology and pharmacology have recently received lots of attention. Organoids as 3D mini-organs are created from multiple cell types (i.e. induced pluripotent stem cells (iPSCs) or embryonic stem cells (ESCs)) with other supporting co-cultured cells such as endothelial cells or fibroblasts. Cardiac organoid culture technologies are bringing about significant advances in organ research and allows for the establishment of tissue regeneration and disease modeling. The present review provides an overview of the recent advances in human cardiac organoid platforms in disease biology and for cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Bastani
- Department of Immunology, Leiden University Medical Science, Leiden, Netherlands
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Samira Asghari
- University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | | | - Sepideh Jalilzadeh- Razin
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadreza Dastouri
- Ankara University Biotechnology Institute and SISBIYOTEK Advanced Research Unit, Gumusdere Yerleskesi, Kecioren, Ankara, Turkey
| |
Collapse
|
5
|
Xu G, Fatima A, Breitbach M, Kuzmenkin A, Fügemann CJ, Ivanyuk D, Kim KP, Cantz T, Pfannkuche K, Schoeler HR, Fleischmann BK, Hescheler J, Šarić T. Electrophysiological Properties of Tetraploid Cardiomyocytes Derived from Murine Pluripotent Stem Cells Generated by Fusion of Adult Somatic Cells with Embryonic Stem Cells. Int J Mol Sci 2023; 24:ijms24076546. [PMID: 37047520 PMCID: PMC10095437 DOI: 10.3390/ijms24076546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Most cardiomyocytes (CMs) in the adult mammalian heart are either binucleated or contain a single polyploid nucleus. Recent studies have shown that polyploidy in CMs plays an important role as an adaptive response to physiological demands and environmental stress and correlates with poor cardiac regenerative ability after injury. However, knowledge about the functional properties of polyploid CMs is limited. In this study, we generated tetraploid pluripotent stem cells (PSCs) by fusion of murine embryonic stem cells (ESCs) and somatic cells isolated from bone marrow or spleen and performed a comparative analysis of the electrophysiological properties of tetraploid fusion-derived PSCs and diploid ESC-derived CMs. Fusion-derived PSCs exhibited characteristics of genuine ESCs and contained a near-tetraploid genome. Ploidy features and marker expression were also retained during the differentiation of fusion-derived cells. Fusion-derived PSCs gave rise to CMs, which were similar to their diploid ESC counterparts in terms of their expression of typical cardiospecific markers, sarcomeric organization, action potential parameters, response to pharmacologic stimulation with various drugs, and expression of functional ion channels. These results suggest that the state of ploidy does not significantly affect the structural and electrophysiological properties of murine PSC-derived CMs. These results extend our knowledge of the functional properties of polyploid CMs and contribute to a better understanding of their biological role in the adult heart.
Collapse
|
6
|
Castells F, Ruipérez-Campillo S, Segarra I, Cervigón R, Casado-Arroyo R, Merino JL, Millet J. Performance assessment of electrode configurations for the estimation of omnipolar electrograms from high density arrays. Comput Biol Med 2023; 154:106604. [PMID: 36709520 DOI: 10.1016/j.compbiomed.2023.106604] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/18/2023] [Accepted: 01/22/2023] [Indexed: 01/26/2023]
Abstract
OBJECTIVE The aim of this study is to propose a method to reduce the sensitivity of the estimated omnipolar electrogram (oEGM) with respect to the angle of the propagation wavefront. METHODS A novel configuration of cliques taking into account all four electrodes of a squared cell is proposed. To test this approach, simulations of HD grids of cardiac activations at different propagation angles, conduction velocities, interelectrode distance and electrogram waveforms are considered. RESULTS The proposed approach successfully provided narrower loops (essentially a straight line) of the electrical field described by the bipole pair with respect to the conventional approach. Estimation of the direction of propagation was improved. Additionally, estimated oEGMs presented larger amplitude, and estimations of the local activation times were more accurate. CONCLUSIONS A novel method to improve the estimation of oEGMs in HD grid of electrodes is proposed. This approach is superior to the existing methods and avoids pitfalls not yet resolved. RELEVANCE Robust tools for quantifying the cardiac substrate are crucial to determine with accuracy target ablation sites during an electrophysiological procedure.
Collapse
Affiliation(s)
| | - Samuel Ruipérez-Campillo
- ITACA Institute, Universitat Politècnica de València, Valencia, Spain; Department of Bioengineering, University of California, Berkeley, CA, USA; School of Medicine, Stanford University, Palo Alto, CA, USA.
| | - Izan Segarra
- ITACA Institute, Universitat Politècnica de València, Valencia, Spain
| | | | | | - José Luis Merino
- Arrhythmia and Robotic Electrophysiology Unit, Hospital Universitario La Paz, Madrid, Spain
| | - José Millet
- ITACA Institute, Universitat Politècnica de València, Valencia, Spain
| |
Collapse
|
7
|
Malihi G, Nikoui V, Elson EL. A review on qualifications and cost effectiveness of induced pluripotent stem cells (IPSCs)-induced cardiomyocytes in drug screening tests. Arch Physiol Biochem 2023; 129:131-142. [PMID: 32783745 DOI: 10.1080/13813455.2020.1802600] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human induced pluripotent stem cells (hIPSCs) have initiated a higher degree of successes in disease modelling, preclinical evaluation of drug therapy and pharmaco-toxicological testing. Since the discovery of iPSCs in 2006, many advanced techniques have been introduced to differentiate iPSCs to cardiomyocytes, which have been progressively improved. The disease models from iPSC-induced cardiomyocytes (iPSC-CM) have been successfully helping to study a variety of cardiac diseases such as long QT syndrome, drug-induced long QT, different cardiomyopathies related to mutations in mitochondria or desmosomal proteins and other rare genetic diseases. IPSC-CMs have also been used to screen the role of chemicals in cardiovascular drug discovery and individualisation of drug dosages. In this review, the quality of current procedures for characterisation and maturation of iPSC-CM lines will be discussed. Also, we will focus on time efficiency and cost of standard differentiation methods after reprogramming.
Collapse
Affiliation(s)
| | - Vahid Nikoui
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Elliot L Elson
- Department of Biochemistry and Molecular Biophysics, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
8
|
Healing the Broken Hearts: A Glimpse on Next Generation Therapeutics. HEARTS 2022. [DOI: 10.3390/hearts3040013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, accounting for 32% of deaths globally and thus representing almost 18 million people according to WHO. Myocardial infarction, the most prevalent adult cardiovascular pathology, affects over half a million people in the USA according to the last records of the AHA. However, not only adult cardiovascular diseases are the most frequent diseases in adulthood, but congenital heart diseases also affect 0.8–1.2% of all births, accounting for mild developmental defects such as atrial septal defects to life-threatening pathologies such as tetralogy of Fallot or permanent common trunk that, if not surgically corrected in early postnatal days, they are incompatible with life. Therefore, both congenital and adult cardiovascular diseases represent an enormous social and economic burden that invariably demands continuous efforts to understand the causes of such cardiovascular defects and develop innovative strategies to correct and/or palliate them. In the next paragraphs, we aim to briefly account for our current understanding of the cellular bases of both congenital and adult cardiovascular diseases, providing a perspective of the plausible lines of action that might eventually result in increasing our understanding of cardiovascular diseases. This analysis will come out with the building blocks for designing novel and innovative therapeutic approaches to healing the broken hearts.
Collapse
|
9
|
Wu J, Lin S, Huang P, Qiu L, Jiang Y, Zhang Y, Meng N, Meng M, Wang L, Deng W, Liu Z, Guo C, Lu J, Wang H, Kong S. Maternal anxiety affects embryo implantation via impairing adrenergic receptor signaling in decidual cells. Commun Biol 2022; 5:840. [PMID: 35982177 PMCID: PMC9388523 DOI: 10.1038/s42003-022-03694-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 07/10/2022] [Indexed: 11/09/2022] Open
Abstract
Recurrent implantation failure (RIF) is defined as the failed pregnancy after good embryo transfer over 3 cycles during in vitro fertilization (IVF).The human endometrium plays a vital role in providing the site for embryo implantation, with several factors implicated in unsatisfactory endometrial receptivity in RIF. Our present results revealed that women with pregnancy loss or infertility have a higher serum epinephrine level, indicating a potential correlation between psychological stress and pregnancy failure. RNA-sequencing of the tissues collected at the endometrial receptive phase in normal and RIF women showed that stress hormones could affect the functional status of endometrial receptivity. Subsequent analysis revealed that the epinephrine signaling acts as an important regulator of endometrial receptivity through the PI3K-AKT and FOXO1 signaling pathways. We also found that patients with RIF show attenuated expression of the alpha-2C-adrenergic receptor (ADRA2C) and that its down regulation induced by high level epinephrine could inhibit the decidualization. Early pregnant mice treated with stress showed high serum epinephrine levels, defective uterine adrenergic receptor expression, and low pregnancy rates. Altogether, our findings indicate that mental stress during early pregnancy can alter the functional status of endometrial receptivity.
Collapse
Affiliation(s)
- Jinxiang Wu
- Department of Reproductive Medicine, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.
| | - Shu Lin
- Centre of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China.,Diabetes and Metabolism Division, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Pinxiu Huang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Lingling Qiu
- Department of Reproductive Medicine, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Yufei Jiang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ying Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Nan Meng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Meiqing Meng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Lemeng Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zhao Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chuanhui Guo
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
10
|
Ma R, Zhao L, Zhao Y, Li Y. Puerarin action on stem cell proliferation, differentiation and apoptosis: Therapeutic implications for geriatric diseases. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153915. [PMID: 35026503 DOI: 10.1016/j.phymed.2021.153915] [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] [Received: 06/16/2021] [Revised: 12/20/2021] [Accepted: 12/25/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Aging is associated with a decline in cognitive and physical functions and various geriatric diseases, such as cardiovascular and neurodegenerative diseases. Puerarin (Pue), one of the main active flavonoids of Radix Puerariae (R. pueraria), is reportedly effective in treating geriatric diseases, including cardiovascular disease and hypertension. PURPOSE This review aims to summarize and discuss the profound physiological impact of Pue on various stem cell populations and provide new insights into the use of Pue for the prevention and treatment of geriatric diseases. METHODS The literature was retrieved from the core collection of electronic databases, such as Web of Science, Google Scholar, PubMed, and Science Direct, using the following keywords and terms: Puerarin, Stem Cell, Proliferation, Differentiation, Apoptosis, and Geriatric diseases. These keywords were used in multiple overlapping combinations. RESULTS Pue is effective in the treatment and management of age-related diseases, such as cardiovascular disease, diabetes, hypertension, and cerebrovascular disease. Pue exerts significant physiological effects on various stem cell populations, including their self-renewal/proliferation, differentiation and apoptosis. Most importantly, it could improve the efficiency and accuracy of stem cell therapy for treating various geriatric diseases. Further studies are essential to improve our understanding of the underlying mechanisms and elucidate their significance for future clinical applications. CONCLUSION The effects of Pue on various stem cell populations and their regulatory mechanisms are discussed in detail to provide new insights into the use of Pue in the prevention and treatment of geriatric diseases.
