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
|
Adegoke TE, Sabinari IW, Usman TO, Abdulkareem TO, Michael OS, Adeyanju OA, Dibia C, Omotoye OO, Oyabambi AO, Olatunji LA. Allopurinol and valproic acid improve cardiac triglyceride and Na +-K +-ATPase activity independent of circulating aldosterone in female rats with glucose intolerance. Arch Physiol Biochem 2022; 128:1283-1289. [PMID: 32447998 DOI: 10.1080/13813455.2020.1767148] [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
Context: Studies have shown that cardiac triglyceride accumulation and impaired Na+-K+-ATPase activity are linked to diabetes- related cardiovascular disease, particularly in women.Objectives: We hypothesised that allopurinol (ALL) and valproic acid (VPA) treatment would improve cardiac triglyceride and Na+-K+-ATPase activity independent of circulating aldosterone in Combined Oral Contraceptive (COC)-induced dysglycemiaMaterials and methods: Rats received COC (1.0 μg ethinylestradiol and 5.0 μg levonorgestrel; po) with or without ALL (1 mg; po) and VPA (20 mg; po) for 6 weeks.Results: COC-treatment led to impaired glucose tolerance, accumulated abdominal fat, dyslipidemia, elevated plasma MDA, PAI-1 and aldosterone levels and also reduced plasma nitric oxide bioavailability and cardiac Na+-K+-ATPase activity. However, either ALL or VPA treatment ameliorated these alterations comparably independent of elevated aldosterone levelDiscussion and conclusion: Our results suggest that either ALL or VPA would improve cardiac TG and Na+-K+-ATPase activity comparably in COC-treated rats, regardless of circulating aldosterone level.
Collapse
Affiliation(s)
- Tolulope E Adegoke
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Isiah W Sabinari
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Taofeek O Usman
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology, Cardiometabolic Research Unit, College of Health Sciences, Osun State University, Osogbo, Nigeria
| | - Toyyib O Abdulkareem
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olugbenga S Michael
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology, Cardiometabolic Research Unit, College of Health and Medical Sciences, Bowen University, Iwo, Nigeria
| | - Oluwaseun A Adeyanju
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology, Cardiometabolic Research Unit, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Chinaza Dibia
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Omotola O Omotoye
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Adewumi O Oyabambi
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A Olatunji
- Department of Physiology, HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
3
|
Portillo Esquivel LE, Zhang B. Application of Cell, Tissue, and Biomaterial Delivery in Cardiac Regenerative Therapy. ACS Biomater Sci Eng 2021; 7:1000-1021. [PMID: 33591735 DOI: 10.1021/acsbiomaterials.0c01805] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death around the world, being responsible for 31.8% of all deaths in 2017 (Roth, G. A. et al. The Lancet 2018, 392, 1736-1788). The leading cause of CVD is ischemic heart disease (IHD), which caused 8.1 million deaths in 2013 (Benjamin, E. J. et al. Circulation 2017, 135, e146-e603). IHD occurs when coronary arteries in the heart are narrowed or blocked, preventing the flow of oxygen and blood into the cardiac muscle, which could provoke acute myocardial infarction (AMI) and ultimately lead to heart failure and death. Cardiac regenerative therapy aims to repair and refunctionalize damaged heart tissue through the application of (1) intramyocardial cell delivery, (2) epicardial cardiac patch, and (3) acellular biomaterials. In this review, we aim to examine these current approaches and challenges in the cardiac regenerative therapy field.
Collapse
Affiliation(s)
| | - Boyang Zhang
- Department of Chemical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4L8, Canada.,School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, Ontaria L8S 4L8, Canada
| |
Collapse
|
4
|
Laiva AL, Venugopal JR, Navaneethan B, Karuppuswamy P, Ramakrishna S. Biomimetic approaches for cell implantation to the restoration of infarcted myocardium. Nanomedicine (Lond) 2015; 10:2907-30. [PMID: 26371367 DOI: 10.2217/nnm.15.124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Compelling evidences accumulated over the years have proven stem cells as a promising source for regenerative medicine. However, the inadequacy with the design of delivery modalities has prolonged the research in realizing an ideal cell-based approach for the regeneration of infarcted myocardium. Currently, some modest improvements in cardiac function have been documented in clinical trials with stem cell treatments, although regenerating a fully functional myocardium remains a dream for cardiac surgeons. This review provides an overview on the significance of stem cell therapy, the current attempts to resolve the drawbacks with the cell implantation approach and the various stratagems adopted with electrospun hybrid nanofibers for implementation in myocardial regenerative therapy.