Collapse
Affiliation(s)
- Ruishuang Ma
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lucy Zhao
- Institute for Pharmacy and Molecular Biotechnology, Functional Genomics, University of Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Yuming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| | - Yue Li
- State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
11
|
QuinoMit Q10-Fluid attenuates hydrogen peroxide-induced irregular beating in mouse pluripotent stem cell-derived cardiomyocytes. Biomed Pharmacother 2021; 142:112089. [PMID: 34449318 DOI: 10.1016/j.biopha.2021.112089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Coenzyme Q10 (CoQ10) is a crucial component of the mitochondrial structure which is involved in producing more than 90% of cellular ATP. This study aimed to investigate the protective effects and underlying mechanisms of QuinoMit Q10-Fluid against hydrogen peroxide (H2O2)-induced arrhythmias on cardiomyocytes (CMs). METHODS Undifferentiated stem cell-derived CMs were cultured in the presence of different concentrations of QuinoMit Q10-Fluid. To investigate if CoQ10 has anti-apoptotic activity, CMs were exposed to H2O2 for up to 100 h with or without CoQ10. The expression levels of cardiac reference genes were determined by RT-PCR. The structural and functional properties of CMs were examined by immunofluorescence and the xCELLigence system. Caspase 3/7 assay was also performed for cell apoptosis study. RESULTS The study showed that QuinoMit Q10-Fluid inhibits the proliferation of pluripotent stem cells at high concentrations and had less effect on cardiomyogenesis. However, the beating rate of clusters containing CMs generated under QuinoMit Q10-Fluid (1:100) was significantly increased. This increase was accompanied by the up-regulated expression level of some important cardiac markers during differentiation. Treatment of CMs with H2O2 notably induced irregular beating and decreased the amplitude of the beating signal of CMs, concomitantly with increased caspase-3/7 activity. However, CMs pretreated with QuinoMit exhibited a protective effect against H2O2-induced arrhythmia. CONCLUSION Our results reveal that QuinoMit Q10-Fluid attenuates H2O2-induced irregular beating in mouse pluripotent stem cell-derived CMs, at least partly by reducing the generation of ROS, suggesting a protective effect against CM dysfunctions.
Collapse
|
12
|
van der Pol A, Bouten CVC. A Brief History in Cardiac Regeneration, and How the Extra Cellular Matrix May Turn the Tide. Front Cardiovasc Med 2021; 8:682342. [PMID: 34095264 PMCID: PMC8172606 DOI: 10.3389/fcvm.2021.682342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
Tissue homeostasis is perturbed by stressful events, which can lead to organ dysfunction and failure. This is particularly true for the heart, where injury resulting from myocardial infarction or ischemic heart disease can result in a cascading event ultimately ending with the loss of functional myocardial tissue and heart failure. To help reverse this loss of healthy contractile tissue, researchers have spent decades in the hopes of characterizing a cell source capable of regenerating the injured heart. Unfortunately, these strategies have proven to be ineffective. With the goal of truly understanding cardiac regeneration, researchers have focused on the innate regenerative abilities of zebrafish and neonatal mammals. This has led to the realization that although cells play an important role in the repair of the diseased myocardium, inducing cardiac regeneration may instead lie in the composition of the extra cellular milieu, specifically the extra cellular matrix. In this review we will briefly summarize the current knowledge regarding cell sources used for cardiac regenerative approaches, since these have been extensively reviewed elsewhere. More importantly, by revisiting innate cardiac regeneration observed in zebrafish and neonatal mammals, we will stress the importance the extra cellular matrix has on reactivating this potential in the adult myocardium. Finally, we will address how we can harness the ability of the extra cellular matrix to guide cardiac repair thereby setting the stage of next generation regenerative strategies.
Collapse
Affiliation(s)
- Atze van der Pol
- Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, Netherlands
| | | |
Collapse
|
13
|
Peinkofer G, Maass M, Pfannkuche K, Sachinidis A, Baldus S, Hescheler J, Saric T, Halbach M. Persistence of intramyocardially transplanted murine induced pluripotent stem cell-derived cardiomyocytes from different developmental stages. Stem Cell Res Ther 2021; 12:46. [PMID: 33419458 PMCID: PMC7792075 DOI: 10.1186/s13287-020-02089-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/09/2020] [Indexed: 01/16/2023] Open
Abstract
Background Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) are regarded as promising cell type for cardiac cell replacement therapy, but it is not known whether the developmental stage influences their persistence and functional integration in the host tissue, which are crucial for a long-term therapeutic benefit. To investigate this, we first tested the cell adhesion capability of murine iPSC-CM in vitro at three different time points during the differentiation process and then examined cell persistence and quality of electrical integration in the infarcted myocardium in vivo. Methods To test cell adhesion capabilities in vitro, iPSC-CM were seeded on fibronectin-coated cell culture dishes and decellularized ventricular extracellular matrix (ECM) scaffolds. After fixed periods of time, stably attached cells were quantified. For in vivo experiments, murine iPSC-CM expressing enhanced green fluorescent protein was injected into infarcted hearts of adult mice. After 6–7 days, viable ventricular tissue slices were prepared to enable action potential (AP) recordings in transplanted iPSC-CM and surrounding host cardiomyocytes. Afterwards, slices were lysed, and genomic DNA was prepared, which was then used for quantitative real-time PCR to evaluate grafted iPSC-CM count. Results The in vitro results indicated differences in cell adhesion capabilities between day 14, day 16, and day 18 iPSC-CM with day 14 iPSC-CM showing the largest number of attached cells on ECM scaffolds. After intramyocardial injection, day 14 iPSC-CM showed a significant higher cell count compared to day 16 iPSC-CM. AP measurements revealed no significant difference in the quality of electrical integration and only minor differences in AP properties between d14 and d16 iPSC-CM. Conclusion The results of the present study demonstrate that the developmental stage at the time of transplantation is crucial for the persistence of transplanted iPSC-CM. iPSC-CM at day 14 of differentiation showed the highest persistence after transplantation in vivo, which may be explained by a higher capability to adhere to the extracellular matrix. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-020-02089-5.
Collapse
Affiliation(s)
- Gabriel Peinkofer
- Department of Internal Medicine III, University Hospital of Cologne, Cologne, Germany. .,Center for Physiology and Pathophysiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch Str. 37, Cologne, 50931, Germany. .,Marga-and-Walter-Boll Laboratory for Cardiac Tissue Engineering, University of Cologne, Cologne, Germany.
| | - Martina Maass
- Department of Internal Medicine III, University Hospital of Cologne, Cologne, Germany.,Department of Ophthalmology and Ocular GvHD Competence Center (P.S.), Medical Faculty, University of Cologne, Cologne, Germany
| | - Kurt Pfannkuche
- Center for Physiology and Pathophysiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch Str. 37, Cologne, 50931, Germany.,Marga-and-Walter-Boll Laboratory for Cardiac Tissue Engineering, University of Cologne, Cologne, Germany.,Department of Pediatric Cardiology, University Hospital of Cologne, Cologne, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Agapios Sachinidis
- Center for Physiology and Pathophysiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch Str. 37, Cologne, 50931, Germany.,Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Stephan Baldus
- Department of Internal Medicine III, University Hospital of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch Str. 37, Cologne, 50931, Germany
| | - Tomo Saric
- Center for Physiology and Pathophysiology, Institute of Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch Str. 37, Cologne, 50931, Germany
| | - Marcel Halbach
- Department of Internal Medicine III, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
14
|
Liew LC, Ho BX, Soh BS. Mending a broken heart: current strategies and limitations of cell-based therapy. Stem Cell Res Ther 2020; 11:138. [PMID: 32216837 PMCID: PMC7098097 DOI: 10.1186/s13287-020-01648-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
The versatility of pluripotent stem cells, attributable to their unlimited self-renewal capacity and plasticity, has sparked a considerable interest for potential application in regenerative medicine. Over the past decade, the concept of replenishing the lost cardiomyocytes, the crux of the matter in ischemic heart disease, with pluripotent stem cell-derived cardiomyocytes (PSC-CM) has been validated with promising pre-clinical results. Nevertheless, clinical translation was hemmed in by limitations such as immature cardiac properties, long-term engraftment, graft-associated arrhythmias, immunogenicity, and risk of tumorigenicity. The continuous progress of stem cell-based cardiac therapy, incorporated with tissue engineering strategies and delivery of cardio-protective exosomes, provides an optimistic outlook on the development of curative treatment for heart failure. This review provides an overview and current status of stem cell-based therapy for heart regeneration, with particular focus on the use of PSC-CM. In addition, we also highlight the associated challenges in clinical application and discuss the potential strategies in developing successful cardiac-regenerative therapy.
Collapse
Affiliation(s)
- Lee Chuen Liew
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore
| | - Beatrice Xuan Ho
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Boon-Seng Soh
- Disease Modeling and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, 61 Biopolis Drive Proteos, Singapore, 138673, Singapore. .,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore. .,Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
15
|
Tompkins BA, Balkan W, Winkler J, Gyöngyösi M, Goliasch G, Fernández-Avilés F, Hare JM. Preclinical Studies of Stem Cell Therapy for Heart Disease. Circ Res 2019; 122:1006-1020. [PMID: 29599277 DOI: 10.1161/circresaha.117.312486] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As part of the TACTICS (Transnational Alliance for Regenerative Therapies in Cardiovascular Syndromes) series to enhance regenerative medicine, here, we discuss the role of preclinical studies designed to advance stem cell therapies for cardiovascular disease. The quality of this research has improved over the past 10 to 15 years and overall indicates that cell therapy promotes cardiac repair. However, many issues remain, including inability to provide complete cardiac recovery. Recent studies question the need for intact cells suggesting that harnessing what the cells release is the solution. Our contribution describes important breakthroughs and current directions in a cell-based approach to alleviating cardiovascular disease.