Collapse
Affiliation(s)
- Ashang Luwang Laiva
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576.,Amity Institute of Nanotechnology, Amity University, Noida, UP, India
| | - Jayarama Reddy Venugopal
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| | - Balchandar Navaneethan
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| | - Priyadharsini Karuppuswamy
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| | - Seeram Ramakrishna
- Center for Nanofibers & Nanotechnology, Nanoscience & Nanotechnology Initiative, Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Block E3, #05-12, 2 Engineering Drive 3, Singapore 117576
| |
Collapse
|
5
|
Yu S, Zhu Y, Li F, Zhang Y, Xia C. Differentiation of human embryonic germ cells and transplantation in rats with acute myocardial infarction. Exp Ther Med 2014; 7:615-620. [PMID: 24520255 PMCID: PMC3919870 DOI: 10.3892/etm.2014.1474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 12/19/2013] [Indexed: 01/24/2023] Open
Abstract
Human embryonic germ cells (hEGCs) are stem cells cultured from primordial germ cells, which reside in human embryonic genital ridges in vivo. In this study, hEGCs were induced to differentiate into cardiomyocytes by treatment with ascorbic acid in vitro and the effects of hEGC transplantation on rat models of acute myocardial infarction (AMI) were investigated. hEGCs were incubated with differentiation medium containing ascorbic acid at various concentrations. Levels of GATA-4 expression were measured to identify the optimal concentration of the inductor. Immunofluorescence microscopy was used to detect the expression of Cx43 on the induced cells. The hEGCs were injected into the myocardium of rats with AMI. The expression levels of MAB1281 and GATA-4 were used to indicate the survival, migration, distribution and differentiation of transplanted cells. The results revealed the positive expression of GATA-4, Cx43 and cardiac troponin T (cTnT) in differentiated cells, and immunocytochemistry showed that transplanted cells highly expressed GATA-4 and MAB1281. hEGCs were successfully induced to differentiate into cardiomyocytes by ascorbic acid in optimal concentrations in vitro and the transplanted hEGCs survived and differentiated into cardiomyocytes.
Collapse
Affiliation(s)
- Shuichang Yu
- Department of Histology and Embryology, Medical College of Soochow University, Suzhou 215123, P.R. China
| | - Yanbo Zhu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, P.R. China
| | - Fang Li
- Department of Histology and Embryology, Medical College of Soochow University, Suzhou 215123, P.R. China
| | - Yujuan Zhang
- Department of Histology and Embryology, Medical College of Soochow University, Suzhou 215123, P.R. China
| | - Chunlin Xia
- Boxi Institute of Clinical Anatomy and Cytoneurobiology Laboratory, Medical College of Soochow University, Suzhou 215123, P.R. China
| |
Collapse
|
6
|
Grajales L, García J, Geenen DL. Induction of cardiac myogenic lineage development differs between mesenchymal and satellite cells and is accelerated by bone morphogenetic protein-4. J Mol Cell Cardiol 2012; 53:382-91. [PMID: 22709559 PMCID: PMC3426454 DOI: 10.1016/j.yjmcc.2012.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Revised: 05/29/2012] [Accepted: 06/08/2012] [Indexed: 10/28/2022]
Abstract
Our aim was to further elucidate the cardiac lineage development of bone marrow-derived mesenchymal stem cells (MSC) and to identify cells which had the potential for cardiac myogenic differentiation when compared to skeletal muscle satellite (Sk-sat) myogenesis. Unlike Sk-sat, MSC expressed the early cardiac markers Nkx2.5 and GATA4. Their expression was significantly increased by culturing MSC with Bone Morphogenetic Protein 4 (BMP4). Enhanced cardiac myogenic lineage differentiation and loss of stem cell characteristics induced by BMP4 were further confirmed by flow cytometry of cells stained for Nkx2.5 and Sca-1 expression. MSC also expressed skeletal genes (MyoG, ssTnI, Sk-Act) early in culture but their expression was suppressed when BMP4 was added from day 0 to day 6 (p<0.05). BMP4 treated MSC also exhibited a 6-fold increase in cTnI expression by day 12 in culture. The average MSC action potential time duration at 90% (APD90) was 32.3±4ms, with some cells exhibiting action potentials closer to Sk-sat APD90 of 13.7±0.9ms. After treatment with BMP4, MSC significantly increased their APD90 to 54.4±7.6ms, shifting from the shorter skeletal-like signature, towards a longer action potential duration more characteristic of a cardiomyocyte signature. Our results show that MSC and Sk-sat exhibit similarities in myogenic lineage development early in culture but that BMP4 clearly enhances cardiac myogenic development, suppresses skeletal myogenesis, and leads to loss of "stemness" in MSC. These findings provide novel information regarding the use of BMP4 to accelerate cardiac myogenic development in harvested MSC and further support the use of MSC in cardiac regenerative therapy.
Collapse
Affiliation(s)
- Liliana Grajales
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | |
Collapse
|
7
|
Haider KH, Ashraf M. Preconditioning approach in stem cell therapy for the treatment of infarcted heart. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 111:323-56. [PMID: 22917238 DOI: 10.1016/b978-0-12-398459-3.00015-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nearly two decades of research in regenerative medicine have been focused on the development of stem cells as a therapeutic option for treatment of the ischemic heart. Given the ability of stem cells to regenerate the damaged tissue, stem-cell-based therapy is an ideal approach for cardiovascular disorders. Preclinical studies in experimental animal models and clinical trials to determine the safety and efficacy of stem cell therapy have produced encouraging results that promise angiomyogenic repair of the ischemically damaged heart. Despite these promising results, stem cell therapy is still confronted with issues ranging from uncertainty about the as-yet-undetermined "ideal" donor cell type to the nonoptimized cell delivery strategies to harness optimal clinical benefits. Moreover, these lacunae have significantly hampered the progress of the heart cell therapy approach from bench to bedside for routine clinical applications. Massive death of donor cells in the infarcted myocardium during acute phase postengraftment is one of the areas of prime concern, which immensely lowers the efficacy of the procedure. An overview of the published data relevant to stem cell therapy is provided here and the various strategies that have been adopted to develop and optimize the protocols to enhance donor stem cell survival posttransplantation are discussed, with special focus on the preconditioning approach.
Collapse
Affiliation(s)
- Khawaja Husnain Haider
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | | |
Collapse
|
8
|
Venugopal JR, Prabhakaran MP, Mukherjee S, Ravichandran R, Dan K, Ramakrishna S. Biomaterial strategies for alleviation of myocardial infarction. J R Soc Interface 2011; 9:1-19. [PMID: 21900319 PMCID: PMC3223634 DOI: 10.1098/rsif.2011.0301] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
World Health Organization estimated that heart failure initiated by coronary artery disease and myocardial infarction (MI) leads to 29 per cent of deaths worldwide. Heart failure is one of the leading causes of death in industrialized countries and is expected to become a global epidemic within the twenty-first century. MI, the main cause of heart failure, leads to a loss of cardiac tissue impairment of left ventricular function. The damaged left ventricle undergoes progressive ‘remodelling’ and chamber dilation, with myocyte slippage and fibroblast proliferation. Repair of diseased myocardium with in vitro-engineered cardiac muscle patch/injectable biopolymers with cells may become a viable option for heart failure patients. These events reflect an apparent lack of effective intrinsic mechanism for myocardial repair and regeneration. Motivated by the desire to develop minimally invasive procedures, the last 10 years observed growing efforts to develop injectable biomaterials with and without cells to treat cardiac failure. Biomaterials evaluated include alginate, fibrin, collagen, chitosan, self-assembling peptides, biopolymers and a range of synthetic hydrogels. The ultimate goal in therapeutic cardiac tissue engineering is to generate biocompatible, non-immunogenic heart muscle with morphological and functional properties similar to natural myocardium to repair MI. This review summarizes the properties of biomaterial substrates having sufficient mechanical stability, which stimulates the native collagen fibril structure for differentiating pluripotent stem cells and mesenchymal stem cells into cardiomyocytes for cardiac tissue engineering.