Collapse
Affiliation(s)
- Bryon A Tompkins
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Wayne Balkan
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Johannes Winkler
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Mariann Gyöngyösi
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Georg Goliasch
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Francisco Fernández-Avilés
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.)
| | - Joshua M Hare
- From the Interdisciplinary Stem Cell Institute (B.A.T., W.B., J.M.H.), Department of Surgery (B.A.T.), and Department of Medicine (W.B., J.M.H.), University of Miami Miller School of Medicine, FL; Department of Cardiology, Medical University of Vienna, Austria (J.W., M.G., G.G.); Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Universidad Complutense, Madrid, Spain (F.F.-A.); and CIBERCV, ISCIII, Madrid, Spain (F.F.-A.).
| |
Collapse
|
16
|
Heras-Bautista CO, Mikhael N, Lam J, Shinde V, Katsen-Globa A, Dieluweit S, Molcanyi M, Uvarov V, Jütten P, Sahito RG, Mederos-Henry F, Piechot A, Brockmeier K, Hescheler J, Sachinidis A, Pfannkuche K. Cardiomyocytes facing fibrotic conditions re-express extracellular matrix transcripts. Acta Biomater 2019; 89:180-192. [PMID: 30862552 DOI: 10.1016/j.actbio.2019.03.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 01/14/2023]
Abstract
Pathophysiological conditions, such as myocardial infarction and mechanical overload affect the mammalian heart integrity, leading to a stiffened fibrotic tissue. With respect to the pathophysiology of cardiac fibrosis but also in the limelight of upcoming approaches of cardiac cell therapy it is of interest to decipher the interaction of cardiomyocytes with fibrotic matrix. Therefore, we designed a hydrogel-based model to engineer fibrotic tissue in vitro as an approach to predict the behavior of cardiomyocytes facing increased matrix rigidity. Here, we generated pure induced pluripotent stem cell-derived cardiomyocytes and cultured them on engineered polyacrylamide hydrogels matching the elasticities of healthy as well as fibrotic cardiac tissue. Only in cardiomyocytes cultured on matrices with fibrotic-like elasticity, transcriptional profiling revealed a substantial up-regulation of a whole panel of cardiac fibrosis-associated transcripts, including collagen I and III, decorin, lumican, and periostin. In addition, matrix metalloproteinases and their inhibitors, known to be essential in cardiac remodeling, were found to be elevated as well as insulin-like growth factor 2. Control experiments with primary cardiac fibroblasts were analyzed and did not show comparable behavior. In conclusion, we do not only present a snapshot on the transcriptomic fingerprint alterations in cardiomyocytes under pathological conditions but also provide a new reproducible approach to study the effects of fibrotic environments to various cell types. STATEMENT OF SIGNIFICANCE: The ageing population in many western countries is faced with an increasing burden of ageing-related diseases such as heart failure which is associated with cardiac fibrosis. A deeper understanding of the interaction of organotypic cells with altered extracellular matrix mechanical properties is of pivotal importance to understand the underlying mechanisms. Here, we present a strategy to combine hydrogel matrices with induced pluripotent stem cell derived cardiomyocytes to study the effect of matrix stiffening on these cells. Our findings suggest an active role of matrix stiffening on cardiomyocyte function and heart failure progression.
Collapse
|
17
|
Different Responses to Drug Safety Screening Targets between Human Neonatal and Infantile Heart Tissue and Cardiac Bodies Derived from Human-Induced Pluripotent Stem Cells. Stem Cells Int 2019; 2019:6096294. [PMID: 30956672 PMCID: PMC6431377 DOI: 10.1155/2019/6096294] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/03/2018] [Indexed: 02/03/2023] Open
Abstract
Aims Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) have become a promising tool in cardiovascular safety pharmacology. Immaturity of iPS-CMs remains an ongoing concern. We compared electrophysiological and contractile features of cardiac bodies (hiPS-CBs) derived from human-induced pluripotent stem cells and human neonatal and infantile myocardial slices relevant for drug screening. Methods and Results Myocardial tissue slices were prepared from biopsies obtained from patients undergoing surgery for hypoplastic left heart syndrome (HLHS) and tetralogy of Fallot (TOF). Electrophysiological features and response to Ik,r blockade as well as contractile properties were investigated using microelectrodes and isometric force measurements and were compared to hiPS-CBs. Both native myocardial tissue slices as well as hiPS-CBs showed action potential prolongation after Ik,r blockade, but early afterdepolarisations could be observed in native myocardial tissue slices only. The force-frequency relationship (FFR) varied at lower frequencies and was negative throughout at higher frequencies in hiPS-CBs. In contrast, native myocardial tissue slices exhibited positive, negative, and biphasic FFRs. In contrast to native myocardial tissue slices, hiPS-CBs failed to show an inotropic response to ß-adrenergic stimulation. Although all groups showed ß-adrenergic induced positive lusitropy, the effect was more pronounced in myocardial tissue slices. Conclusion hiPS-CBs were able to reproduce AP prolongation after Ik,r blockade, but to a lesser extent compared to human neonatal and infantile myocardial tissue slices. Early afterdepolarisations could not be induced in hiPS-CBs. Contractile force was differently regulated by β-adrenergic stimulation in hiPS-CBs and the native myocardium. If used for cardiotoxicity screening, caution is warranted as hiPS-CBs might be less sensitive to pharmacologic targets compared to the native myocardium of neonates and infants.
Collapse
|
18
|
Rikhtegar R, Pezeshkian M, Dolati S, Safaie N, Afrasiabi Rad A, Mahdipour M, Nouri M, Jodati AR, Yousefi M. Stem cells as therapy for heart disease: iPSCs, ESCs, CSCs, and skeletal myoblasts. Biomed Pharmacother 2018; 109:304-313. [PMID: 30396088 DOI: 10.1016/j.biopha.2018.10.065] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 10/04/2018] [Accepted: 10/12/2018] [Indexed: 01/14/2023] Open
Abstract
Heart Diseases are serious and global public health concern. In spite of remarkable therapeutic developments, the prediction of patients with Heart Failure (HF) is weak, and present therapeutic attitudes do not report the fundamental problem of the cardiac tissue loss. Innovative therapies are required to reduce mortality and limit or abolish the necessity for cardiac transplantation. Stem cell-based therapies applied to the treatment of heart disease is according to the understanding that natural self-renewing procedures are inherent to the myocardium, nonetheless may not be adequate to recover the infarcted heart muscle. Following the first account of cell therapy in heart diseases, examination has kept up to rapidity; besides, several animals and human clinical trials have been conducted to preserve the capacity of numerous stem cell population in advance cardiac function and decrease infarct size. The purpose of this study was to censoriously evaluate the works performed regarding the usage of four major subgroups of stem cells, including induced Pluripotent Stem Cells (iPSC), Embryonic Stem Cells (ESCs), Cardiac Stem Cells (CDC), and Skeletal Myoblasts, in heart diseases, at the preclinical and clinical studies. Moreover, it is aimed to argue the existing disagreements, unsolved problems, and prospect directions.
Collapse
Affiliation(s)
- Reza Rikhtegar
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Pezeshkian
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanam Dolati
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Safaie
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Afrasiabi Rad
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Reza Jodati
- Department of Cardiac Surgery, Tabriz University of Medical, Tabriz, Iran; Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
19
|
Mukae Y, Itoh M, Noguchi R, Furukawa K, Arai KI, Oyama JI, Toda S, Nakayama K, Node K, Morita S. The addition of human iPS cell-derived neural progenitors changes the contraction of human iPS cell-derived cardiac spheroids. Tissue Cell 2018; 53:61-67. [PMID: 30060828 DOI: 10.1016/j.tice.2018.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 04/14/2018] [Accepted: 05/04/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND We havebeen attempting to use cardiac spheroids to construct three-dimensional contractilestructures for failed hearts. Recent studies have reported that neuralprogenitors (NPs) play significant roles in heart regeneration. However, theeffect of NPs on the cardiac spheroid has not yet been elucidated. OBJECTIVE This studyaims to demonstrate the influence of NPs on the function of cardiac spheroids. METHODS Thespheroids were constructed on a low-attachment-well plate by mixing humaninduced pluripotent stem (hiPS) cell-derived cardiomyocytes and hiPScell-derived NPs (hiPS-NPs). The ratio of hiPS-NPs was set at 0%, 10%, 20%,30%, and 40% of the total cell number of spheroids, which was 2500. The motionwas recorded, and the fractional shortening and the contraction velocity weremeasured. RESULTS Spheroidswere formed within 48 h after mixing the cells, except for the spheroidscontaining 0% hiPS-NPs. Observation at day 7 revealed significant differencesin the fractional shortening (analysis of variance; p = 0.01). The bestfractional shortening was observed with the spheroids containing 30% hiPS-NPs.Neuronal cells were detected morphologically within the spheroids under aconfocal microscope. CONCLUSION Theaddition of hiPS-NPs influenced the contractile function of the cardiacspheroids. Further studies are warranted to elucidate the underlying mechanism.
Collapse
Affiliation(s)
- Yosuke Mukae
- Department of Thoracic and Cardiovascular Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Manabu Itoh
- Department of Thoracic and Cardiovascular Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Ryo Noguchi
- Department of Thoracic and Cardiovascular Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Kojiro Furukawa
- Department of Thoracic and Cardiovascular Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Ken-Ichi Arai
- Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University, Saga, Japan
| | - Jun-Ichi Oyama
- Department of Cardiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Shuji Toda
- Department of Pathology & Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichi Nakayama
- Department of Regenerative Medicine and Biomedical Engineering, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichi Node
- Department of Cardiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Shigeki Morita
- Department of Cardiovascular Surgery, Kyushu Medical Center, A Hospital of National Hospital Organization, Fukuoka, Japan.
| |
Collapse
|
20
|
Wheelwright M, Win Z, Mikkila JL, Amen KY, Alford PW, Metzger JM. Investigation of human iPSC-derived cardiac myocyte functional maturation by single cell traction force microscopy. PLoS One 2018; 13:e0194909. [PMID: 29617427 PMCID: PMC5884520 DOI: 10.1371/journal.pone.0194909] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 03/13/2018] [Indexed: 11/24/2022] Open
Abstract
Recent advances have made it possible to readily derive cardiac myocytes from human induced pluripotent stem cells (hiPSC-CMs). HiPSC-CMs represent a valuable new experimental model for studying human cardiac muscle physiology and disease. Many laboratories have devoted substantial effort to examining the functional properties of isolated hiPSC-CMs, but to date, force production has not been adequately characterized. Here, we utilized traction force microscopy (TFM) with micro-patterning cell printing to investigate the maximum force production of isolated single hiPSC-CMs under varied culture and assay conditions. We examined the role of length of differentiation in culture and the effects of varied extracellular calcium concentration in the culture media on the maturation of hiPSC-CMs. Results show that hiPSC-CMs developing in culture for two weeks produced significantly less force than cells cultured from one to three months, with hiPSC-CMs cultured for three months resembling the cell morphology and function of neonatal rat ventricular myocytes in terms of size, dimensions, and force production. Furthermore, hiPSC-CMs cultured long term in conditions of physiologic calcium concentrations were larger and produced more force than hiPSC-CMs cultured in standard media with sub-physiological calcium. We also examined relationships between cell morphology, substrate stiffness and force production. Results showed a significant relationship between cell area and force. Implementing directed modifications of substrate stiffness, by varying stiffness from embryonic-like to adult myocardium-like, hiPSC-CMs produced maximal forces on substrates with a lower modulus and significantly less force when assayed on increasingly stiff adult myocardium-like substrates. Calculated strain energy measurements paralleled these findings. Collectively, these findings further establish single cell TFM as a valuable approach to illuminate the quantitative physiological maturation of force in hiPSC-CMs.