Collapse
Affiliation(s)
- Jayarama Reddy Venugopal
- Healthcare and Energy Materials Laboratory, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, National University of Singapore, Singapore.
| | | | | | | | | | | |
Collapse
|
9
|
Guo HD, Cui GH, Wang HJ, Tan YZ. Transplantation of marrow-derived cardiac stem cells carried in designer self-assembling peptide nanofibers improves cardiac function after myocardial infarction. Biochem Biophys Res Commun 2010; 399:42-8. [DOI: 10.1016/j.bbrc.2010.07.031] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 07/11/2010] [Indexed: 12/24/2022]
|
10
|
|
11
|
Schabort EJ, Myburgh KH, Wiehe JM, Torzewski J, Niesler CU. Potential Myogenic Stem Cell Populations: Sources, Plasticity, and Application for Cardiac Repair. Stem Cells Dev 2009; 18:813-30. [DOI: 10.1089/scd.2008.0387] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Elske J. Schabort
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - Kathryn H. Myburgh
- Department of Physiological Sciences, University of Stellenbosch, Stellenbosch, South Africa
| | - Juliane M. Wiehe
- Department of Internal Medicine II, University of Ulm, Ulm, Germany
| | - Jan Torzewski
- Cardiovascular Unit, Oberallgäu Kliniken GmbH, Immenstadt, Germany
| | - Carola U. Niesler
- Department of Biochemistry, School of Biochemistry, Genetics, Microbiology, and Plant Pathology, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| |
Collapse
|
12
|
Anderson JE. The satellite cell as a companion in skeletal muscle plasticity: currency, conveyance, clue, connector and colander. ACTA ACUST UNITED AC 2006; 209:2276-92. [PMID: 16731804 DOI: 10.1242/jeb.02088] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Satellite cells are companions to voluntary muscle fibres, and are named for their intimate positional or ;satellite' relationship, as if revolving around fibres, like a satellite moon around the earth. Studies on the nature of at least some satellite cells, including their capabilities for self-renewal and for giving rise to multiple lineages in a stem cell-like function, are exploring the molecular basis of phenotypes described by markers of specialized function and gene expression in normal development, neuromuscular disease and aging. In adult skeletal muscle, the self-renewing capacity of satellite cells contributes to muscle growth, adaptation and regeneration. Muscle remodeling, such as demonstrated by changes in myofibre cross-sectional area and length, nerve and tendon junctions, and fibre-type distribution, occur in the absence of injury and provide broad functional and structural diversity among skeletal muscles. Those contributions to plasticity involve the satellite cell in at least five distinct roles, here described using metaphors for behaviour or the investigator's perspective. Satellite cells are the 'currency' of muscle; have a 'conveyance' role in adaptation by domains of cytoplasm along a myofibre; serve researchers, through a marker role, as 'clues' to various activities of muscle; are 'connectors' that physically, and through signalling and cell-fibre communications, bridge myofibres to the intra- and extra-muscular environment; and are equipped as metabolic and genetic filters or 'colanders' that can rectify or modulate particular signals. While all these roles are still under exploration, each contributes to the plasticity of skeletal muscle and thence to the overall biology and function of an organism. The use of metaphor for describing these roles helps to clarify and scrutinize the definitions that form the basis of our understanding of satellite cell biology: the metaphors provide the construct for various approaches to detect or test the nature of satellite cell functions in skeletal muscle plasticity.
Collapse
Affiliation(s)
- Judy E Anderson
- Department of Human Anatomy and Cell Science, Faculty of Medicine, University of Manitoba, Winnipeg, MB, R3E 0W3, Canada.
| |
Collapse
|
13
|
Tolmachov O, Ma YL, Themis M, Patel P, Spohr H, MacLeod KT, Ullrich ND, Kienast Y, Coutelle C, Peters NS. Overexpression of connexin 43 using a retroviral vector improves electrical coupling of skeletal myoblasts with cardiac myocytes in vitro. BMC Cardiovasc Disord 2006; 6:25. [PMID: 16756651 PMCID: PMC1513252 DOI: 10.1186/1471-2261-6-25] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2005] [Accepted: 06/06/2006] [Indexed: 01/02/2023] Open
Abstract
Background Organ transplantation is presently often the only available option to repair a damaged heart. As heart donors are scarce, engineering of cardiac grafts from autologous skeletal myoblasts is a promising novel therapeutic strategy. The functionality of skeletal muscle cells in the heart milieu is, however, limited because of their inability to integrate electrically and mechanically into the myocardium. Therefore, in pursuit of improved cardiac integration of skeletal muscle grafts we sought to modify primary skeletal myoblasts by overexpression of the main gap-junctional protein connexin 43 and to study electrical coupling of connexin 43 overexpressing myoblasts to cardiac myocytes in vitro. Methods To create an efficient means for overexpression of connexin 43 in skeletal myoblasts we constructed a bicistronic retroviral vector MLV-CX43-EGFP expressing the human connexin 43 cDNA and the marker EGFP gene. This vector was employed to transduce primary rat skeletal myoblasts in optimised conditions involving a concomitant use of the retrovirus immobilising protein RetroNectin® and the polycation transduction enhancer Transfectam®. The EGFP-positive transduced cells were then enriched by flow cytometry. Results More than four-fold overexpression of connexin 43 in the transduced skeletal myoblasts, compared with non-transduced cells, was shown by Western blotting. Functionality of the overexpressed connexin 43 was demonstrated by microinjection of a fluorescent dye showing enhanced gap-junctional intercellular transfer in connexin 43 transduced myoblasts compared with transfer in non-transduced myoblasts. Rat cardiac myocytes were cultured in multielectrode array culture dishes together with connexin 43/EGFP transduced skeletal myoblasts, control non-transduced skeletal myoblasts or alone. Extracellular field action potential activation rates in the co-cultures of connexin 43 transduced skeletal myoblasts with cardiac myocytes were significantly higher than in the co-cultures of non-transduced skeletal myoblasts with cardiac myocytes and similar to the rates in pure cultures of cardiac myocytes. Conclusion The observed elevated field action potential activation rate in the co-cultures of cardiac myocytes with connexin 43 transduced skeletal myoblasts indicates enhanced cell-to-cell electrical coupling due to overexpression of connexin 43 in skeletal myoblasts. This study suggests that retroviral connexin 43 transduction can be employed to augment engineering of the electrocompetent cardiac grafts from patients' own skeletal myoblasts.