Collapse
Affiliation(s)
- Matthew Wheelwright
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Zaw Win
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Jennifer L. Mikkila
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Kamilah Y. Amen
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Patrick W. Alford
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
21
|
The impact of growth factors on human induced pluripotent stem cells differentiation into cardiomyocytes. Life Sci 2018; 196:38-47. [DOI: 10.1016/j.lfs.2018.01.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 01/05/2018] [Accepted: 01/10/2018] [Indexed: 01/29/2023]
|
22
|
Steinhoff G, Nesteruk J, Wolfien M, Große J, Ruch U, Vasudevan P, Müller P. Stem cells and heart disease - Brake or accelerator? Adv Drug Deliv Rev 2017; 120:2-24. [PMID: 29054357 DOI: 10.1016/j.addr.2017.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/11/2022]
Abstract
After two decades of intensive research and attempts of clinical translation, stem cell based therapies for cardiac diseases are not getting closer to clinical success. This review tries to unravel the obstacles and focuses on underlying mechanisms as the target for regenerative therapies. At present, the principal outcome in clinical therapy does not reflect experimental evidence. It seems that the scientific obstacle is a lack of integration of knowledge from tissue repair and disease mechanisms. Recent insights from clinical trials delineate mechanisms of stem cell dysfunction and gene defects in repair mechanisms as cause of atherosclerosis and heart disease. These findings require a redirection of current practice of stem cell therapy and a reset using more detailed analysis of stem cell function interfering with disease mechanisms. To accelerate scientific development the authors suggest intensifying unified computational data analysis and shared data knowledge by using open-access data platforms.
Collapse
Affiliation(s)
- Gustav Steinhoff
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Julia Nesteruk
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Markus Wolfien
- University Rostock, Institute of Computer Science, Department of Systems Biology and Bioinformatics, Ulmenstraße 69, 18057 Rostock, Germany.
| | - Jana Große
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Ulrike Ruch
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Praveen Vasudevan
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| | - Paula Müller
- University Medicine Rostock, Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy, University Medical Center Rostock, Schillingallee 35, 18055 Rostock, Germany.
| |
Collapse
|
23
|
Ghafarzadeh M, Namdari M, Eatemadi A. Stem cell therapies for congenital heart disease. Biomed Pharmacother 2016; 84:1163-1171. [PMID: 27780147 DOI: 10.1016/j.biopha.2016.10.055] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/16/2016] [Accepted: 10/17/2016] [Indexed: 01/15/2023] Open
Abstract
Congenital heart disease (CHD) is the most prevalent congenital anomaly in newborn babies. Cardiac malformations have been induced in different animal model experiments, by perturbing some molecules that take part in the developmental pathways associated with myocyte differentiation, specification, or cardiac morphogenesis. The exact epigenetic, environmental, or genetic, basis for these molecules perturbations is yet to be understood. But, scientist have bridged this gap by introducing autologous stem cell into the defective hearts to treat CHD. The choice of stem cells to use has also raised an issue. In this review, we explore different stem cells that have been recently used, as an update into the pool of this knowledge and we suggested the future perspective into the choice of stem cells to control this disease. We propose that isolating mesenchymal stem cells from neonate will give a robust heart regeneration as compared to adults. This source are easily isolated. To unveil stem cell therapy beyond its possibility and safety, further study is required, including largescale randomized, and clinical trials to certify the efficacy of stem cell therapy.
Collapse
Affiliation(s)
- Masoumeh Ghafarzadeh
- Assalian Hospital, Center for Obstetrics and Gynecology, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Mehrdad Namdari
- Department of Cardiology, Lorestan University of Medical Sciences, Postal address: 6997118544, Khoramabad, Iran.
| | - Ali Eatemadi
- Department of Medical Biotechnology, School of advance Science in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical Sciences, Lorestan, Iran
| |
Collapse
|
24
|
Peinkofer G, Burkert K, Urban K, Krausgrill B, Hescheler J, Saric T, Halbach M. From Early Embryonic to Adult Stage: Comparative Study of Action Potentials of Native and Pluripotent Stem Cell-Derived Cardiomyocytes. Stem Cells Dev 2016; 25:1397-406. [DOI: 10.1089/scd.2016.0073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Gabriel Peinkofer
- Department of Internal Medicine III, University of Cologne, Cologne, Germany
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Karsten Burkert
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Katja Urban
- Department of Internal Medicine III, University of Cologne, Cologne, Germany
| | - Benjamin Krausgrill
- Department of Internal Medicine III, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Tomo Saric
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Marcel Halbach
- Department of Internal Medicine III, University of Cologne, Cologne, Germany
| |
Collapse
|
25
|
Fatima A, Xu G, Nguemo F, Kuzmenkin A, Burkert K, Hescheler J, Šarić T. Murine transgenic iPS cell line for monitoring and selection of cardiomyocytes. Stem Cell Res 2016; 17:266-272. [PMID: 27879210 DOI: 10.1016/j.scr.2016.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 06/20/2016] [Accepted: 07/19/2016] [Indexed: 10/21/2022] Open
Abstract
We report here a transgenic murine induced pluripotent stem cell (iPSC) line expressing puromycin N-acetyltransferase (PAC) and enhanced green fluorescent protein (EGFP) under the control of α-myosin heavy chain promoter. This transgenic cell line reproducibly differentiates into EGFP-expressing cardiomyocytes (CMs) which can be generated at high purity with puromycin treatment and exhibit molecular and functional properties of immature heart muscle cells. This genetically modified iPSC line can be used for assessment of the utility of CMs for myocardial repair, pharmacological and toxicological applications and development of improved cardiac differentiation protocols.
Collapse
Affiliation(s)
- Azra Fatima
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Guoxing Xu
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Alexey Kuzmenkin
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Karsten Burkert
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
26
|
|
27
|
Pilarczyk G, Raulf A, Gunkel M, Fleischmann BK, Lemor R, Hausmann M. Tissue-Mimicking Geometrical Constraints Stimulate Tissue-Like Constitution and Activity of Mouse Neonatal and Human-Induced Pluripotent Stem Cell-Derived Cardiac Myocytes. J Funct Biomater 2016; 7:E1. [PMID: 26751484 PMCID: PMC4810060 DOI: 10.3390/jfb7010001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 12/15/2022] Open
Abstract
The present work addresses the question of to what extent a geometrical support acts as a physiological determining template in the setup of artificial cardiac tissue. Surface patterns with alternating concave to convex transitions of cell size dimensions were used to organize and orientate human-induced pluripotent stem cell (hIPSC)-derived cardiac myocytes and mouse neonatal cardiac myocytes. The shape of the cells, as well as the organization of the contractile apparatus recapitulates the anisotropic line pattern geometry being derived from tissue geometry motives. The intracellular organization of the contractile apparatus and the cell coupling via gap junctions of cell assemblies growing in a random or organized pattern were examined. Cell spatial and temporal coordinated excitation and contraction has been compared on plain and patterned substrates. While the α-actinin cytoskeletal organization is comparable to terminally-developed native ventricular tissue, connexin-43 expression does not recapitulate gap junction distribution of heart muscle tissue. However, coordinated contractions could be observed. The results of tissue-like cell ensemble organization open new insights into geometry-dependent cell organization, the cultivation of artificial heart tissue from stem cells and the anisotropy-dependent activity of therapeutic compounds.
Collapse
Affiliation(s)
- Götz Pilarczyk
- Kirchhoff Institute für Physik, Im Neuenheimer Feld INF 270, Heidelberg D-69120, Germany.
| | - Alexandra Raulf
- Institut für Physiologie der Unversität Bonn, Life & Brain Center, Sigmund Freud Strasse 25, Bonn D-53127, Germany.
| | - Manuel Gunkel
- ViroQuant Cell Networks RNAi Screening Facility, BioQuant Center, Im Neuenheimer Feld INF 267, Heidelberg D-69120, Germany.
| | - Bernd K Fleischmann
- Institut für Physiologie der Unversität Bonn, Life & Brain Center, Sigmund Freud Strasse 25, Bonn D-53127, Germany.
| | - Robert Lemor
- Luxembourg Institute for Science and Technology, 5 avenue des Hauts-Fourneaux, Esch-Belval L-4362, Luxembourg.
| | - Michael Hausmann
- Kirchhoff Institute für Physik, Im Neuenheimer Feld INF 270, Heidelberg D-69120, Germany.
| |
Collapse
|
28
|
The Interaction between Adult Cardiac Fibroblasts and Embryonic Stem Cell-Derived Cardiomyocytes Leads to Proarrhythmic Changes in In Vitro Cocultures. Stem Cells Int 2016; 2016:2936126. [PMID: 26880949 PMCID: PMC4736407 DOI: 10.1155/2016/2936126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/16/2015] [Accepted: 12/14/2015] [Indexed: 01/14/2023] Open
Abstract
Transplantation of stem cell-derived cardiomyocytes is one of the most promising therapeutic approaches after myocardial infarction, as loss of cardiomyocytes is virtually irreversible by endogenous repair mechanisms. In myocardial scars, transplanted cardiomyocytes will be in immediate contact with cardiac fibroblasts. While it is well documented how the electrophysiology of neonatal cardiomyocytes is modulated by cardiac fibroblasts of the same developmental stage, it is unknown how adult cardiac fibroblasts (aCFs) affect the function of embryonic stem cell-derived cardiomyocytes (ESC-CMs). To investigate the effects of aCFs on ESC-CM electrophysiology, we performed extra- and intracellular recordings of murine aCF-ESC-CM cocultures. We observed that spontaneous beating behaviour was highly irregular in aCF-ESC-CM cocultures compared to cocultures with mesenchymal stem cells (coefficient of variation of the interspike interval: 40.5 ± 15.2% versus 9.3 ± 2.0%, p = 0.008) and that action potential amplitude and maximal upstroke velocity (V max) were reduced (amplitude: 52.3 ± 1.7 mV versus 65.1 ± 1.5 mV, V max: 7.0 ± 1.0 V/s versus 36.5 ± 5.3 V/s), while action potential duration (APD) was prolonged (APD50: 25.6 ± 1.0 ms versus 16.8 ± 1.9 ms, p < 0.001; APD90: 52.2 ± 1.5 ms versus 43.3 ± 3.3 ms, p < 0.01) compared to controls. Similar changes could be induced by aCF-conditioned medium. We conclude that the presence of aCFs changes automaticity and induces potentially proarrhythmic changes of ESC-CM electrophysiology.