Collapse
Affiliation(s)
- Oleg Tolmachov
- Section of Molecular and Cellular Medicine, Division of Biomedical Sciences, Faculty of Life Sciences, Imperial College London, London, UK
| | - Yu-Ling Ma
- Department of Cardiac Electrophysiology, National Heart and Lung Institute, Imperial College London at St. Mary's Hospital, London, UK
| | - Michael Themis
- Section of Molecular and Cellular Medicine, Division of Biomedical Sciences, Faculty of Life Sciences, Imperial College London, London, UK
| | - Pravina Patel
- Department of Cardiac Electrophysiology, National Heart and Lung Institute, Imperial College London at St. Mary's Hospital, London, UK
| | - Hilmar Spohr
- Department of Cardiac Electrophysiology, National Heart and Lung Institute, Imperial College London at St. Mary's Hospital, London, UK
| | - Kenneth T MacLeod
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Nina D Ullrich
- Department of Cardiac Medicine, National Heart and Lung Institute, Imperial College London, London, UK
| | - Yvonne Kienast
- Section of Molecular and Cellular Medicine, Division of Biomedical Sciences, Faculty of Life Sciences, Imperial College London, London, UK
| | - Charles Coutelle
- Section of Molecular and Cellular Medicine, Division of Biomedical Sciences, Faculty of Life Sciences, Imperial College London, London, UK
| | - Nicholas S Peters
- Department of Cardiac Electrophysiology, National Heart and Lung Institute, Imperial College London at St. Mary's Hospital, London, UK
| |
Collapse
|
14
|
Steendijk P, Smits PC, Valgimigli M, van der Giessen WJ, Onderwater EEM, Serruys PW. Intramyocardial injection of skeletal myoblasts: long-term follow-up with pressure–volume loops. ACTA ACUST UNITED AC 2006; 3 Suppl 1:S94-100. [PMID: 16501641 DOI: 10.1038/ncpcardio0416] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Accepted: 10/18/2005] [Indexed: 11/09/2022]
Abstract
The human heart has a limited capacity for self-repair because, unlike most other cells, cardiomyocytes do not regenerate. Therefore, if a substantial number of myocytes is lost after a myocardial infarction, the performance of the heart may become severely limited, leading to a condition of heart failure. Recently, cell transplantation has emerged as a potential therapy for patients with end-stage heart failure. Of the various cell types being investigated for this purpose, skeletal myoblasts are an attractive option, because they are readily available from muscle biopsies and, if autologous cells are used, immunosuppression is not required and ethical issues are avoided. Several studies have shown that the cells can survive and differentiate after transplantation, and promising clinical results have been reported. However, effects of this therapy on left ventricular function remain largely unknown. In the present study, we investigated the long-term hemodynamic effects of intramyocardial injection of autologous skeletal myoblasts in patients with ischemic heart failure. Our findings indicate hemodynamic improvement after follow-up for up to 1 year, which is especially promising in view of the expected decline in left ventricular function in these patients.
Collapse
Affiliation(s)
- Paul Steendijk
- Department of Cardiology, Leiden University Medical Center, Leiden, the Netherlands.
| | | | | | | | | | | |
Collapse
|
15
|
Kofidis T, Lebl DR, Martinez EC, Hoyt G, Tanaka M, Robbins RC. Novel injectable bioartificial tissue facilitates targeted, less invasive, large-scale tissue restoration on the beating heart after myocardial injury. Circulation 2006; 112:I173-7. [PMID: 16159811 DOI: 10.1161/circulationaha.104.526178] [Citation(s) in RCA: 136] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Implantation of bioartificial patches distorts myocardial geometry, and functional improvement of the recipient heart is usually attributed to reactive angiogenesis around the graft. With the liquid bioartificial tissue compound used in this study, we achieved targeted large-scale support of the infarcted left ventricular wall and improvement of heart function. METHODS AND RESULTS A liquid compound consisting of growth factor-free Matrigel and 10(6) green fluorescent protein (GFP)-positive mouse (129sv) embryonic stem cells (ESCs) was generated and injected into the area of ischemia after ligation of the left anterior descending artery in BALB/c mice (group I). Left anterior descending artery-ligated mice (group II) and mice with Matrigel (group III) or ESC treatment alone (group IV) were used as the control groups (n=5 in all groups). The hearts were harvested for histology 2 weeks later after echocardiographic assessment with a 15-MHz probe. The liquid injectable tissue solidified at body temperature and retained the geometry of the infarcted lateral wall. Immunofluorescence stains revealed voluminous GFP grafts. The quality of restoration (graft/infarct area ratio) was 45.5+/-10.8% in group I and 29.1+/-6.7% in group IV (P=0.034). ESCs expressed connexin 43 at intercellular contact sites. The mice treated with the compound had a superior heart function compared with the controls (P<0.0001 by ANOVA/Bonferroni test; group I: 27.1+/-5.4, group II:11.9+/-2.4, group III:16.2+/-2.8, group IV: 19.1+/-2.7). CONCLUSIONS Injectable bioartificial tissue restores the heart's geometry and function in a targeted and nondistorting fashion. This new method paves the way for novel interventional approaches to myocardial repair, using both stem cells and matrices.