Collapse
|
29
|
Bedada FB, Wheelwright M, Metzger JM. Maturation status of sarcomere structure and function in human iPSC-derived cardiac myocytes. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1829-38. [PMID: 26578113 DOI: 10.1016/j.bbamcr.2015.11.005] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/05/2015] [Accepted: 11/09/2015] [Indexed: 12/15/2022]
Abstract
Human heart failure due to myocardial infarction is a major health concern. The paucity of organs for transplantation limits curative approaches for the diseased and failing adult heart. Human induced pluripotent stem cell-derived cardiac myocytes (hiPSC-CMs) have the potential to provide a long-term, viable, regenerative-medicine alternative. Significant progress has been made with regard to efficient cardiac myocyte generation from hiPSCs. However, directing hiPSC-CMs to acquire the physiological structure, gene expression profile and function akin to mature cardiac tissue remains a major obstacle. Thus, hiPSC-CMs have several hurdles to overcome before they find their way into translational medicine. In this review, we address the progress that has been made, the void in knowledge and the challenges that remain. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.
Collapse
Affiliation(s)
- Fikru B Bedada
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA
| | - Matthew Wheelwright
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School Minneapolis, MN 55455, USA.
| |
Collapse
|
30
|
Nembo EN, Atsamo AD, Nguelefack TB, Kamanyi A, Hescheler J, Nguemo F. In vitro chronotropic effects of Erythrina senegalensis DC (Fabaceae) aqueous extract on mouse heart slice and pluripotent stem cell-derived cardiomyocytes. JOURNAL OF ETHNOPHARMACOLOGY 2015; 165:163-172. [PMID: 25680843 DOI: 10.1016/j.jep.2015.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 01/27/2015] [Accepted: 02/02/2015] [Indexed: 06/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Erythrina senegalensis DC (Fabaceae) bark is commonly used in sub-Saharan traditional medicine for the treatment of many diseases including gastrointestinal disorders and cardiovascular diseases. In this study, we investigated the effect of the aqueous extract of the stem bark of Erythrina senegalensis on the contractile properties of mouse ventricular slices and human induced pluripotent stem (hiPS) cell-derived cardiomyocytes. We also investigated the cytotoxic effect of the extract on mouse embryonic stem (ES) cells differentiating into cardiomyocytes (CMs). MATERIALS AND METHODS We used well-established electrophysiological technologies to assess the effect of Erythrina senegalensis aqueous extract (ESAE) on the beating activity of mouse ventricular slices, mouse ES and hiPS cell-derived CMs. To study the cytotoxic effect of our extract, differentiating mouse ES cells were exposed to different concentrations of ESAE. EB morphology was assessed by microscopy at different stages of differentiation whereas cell viability was measured by flow cytometry, fluorometry and immunocytochemistry. The electrical activity of CMs and heart slices were respectively captured by the patch clamp technique and microelectrode array (MEA) method following ESAE acute exposure. RESULTS Our findings revealed that ESAE exhibits a biphasic chronotropic activity on mouse ventricular slices with an initial low dose (0.001 and 0.01 µg/mL) decrease in beating activity followed by a corresponding significant increase in chronotropic activity at higher doses above 10 µg/mL. The muscarinic receptor blocker, atropine abolished the negative chronotropic activity of ESAE, while propranolol successfully blocked its positive chronotropic activity. ESAE showed a significant dose-dependent positive chronotropic activity on hiPS cell-derived CMs. Also, though not significantly, ESAE decreased cell viability and increased total caspase-3/7 activity of mouse ES cells in a concentration-dependent manner. CONCLUSION Erythrina senegalensis aqueous extract exhibits a biphasic chronotropic effect on mouse heart and a positive chronotropic activity on hiPS cell-derived CMs, suggesting a possible mechanism through muscarinic and β-adrenergic receptor pathways. Also, ESAE is not cytotoxic on mouse ES cells at concentrations up to 100 µg/mL.
Collapse
Affiliation(s)
| | - Albert Donatien Atsamo
- Laboratory of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon
| | - Télesphore Benoît Nguelefack
- Laboratory of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon
| | - Albert Kamanyi
- Laboratory of Animal Physiology and Phytopharmacology, Faculty of Science, University of Dschang, P.O. Box 67, Dschang, Cameroon
| | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany
| | - Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
31
|
Brodarac A, Šarić T, Oberwallner B, Mahmoodzadeh S, Neef K, Albrecht J, Burkert K, Oliverio M, Nguemo F, Choi YH, Neiss WF, Morano I, Hescheler J, Stamm C. Susceptibility of murine induced pluripotent stem cell-derived cardiomyocytes to hypoxia and nutrient deprivation. Stem Cell Res Ther 2015; 6:83. [PMID: 25900017 PMCID: PMC4445302 DOI: 10.1186/s13287-015-0057-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 05/23/2014] [Accepted: 03/19/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) may be suitable for myocardial repair. While their functional and structural properties have been extensively investigated, their response to ischemia-like conditions has not yet been clearly defined. Methods iPS-CMs were differentiated and enriched from murine induced pluripotent stem cells expressing enhanced green fluorescent protein (eGFP) and puromycin resistance genes under the control of an α-myosin heavy chain (α-MHC) promoter. iPS-CMs maturity and function were characterized by microscopy, real-time PCR, calcium transient recordings, electrophysiology, and mitochondrial function assays, and compared to those from neonatal murine cardiomyocytes. iPS-CMs as well as neonatal murine cardiomyocytes were exposed for 3 hours to hypoxia (1% O2) and glucose/serum deprivation, and viability, apoptosis markers, reactive oxygen species, mitochondrial membrane potential and intracellular stress signaling cascades were investigated. Then, the iPS-CMs response to mesenchymal stromal cell-conditioned medium was determined. Results iPS-CMs displayed key morphological and functional properties that were comparable to those of neonatal cardiomyocytes, but several parameters indicated an earlier iPS-CMs maturation stage. During hypoxia and glucose/serum deprivation, iPS-CMs exhibited a significantly higher proportion of poly-caspase-active, 7-aminoactinomycin D-positive and TUNEL-positive cells than neonatal cardiomyocytes. The average mitochondrial membrane potential was reduced in “ischemic” iPS-CMs but remained unchanged in neonatal cardiomyocytes; reactive oxygen species production was only increased in “ischemic” iPS-CMs, and oxidoreductase activity in iPS-CMs dropped more rapidly than in neonatal cardiomyocytes. In iPS-CMs, hypoxia and glucose/serum deprivation led to upregulation of Hsp70 transcripts and decreased STAT3 phosphorylation and total PKCε protein expression. Treatment with mesenchymal stromal cell-conditioned medium preserved oxidoreductase activity and restored pSTAT3 and PKCε levels. Conclusion iPS-CMs appear to be particularly sensitive to hypoxia and nutrient deprivation. Counteracting the ischemic susceptibility of iPS-CMs with mesenchymal stromal cell-conditioned medium may help enhance their survival and efficacy in cell-based approaches for myocardial repair.
Collapse
Affiliation(s)
- Andreja Brodarac
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany.
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Barbara Oberwallner
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany.
| | | | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center, University Hospital Cologne, Cologne, Germany.
| | - Julie Albrecht
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Karsten Burkert
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Matteo Oliverio
- Max-Planck-Institute for Metabolism Research, Cologne, Germany.
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center, University Hospital Cologne, Cologne, Germany.
| | - Wolfram F Neiss
- Department of Anatomy I, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Ingo Morano
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany.
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany. .,Deutsches Herzzentrum Berlin, Berlin, Germany.
| |
Collapse
|
32
|
Nembo EN, Dimo T, Bopda OSM, Hescheler J, Nguemo F. The proliferative and chronotropic effects of Brillantaisia nitens Lindau (Acanthaceae) extracts on pluripotent stem cells and their cardiomyocytes derivatives. JOURNAL OF ETHNOPHARMACOLOGY 2014; 156:73-81. [PMID: 25086409 DOI: 10.1016/j.jep.2014.07.046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/04/2014] [Accepted: 07/19/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Brillantaisia nitens Lindau (Acanthaceae) leaves are commonly used in traditional medicine in Africa for the treatment of many disorders including heart diseases and malaria. In this study, we therefore evaluated the effect of the methylene chloride/methanol leaf extract of Brillantaisia nitens on the proliferation of mouse pluripotent stem cells and their cardiomyocyte derivatives. MATERIALS AND METHODS In this study, we combined two emerging technologies, pluripotent stem cell-derived cardiomyocytes and modern electrophysiology systems (impedance-based real-time) to assess the cytotoxicity of Brillantaisia nitens extract (BNE). Undifferentiated pluripotent cells and cardiomyocytes were exposed to different concentrations of BNE. Cell viability and contraction were monitored by impedance using the xCELLigence system for short- and long-term treatment whereas the excitability of single cardiomyocytes was captured by patch clamp technique after BNE acute exposure. RESULTS Brillantaisia nitens extract inhibited the proliferation and increased cytotoxicity of embryonic stem cells in a concentration-dependent manner. With the increase in concentration of BNE, beating rate and the contractile amplitude of cardiomyocytes changed significantly. Spontaneous rhythmic activity of cardiomyocytes was completely suppressed after 48 and 24h exposures to relatively low (4.16 mg/ml) and high (8.32 mg/ml) concentrations of BNE, respectively. Moreover, acute application of 4.16 mg/ml of BNE led to a significant alteration of action potential (AP) parameters such as beating frequency, amplitude and AP duration at 90% of repolarization. CONCLUSION Brillantaisia nitens extract inhibits the proliferative capacity of pluripotent stem cells and reduces electrical activity of cardiomyocytes, confirming its depressant action on the heart.
Collapse
Affiliation(s)
- Erastus Nembu Nembo
- Center for Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Theophile Dimo
- Department of Animal Biology and Physiology, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon
| | | | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute of Neurophysiology, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany.
| |
Collapse
|
33
|
Lepperhof V, Polchynski O, Kruttwig K, Brüggemann C, Neef K, Drey F, Zheng Y, Ackermann JP, Choi YH, Wunderlich TF, Hoehn M, Hescheler J, Šarić T. Bioluminescent imaging of genetically selected induced pluripotent stem cell-derived cardiomyocytes after transplantation into infarcted heart of syngeneic recipients. PLoS One 2014; 9:e107363. [PMID: 25226590 PMCID: PMC4167328 DOI: 10.1371/journal.pone.0107363] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 08/15/2014] [Indexed: 01/16/2023] Open
Abstract
Cell loss after transplantation is a major limitation for cell replacement approaches in regenerative medicine. To assess the survival kinetics of induced pluripotent stem cell (iPSC)-derived cardiomyocytes (CM) we generated transgenic murine iPSC lines which, in addition to CM-specific expression of puromycin N-acetyl-transferase and enhanced green fluorescent protein (EGFP), also constitutively express firefly luciferase (FLuc) for bioluminescence (BL) in vivo imaging. While undifferentiated iPSC lines generated by random integration of the transgene into the genome retained stable FLuc activity over many passages, the BL signal intensity was strongly decreased in purified iPS-CM compared to undifferentiated iPSC. Targeted integration of FLuc-expression cassette into the ROSA26 genomic locus using zinc finger nuclease (ZFN) technology strongly reduced transgene silencing in iPS-CM, leading to a several-fold higher BL compared to iPS-CM expressing FLuc from random genomic loci. To investigate the survival kinetics of iPS-CM in vivo, purified CM obtained from iPSC lines expressing FLuc from a random or the ROSA26 locus were transplanted into cryoinfarcted hearts of syngeneic mice. Engraftment of viable cells was monitored by BL imaging over 4 weeks. Transplanted iPS-CM were poorly retained in the myocardium independently of the cell line used. However, up to 8% of cells survived for 28 days at the site of injection, which was confirmed by immunohistological detection of EGFP-positive iPS-CM in the host tissue. Transplantation of iPS-CM did not affect the scar formation or capillary density in the periinfarct region of host myocardium. This report is the first to determine the survival kinetics of drug-selected iPS-CM in the infarcted heart using BL imaging and demonstrates that transgene silencing in the course of iPSC differentiation can be greatly reduced by employing genome editing technology. FLuc-expressing iPS-CM generated in this study will enable further studies to reduce their loss, increase long-term survival and functional integration upon transplantation.
Collapse
Affiliation(s)
- Vera Lepperhof
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Olga Polchynski
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Klaus Kruttwig
- In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, Cologne, Germany
| | - Chantal Brüggemann
- In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, Cologne, Germany
| | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Florian Drey
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany
| | - Yunjie Zheng
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Justus P. Ackermann
- Max Planck Institute for Metabolism Research and Institute for Genetics, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Thomas F. Wunderlich
- Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
- Max Planck Institute for Metabolism Research and Institute for Genetics, Cologne, Germany
- Cologne Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Tomo Šarić
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
34
|
Mezzanotte L, Aswendt M, Tennstaedt A, Hoeben R, Hoehn M, Löwik C. Evaluating reporter genes of different luciferases for optimized in vivo bioluminescence imaging of transplanted neural stem cells in the brain. CONTRAST MEDIA & MOLECULAR IMAGING 2014; 8:505-13. [PMID: 24375906 DOI: 10.1002/cmmi.1549] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 05/07/2013] [Accepted: 05/10/2013] [Indexed: 11/05/2022]
Abstract
Bioluminescence imaging (BLI) has become the method of choice for optical tracking of cells in small laboratory animals. However, the use of luciferases from different species, depending on different substrates and emitting at distinct wavelengths, has not been optimized for sensitive neuroimaging. In order to identify the most suitable luciferase, this quantitative study compared the luciferases Luc2, CBG99, PpyRE9 and hRluc. Human embryonic kidney (HEK-293) cells and mouse neural stem cells were transduced by lentiviral vector-mediated transfer to express one of the four luciferases, together with copGFP. A T2A peptide linker promoted stoichiometric expression between both imaging reporters and the comparison of cell populations upon flow cytometry. Cell dilution series were used to determine highest BLI sensitivity in vitro for Luc2. However, Coelenterazine h-dependent hRluc signals clearly exceeded d-luciferin-dependent BLI in vitro. For the quantitative in vivo analysis, cells were transplanted into mouse brain and BLI was performed including the recording of emission kinetics and spectral characteristics. Differences in light kinetics were observed for d-luciferin vs Coelenterazine h. The emission spectra of Luc2 and PpyRE9 remained almost unchanged, while the emission spectrum of CBG99 became biphasic. Most importantly, photon emission decreased in the order of Luc2, CBG99, PpyRE9 to hRluc. The feasibility of combining different luciferases for dual color and dual substrate neuroimaging was tested and discussed. This investigation provides the first complete quantitative comparison of different luciferases expressed by neural stem cells. It results in a clear recommendation of Luc2 as the best luciferase selection for in vivo neuroimaging.
Collapse
Affiliation(s)
- Laura Mezzanotte
- Experimental Molecular Imaging, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
35
|
Heras-Bautista CO, Katsen-Globa A, Schloerer NE, Dieluweit S, Abd El Aziz OM, Peinkofer G, Attia WA, Khalil M, Brockmeier K, Hescheler J, Pfannkuche K. The influence of physiological matrix conditions on permanent culture of induced pluripotent stem cell-derived cardiomyocytes. Biomaterials 2014; 35:7374-85. [PMID: 24889032 DOI: 10.1016/j.biomaterials.2014.05.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 05/13/2014] [Indexed: 10/25/2022]
Abstract
Cardiomyocytes (CMs) from induced pluripotent stem (iPS) cells mark an important achievement in the development of in vitro pharmacological, toxicological and developmental assays and in the establishment of protocols for cardiac cell replacement therapy. Using CMs generated from murine embryonic stem cells and iPS cells we found increased cell-matrix interaction and more matured embryoid body (EB) structures in iPS cell-derived EBs. However, neither suspension-culture in form of purified cardiac clusters nor adherence-culture on traditional cell culture plastic allowed for extended culture of CMs. CMs grown for five weeks on polystyrene exhibit signs of massive mechanical stress as indicated by α-smooth muscle actin expression and loss of sarcomere integrity. Hydrogels from polyacrylamide allow adapting of the matrix stiffness to that of cardiac tissue. We were able to eliminate the bottleneck of low cell adhesion using 2,5-Dioxopyrrolidin-1-yl-6-acrylamidohexanoate as a crosslinker to immobilize matrix proteins on the gels surface. Finally we present an easy method to generate polyacrylamide gels with a physiological Young's modulus of 55 kPa and defined surface ligand, facilitating the culture of murine and human iPS-CMs, removing excess mechanical stresses and reducing the risk of tissue culture artifacts exerted by stiff substrates.
Collapse
Affiliation(s)
- Carlos O Heras-Bautista
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Alisa Katsen-Globa
- Fraunhofer Institute for Biomedical Engineering IBMT, St. Ingbert, Germany
| | | | - Sabine Dieluweit
- Institute of Complex Systems, ICS-7: Biomechanics, Forschungszentrum Jülich GmbH, Jülich, Germany
| | - Osama M Abd El Aziz
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany; Department of Pediatrics, Cairo University, Cairo, Egypt; Department of Paediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| | - Gabriel Peinkofer
- Department of Internal Medicine III, University Clinics of Cologne, Cologne, Germany
| | - Wael A Attia
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany; Department of Pediatrics, Cairo University, Cairo, Egypt; Department of Paediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| | - Markus Khalil
- Department of Paediatric Cardiology, University Clinics of Cologne, Cologne, Germany; Division of Pediatric Cardiology, University Children's Hospital, Giessen, Germany
| | - Konrad Brockmeier
- Department of Paediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Kurt Pfannkuche
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Cologne, Germany; Department of Paediatric Cardiology, University Clinics of Cologne, Cologne, Germany.
| |
Collapse
|
36
|
Nguemo F, Semmler J, Reppel M, Hescheler J. Modulation of L-type calcium current by intracellular magnesium in differentiating cardiomyocytes derived from induced pluripotent stem cells. Stem Cells Dev 2014; 23:1316-27. [PMID: 24527794 DOI: 10.1089/scd.2013.0549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Intracellular Mg(2+), which is implicated in arrhythmogenesis and transient cardiac ischemia, inhibits L-type Ca(2+) calcium channel current (ICaL) of adult cardiomyocytes (CMs). We take the advantage of an in vitro model of CMs based on induced pluripotent stem cells to investigate the effects of intracellular Mg(2+) on the phosphorylation or dephosphorylation processes of L-type Ca(2+) channels (LTCCs) at early and late stages of cardiac cell differentiation. Using the whole-cell patch-clamp technique, we demonstrate that increasing intracellular Mg(2+) concentration [Mg(2+)]i from 0.2 to 5 mM markedly reduced the peak of ICaL density, showing less effect on both the activation and inactivation properties in the late differentiation stage (LDS) of CMs more so than in the early differentiation stage (EDS). Increasing the [Mg(2+)]i from 0.2 to 2 mM in the presence of cAMP-dependent protein kinase A significantly decreased ICaL in LDS (70%) and in EDS (36%) CMs. In addition, the effect of forskolin was greatly attenuated in the presence of 2 mM [Mg(2+)]i in LDS but not in EDS CMs. The effect of forskolin was enhanced in the presence of ATP-γ-S in LDS CMs compared with EDS CMs. The exposure of both EDS and LDS CMs to 2 mM [Mg(2+)]i considerably reduced the effects of isobutylmethylxanthine (IBMX) and okadaic acid on ICaL. Our results provide evidence for differential regulation of LTCCs activities by cytosolic Mg(2+) concentration in developing cardiac cells and confirm that Mg(2+) acts under conditions that favor opening of the LTCCs caused by channel phosphorylation.
Collapse
Affiliation(s)
- Filomain Nguemo
- 1 Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne , Cologne, Germany
| | | | | | | |
Collapse
|
37
|
Takeuchi A, Shimba K, Takayama Y, Kotani K, Lee JK, Noshiro M, Jimbo Y. Microfabricated device for co-culture of sympathetic neuron and iPS-derived cardiomyocytes. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2013; 2013:3817-20. [PMID: 24110563 DOI: 10.1109/embc.2013.6610376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Induced pluripotent stem (iPS) cell-derived cardiomyocytes (iPS-CMs) has been expected as a cell source for therapy of serious heart failure. However, it is unclear whether the function of iPS-CMs is modulated by the host sympathetic nervous system. Here we developed a device for co-culture of sympathetic neurons and iPS-CMs using microfabrication technique. The device consisted of a culture chamber and a microelectrode-array (MEA) substrate. The superior cervical ganglion (SCG) neurons were co-cultured with iPS-CMs in a microfabricated device, which had multiple compartments. Several days after seeding, synapses were formed between SCG neurons and iPS-CMs, as confirmed by immunostaining. Spontaneous electrical activities of the SCG neurons and the iPS-CMs were observed from the electrode of the MEA substrate. The beat rate of iPS-CMs increased after electrical stimulation of the co-cultured SCG neurons. Such changes in the beat rate were prevented in the presence of propranolol, a β-adrenoreceptor antagonist. These results suggest that the microfabricated device will be utilized for studying the functional modulation of iPS-CMs by connected sympathetic neurons.
Collapse
|
38
|
Halbach M, Peinkofer G, Baumgartner S, Maass M, Wiedey M, Neef K, Krausgrill B, Ladage D, Fatima A, Saric T, Hescheler J, Müller-Ehmsen J. Electrophysiological integration and action potential properties of transplanted cardiomyocytes derived from induced pluripotent stem cells. Cardiovasc Res 2013; 100:432-40. [PMID: 24042016 DOI: 10.1093/cvr/cvt213] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Induced pluripotent stem cell-derived cardiomyocytes (iPSCM) are regarded as promising cell type for cardiac cell replacement therapy. We investigated long-term electrophysiological integration and maturation of transplanted iPSCM, which are essential for therapeutic benefit. METHODS AND RESULTS Murine iPSCM expressing enhanced green fluorescent protein and a puromycin resistance under control of the α-myosin heavy chain promoter were purified by antibiotic selection and injected into adult mouse hearts. After 6-12 days, 3-6 weeks, or 6-8 months, viable slices of recipient hearts were prepared. Slices were focally stimulated by a unipolar electrode placed in host tissue, and intracellular action potentials (APs) were recorded with glass microelectrodes in transplanted cells and neighbouring host tissue within the slices. Persistence and electrical integration of transplanted iPSCM into recipient hearts could be demonstrated at all time points. Quality of coupling improved, as indicated by a maximal stimulation frequency without conduction blocks of 5.77 ± 0.54 Hz at 6-12 days, 8.98 ± 0.38 Hz at 3-6 weeks and 10.82 ± 1.07 Hz at 6-8 months after transplantation. AP properties of iPSCM became more mature from 6-12 days to 6-8 months after transplantation, but still differed significantly from those of host APs. CONCLUSION Transplanted iPSCM can persist in the long term and integrate electrically into host tissue, supporting their potential for cell replacement therapy. Quality of electrical integration improves between 6-12 days and 6-8 months after transplantation, and there are signs of an electrophysiological maturation. However, even after 6-8 months, AP properties of transplanted iPSCM differ from those of recipient cardiomyocytes.
Collapse
Affiliation(s)
- Marcel Halbach
- Department of Internal Medicine III, University Hospital of Cologne, Kerpener Str. 62, D-50937 Cologne, Köln, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sanganalmath SK, Bolli R. Cell therapy for heart failure: a comprehensive overview of experimental and clinical studies, current challenges, and future directions. Circ Res 2013; 113:810-34. [PMID: 23989721 PMCID: PMC3892665 DOI: 10.1161/circresaha.113.300219] [Citation(s) in RCA: 446] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 06/07/2013] [Indexed: 12/28/2022]
Abstract
Despite significant therapeutic advances, the prognosis of patients with heart failure (HF) remains poor, and current therapeutic approaches are palliative in the sense that they do not address the underlying problem of the loss of cardiac tissue. Stem cell-based therapies have the potential to fundamentally transform the treatment of HF by achieving what would have been unthinkable only a few years ago-myocardial regeneration. For the first time since cardiac transplantation, a therapy is being developed to eliminate the underlying cause of HF, not just to achieve damage control. Since the initial report of cell therapy (skeletal myoblasts) in HF in 1998, research has proceeded at lightning speed, and numerous preclinical and clinical studies have been performed that support the ability of various stem cell populations to improve cardiac function and reduce infarct size in both ischemic and nonischemic cardiomyopathy. Nevertheless, we are still at the dawn of this therapeutic revolution. Many important issues (eg, mechanism(s) of action of stem cells, long-term engraftment, optimal cell type(s), and dose, route, and frequency of cell administration) remain to be resolved, and no cell therapy has been conclusively shown to be effective. The purpose of this article is to critically review the large body of work performed with respect to the use of stem/progenitor cells in HF, both at the experimental and clinical levels, and to discuss current controversies, unresolved issues, challenges, and future directions. The review focuses specifically on chronic HF; other settings (eg, acute myocardial infarction, refractory angina) are not discussed.
Collapse
Affiliation(s)
- Santosh K Sanganalmath
- Division of Cardiovascular Medicine and Institute of Molecular Cardiology, University of Louisville, KY, USA
| | | |
Collapse
|
40
|
Oyamada M, Takebe K, Endo A, Hara S, Oyamada Y. Connexin expression and gap-junctional intercellular communication in ES cells and iPS cells. Front Pharmacol 2013; 4:85. [PMID: 23840189 PMCID: PMC3699729 DOI: 10.3389/fphar.2013.00085] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2013] [Accepted: 06/13/2013] [Indexed: 01/23/2023] Open
Abstract
Pluripotent stem cells, i.e., embryonic stem (ES) and induced pluripotent stem (iPS) cells, can indefinitely proliferate without commitment and differentiate into all cell lineages. ES cells are derived from the inner cell mass of the preimplantation blastocyst, whereas iPS cells are generated from somatic cells by overexpression of a few transcription factors. Many studies have demonstrated that mouse and human iPS cells are highly similar but not identical to their respective ES cell counterparts. The potential to generate basically any differentiated cell types from these cells offers the possibility to establish new models of mammalian development and to create new sources of cells for regenerative medicine. ES cells and iPS cells also provide useful models to study connexin expression and gap-junctional intercellular communication (GJIC) during cell differentiation and reprogramming. In 1996, we reported connexin expression and GJIC in mouse ES cells. Because a substantial number of papers on these subjects have been published since our report, this Mini Review summarizes currently available data on connexin expression and GJIC in ES cells and iPS cells during undifferentiated state, differentiation, and reprogramming.
Collapse
Affiliation(s)
- Masahito Oyamada
- Department of Food Science and Human Nutrition, Faculty of Human Life Sciences, Fuji Women's University Ishikarishi, Japan
| | | | | | | | | |
Collapse
|
41
|
Nguemo F, Fleischmann BK, Gupta MK, Šarić T, Malan D, Liang H, Pfannkuche K, Bloch W, Schunkert H, Hescheler J, Reppel M. The L-type Ca2+ channels blocker nifedipine represses mesodermal fate determination in murine embryonic stem cells. PLoS One 2013; 8:e53407. [PMID: 23320083 PMCID: PMC3539992 DOI: 10.1371/journal.pone.0053407] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 11/28/2012] [Indexed: 01/20/2023] Open
Abstract
Dihydropyridines (DHP), which nifedipine is a member of, preferentially block Ca(2+) channels of different cell types. Moreover, influx of Ca(2+) through L-type Ca(2+) channels (LTCCs) activates Ca(2+) signaling pathways, which in turn contribute to numerous cellular processes. Although LTCCs are expressed in undifferentiated cells, very little is known about its contributions to the transcriptional regulation of mesodermal and cardiac genes. This study aimed to examine the contribution of LTCCs and the effect of nifedipine on the commitment of pluripotent stem cells toward the cardiac lineage in vitro. The murine embryonic stem (ES, cell line D3) and induced pluripotent stem (iPS, cell clone 09) cells were differentiated into enhanced green fluorescence protein (EGFP) expressing spontaneously beating cardiomyocytes (CMs). Early treatment of differentiating cells with 10 µM nifedipine led to a significant inhibition of the cardiac mesoderm formation and cardiac lineage commitment as revealed by gene regulation analysis. This was accompanied by the inhibition of spontaneously occurring Ca(2+) transient and reduction of LTCCs current density (I(CaL)) of differentiated CMs. In addition, nifedipine treatment instigated a pronounced delay of the spontaneous beating embryoid body (EB) and led to a poor surface localization of L-type Ca(2+) channel α(1C) (Ca(V)1.2) subunits. Contrary late incubation of pluripotent stem cells with nifedipine was without any impact on the differentiation process and did not affect the derived CMs function. Our data indicate that nifedipine blocks the determined path of pluripotent stem cells to cardiomyogenesis by inhibition of mesodermal commitment at early stages of differentiation, thus the proper upkeep Ca(2+) concentration and pathways are essentially required for cardiac gene expression, differentiation and function.
Collapse
Affiliation(s)
- Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Bernd K. Fleischmann
- Institute of Physiology I, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Manoj K. Gupta
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Tomo Šarić
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Daniela Malan
- Institute of Physiology I, Life and Brain Center, University of Bonn, Bonn, Germany
| | - Huamin Liang
- Department of Physiology, Huazhong University of Science and Technology, Tongji Medical College, Wuhan, China
| | - Kurt Pfannkuche
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, German Sport University, Cologne, Germany
| | | | - Jürgen Hescheler
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Michael Reppel
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
- Department of Cardiology, Medical University of Lübeck, Lübeck, Germany
| |
Collapse
|
42
|
Kozlova EN, Berens C. Guiding Differentiation of Stem Cells in Vivo by Tetracycline-Controlled Expression of Key Transcription Factors. Cell Transplant 2012; 21:2537-54. [DOI: 10.3727/096368911x637407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transplantation of stem or progenitor cells is an attractive strategy for cell replacement therapy. However, poor long-term survival and insufficiently reproducible differentiation to functionally appropriate cells in vivo still present major obstacles for translation of this methodology to clinical applications. Numerous experimental studies have revealed that the expression of just a few transcription factors can be sufficient to drive stem cell differentiation toward a specific cell type, to transdifferentiate cells from one fate to another, or to dedifferentiate mature cells to pluripotent stem/progenitor cells (iPSCs). We thus propose here to apply the strategy of expressing the relevant key transcription factors to guide the differentiation of transplanted cells to the desired cell fate in vivo. To achieve this requires tools allowing us to control the expression of these genes in the transplant. Here, we describe drug-inducible systems that allow us to sequentially and timely activate gene expression from the outside, with a particular emphasis on the Tet system, which has been widely and successfully used in stem cells. These regulatory systems offer a tool for strictly limiting gene expression to the respective optimal stage after transplantation. This approach will direct the differentiation of the immature stem/progenitor cells in vivo to the desired cell type.
Collapse
Affiliation(s)
- Elena N Kozlova
- Department of Neuroscience, Uppsala University, Uppsala, Sweden.
| | | |
Collapse
|
43
|
Hsiao LC, Carr C, Chang KC, Lin SZ, Clarke K. Stem cell-based therapy for ischemic heart disease. Cell Transplant 2012; 22:663-75. [PMID: 23044395 DOI: 10.3727/096368912x655109] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite great advances in therapy over the past decades, ischemic heart disease (IHD) remains the leading cause of death worldwide because the decrease in mortality after acute myocardial infarction (AMI) leads to a longer life span in patients with chronic postinfarct heart failure (HF). There are no existing medical treatments that can cure chronic HF and the only currently available therapeutic option for end-stage HF is heart transplantation. However, transplantation is limited by the shortage of donor organs and patients require lifelong immunosuppression. In the past 10 years, stem cell-based cardiac therapy has been proposed as a promising approach for the treatment of IHD. There is a variety of potential stem cell types for cardiac repair and regeneration, including bone marrow cells (BMCs), resident cardiac stem cells (CSCs) and induced pluripotent stem cells (iPSCs). Stem cell-based therapy may comprise cell transplantation or cardiac tissue engineering (CTE), which might be an attractive alternative to solve the problems of low retention and poor survival of transplanted cells. This review focuses on the characteristics of stem cells from various sources and discusses the strategies of stem cell-based therapy for the treatment of IHD.
Collapse
Affiliation(s)
- Lien-Cheng Hsiao
- Cardiac Metabolism Research Group, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | | | | | | | | |
Collapse
|
44
|
Sheng X, Reppel M, Nguemo F, Mohammad FI, Kuzmenkin A, Hescheler J, Pfannkuche K. Human pluripotent stem cell-derived cardiomyocytes: response to TTX and lidocain reveals strong cell to cell variability. PLoS One 2012; 7:e45963. [PMID: 23029342 PMCID: PMC3459939 DOI: 10.1371/journal.pone.0045963] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 08/28/2012] [Indexed: 02/07/2023] Open
Abstract
Stem cell derived cardiomyocytes generated either from human embryonic stem cells (hESC-CMs) or human induced pluripotent stem cells (hiPSC-CMs) hold great promise for the investigation of early developmental processes in human cardiomyogenesis and future cell replacement strategies. We have analyzed electrophysiological properties of hESC-CMs (HES2) and hiPSC-CMs, derived from reprogrammed adult foreskin fibroblasts that have previously been found to be highly similar in terms of gene expression. In contrast to the similarity found in the expression profile we found substantial differences in action potentials (APs) and sodium currents at late stage (day 60) of in vitro differentiation with higher sodium currents in hiPSC-CMs. Sensitivity to lidocain was considerably reduced in hESC-CMs as compared to hiPSC-CMs, and the effect could not be explained by differences in beating frequency. In contrast, sensitivity to tetrodotoxin (TTX) was higher in hESC-CMs suggesting different contributions of TTX-sensitive and TTX-resistant sodium channels to AP generation. These data point to physiological differences that are not necessarily detected by genomics. We conclude that novel pharmacological screening-assays using hiPSC-CMs need to be applied with some caution.
Collapse
Affiliation(s)
- Xiaowu Sheng
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
- Department of Physiology and German-Chinese Stem Cell Center, Tongji, Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | | | - Filomain Nguemo
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Farooq Ibrahem Mohammad
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
- Biotechnology Research Center, Al Nahrain University, Baghdad, Iraq
| | - Alexey Kuzmenkin
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Kurt Pfannkuche
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
- Clinic and Polyclinic for Paedriatric Cardiology, University of Cologne, Cologne, Germany
| |
Collapse
|
45
|
Abstract
Heart attack remains the leading cause of death in both men and women worldwide. Stem cell-based therapies, including the use of engineered cardiac tissues, have the potential to treat the massive cell loss and pathological remodeling resulting from heart attack. Specifically, embryonic and induced pluripotent stem cells are a promising source for generation of therapeutically relevant numbers of functional cardiomyocytes and engineering of cardiac tissues in vitro. This review will describe methodologies for successful differentiation of pluripotent stem cells towards the cardiovascular cell lineages as they pertain to the field of cardiac tissue engineering. The emphasis will be placed on comparing the functional maturation in engineered cardiac tissues and developing heart and on methods to quantify cardiac electrical and mechanical function at different spatial scales.
Collapse
Affiliation(s)
- Brian Liau
- Department of Biomedical Engineering, Faculty of Cardiology, Duke University, Room 136 Hudson Hall, Durham, NC 27708, USA
| | | | | |
Collapse
|
46
|
Kuboth S, Kramer J, Rohwedel J. Chondrogenic differentiation in vitro of murine two-factor induced pluripotent stem cells is comparable to murine embryonic stem cells. Cells Tissues Organs 2012; 196:481-9. [PMID: 22797361 DOI: 10.1159/000338527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2012] [Indexed: 11/19/2022] Open
Abstract
Differentiation of embryonic stem (ES) cells via embryoid bodies has been established as an appropriate model to study the development of various cell types in vitro. Here, we show that murine induced pluripotent stem (iPS) cells, reprogrammed by exogenous expression of the two transcription factors Oct4 and Klf4 (2F OK iPS), differentiate into chondrocytes in vitro characterized by the appearance of Alcian blue-stained nodules and the expression of cartilage-associated genes and proteins. Quantitatively, the chondrogenic differentiation potential of 2F OK iPS and ES cells was found to be similar. Further, we demonstrate the induction of chondrogenic iPS cell differentiation by certain members of the transforming growth factor-β family (BMP-2, TGF-β(1)). The number of Alcian blue-positive nodules and the expression of the cartilage marker molecule collagen type II increased after application of BMP-2, whereas simultaneous treatment with both BMP-2 and TGF-β(1) showed no significant effect on gene expression.
Collapse
Affiliation(s)
- Sina Kuboth
- Department of Virology and Cell Biology, University of Lübeck, Lübeck, Germany
| | | | | |
Collapse
|
47
|
Gourronc FA, Klingelhutz AJ. Therapeutic opportunities: telomere maintenance in inducible pluripotent stem cells. Mutat Res 2012; 730:98-105. [PMID: 21605571 PMCID: PMC3179558 DOI: 10.1016/j.mrfmmm.2011.05.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Revised: 05/06/2011] [Accepted: 05/08/2011] [Indexed: 12/22/2022]
Abstract
It has been demonstrated that exogenous expression of a combination of transcription factors can reprogram differentiated cells such as fibroblasts and keratinocytes into what have been termed induced pluripotent stem (iPS) cells. These iPS cells are capable of differentiating into all the tissue lineages when placed in the right environment and, in the case of mouse cells, can generate chimeric mice and be transmitted through the germline. Safer and more efficient methods of reprogramming are rapidly being developed. Clearly, iPS cells present a number of exciting possibilities, including disease modeling and therapy. A major question is whether the nuclei of iPS cells are truly rejuvenated or whether they might retain some of the marks of aging from the cells from which they were derived. One measure of cellular aging is the telomere. In this regard, recent studies have demonstrated that telomeres in iPS cells may be rejuvenated. They are not only elongated by reactivated telomerase but they are also epigenetically modified to be similar but not identical to embryonic stem cells. Upon differentiation, the derivative cells turn down telomerase, the telomeres begin to shorten again, and the telomeres and the genome are returned to an epigenetic state that is similar to normal differentiated somatic cells. While these preliminary telomere findings are promising, the overall genomic integrity of reprogrammed cells may still be problematic and further studies are needed to examine the safety and feasibility of using iPS cells in regenerative medicine applications.
Collapse
|
48
|
Induced pluripotent cells in cardiovascular biology: epigenetics, promises, and challenges. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:27-49. [PMID: 22917225 DOI: 10.1016/b978-0-12-398459-3.00002-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cardiovascular diseases are still the leading cause of death worldwide. Despite the improvement shown in the prognosis of patients with acute MI, there remains still a significant mortality risk. Since the main underlying problem after an MI is the loss of cardiomyocytes and microvasculature, treatment strategies aimed at preserving or regenerating myocardial tissue have been examined as potential therapeutic modalities. Toward this goal, many cell types are being investigated as potent sources of cardiomyocytes for cell transplantation. The progress made toward the generation of induced Pluripotent Stem (iPS) cells hold great potential for future use in myocardial repair. We review critical aspects of these cell's potential, such as their generation, their differentiating ability, the known epigenetic mechanisms that allow for their reprogramming, maintenance of pluripotency, their cardiovascular differentiation and therapeutic potential, and the possibility of an epigenetic memory. Understanding the molecular circuitry of these cells will provide a better understanding of their potential as well as limitations in future clinical use.
Collapse
|
49
|
Kraushaar U, Meyer T, Hess D, Gepstein L, L Mummery C, R Braam S, Guenther E. Cardiac safety pharmacology: from human ether-a-gogo related gene channel block towards induced pluripotent stem cell based disease models. Expert Opin Drug Saf 2011; 11:285-98. [DOI: 10.1517/14740338.2012.639358] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
Kujala VJ, Jimenez ZC, Väisänen J, Tanskanen JMA, Kerkelä E, Hyttinen J, Aalto-Setälä K. Averaging in vitro cardiac field potential recordings obtained with microelectrode arrays. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2011; 104:199-205. [PMID: 21645941 DOI: 10.1016/j.cmpb.2011.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 02/13/2011] [Accepted: 04/09/2011] [Indexed: 05/30/2023]
Abstract
Extracellular field potential (FP) recordings with microelectrode arrays (MEAs) from cardiomyocyte cultures offer a non-invasive way of studying the electrophysiological properties of these cells at the population level. Several studies have examined the FP properties of cardiomyocytes of various origins, including stem cell-derived cardiomyocytes. This focus reflects growing importance and interest in the field of MEA. High-quality cardiac FP signals are often difficult to obtain, especially from stem cell-derived cardiomyocyte cultures, which represent an important new field in cardiac electrophysiology. One way to improve the quality of these recordings is to average the cardiac FP signals. To date, however, no studies have examined the effect of averaging on cardiac FP signals. We report here that cardiac FP averaging can yield higher-quality signals than original individual FPs, and therefore promise more accurate detection of different phases and analysis of the cardiac FP signal. Averaged signals improved the signal-to-noise ratio (SNR), and obtaining reliable averages required approximately 50 cardiac cycles. We therefore propose that routine cardiac FP averaging can serve as a tool to compare the effects of different experimental conditions or stimuli on the properties of cardiac FPs.
Collapse
Affiliation(s)
- Ville J Kujala
- Regea - Institute for Regenerative Medicine, University of Tampere and Tampere University Hospital, Tampere, Finland
| | | | | | | | | | | | | |
Collapse
|