Collapse
MESH Headings
- Animals
- Bioartificial Organs
- Chemical Phenomena
- Chemistry, Physical
- Collagen
- Connexin 43/biosynthesis
- Drug Combinations
- Genes, Reporter
- Genes, Synthetic
- Green Fluorescent Proteins/analysis
- Green Fluorescent Proteins/genetics
- Heart Ventricles
- Injections, Intramuscular
- Laminin
- Mice
- Mice, Inbred BALB C
- Myocardial Contraction
- Myocardial Infarction/complications
- Myocardial Infarction/physiopathology
- Myocardial Infarction/surgery
- Peptide Elongation Factor 1/genetics
- Promoter Regions, Genetic
- Proteoglycans
- Stem Cell Transplantation/methods
- Stem Cells/metabolism
- Tissue Engineering
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Dysfunction, Left/surgery
- Ventricular Function, Left
Collapse
Affiliation(s)
- Theo Kofidis
- Cardiovascular Surgery, Stanford University Medical School, Stanford, California, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Mathew AJ, Baust JM, Van Buskirk RG, Baust JG. Cell Preservation in Reparative and Regenerative Medicine: Evolution of Individualized Solution Composition. ACTA ACUST UNITED AC 2004; 10:1662-71. [PMID: 15684675 DOI: 10.1089/ten.2004.10.1662] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The expanding complexity of biologics banked for therapeutic applications necessitates the development of improved preservation technologies for support of the emerging fields of reparative and regenerative medicine. Currently, a number of media or "solutions" are utilized for the preservation of biologics. Given the diversity of cell systems utilized in the regenerative medicine arena, we hypothesized that the development of unique (individualized) preservation solutions designed to meet the distinct molecular biological requirements of individual systems would provide for enhanced and extended preservation. To evaluate this hypothesis, coronary artery smooth muscle cells (CASMCs), coronary artery endothelial cells (CAECs), hepatic cells (C3A), and skeletal muscle cells (SKMCs) were hypothermically preserved for 2 to 7 days at 4 degrees C in either cell culture medium, University of Wisconsin Solution (UW or ViaSpan), or HypoThermosol (HTS) variants. Cells were then assayed for viability, using the alamarBlue assay as well as calcein-AM, subsequent to their return to normothermic (37 degrees C) temperatures for up to 5 days. CASMC viability was best maintained when preserved in HTS plus Trolox/EDTA, CAEC viability was highest when preserved in HTS plus Trolox, SKMCs stored in HTS plus Trolox/RGD demonstrated enhanced viability, and C3A cells were best preserved in HTS plus FK041. The data suggest that solution compositions that address the differences in cell death mechanisms limiting preservation efficacy can result in targeted improvement matched to specific cell types. These observations support the custom solution hypothesis of cell and tissue preservation.
Collapse
Affiliation(s)
- Aby J Mathew
- Institute of Biomedical Technology, State University of New York, Binghamton, New York, USA.
| | | | | | | |
Collapse
|
17
|
Geng YJ. Molecular mechanisms for cardiovascular stem cell apoptosis and growth in the hearts with atherosclerotic coronary disease and ischemic heart failure. Ann N Y Acad Sci 2004; 1010:687-97. [PMID: 15033813 DOI: 10.1196/annals.1299.126] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In the heart with atherosclerotic coronary disease, chronic ischemia causes progressive loss of cardiovascular cells and ultimately triggers myocardial dysfunctions or heart failure. Various types of stem cells from embryonic and adult tissues have potentials for regenerating functional cardiovascular cells in the heart undergoing ischemic injury. However, native or exogenous stem cells in the ischemic hearts are exposed to various proapoptotic or cytotoxic factors. Furthermore, during repopulation and differentiation, certain numbers of newly produced cells may die by apoptosis during neocardiovascular tissue remodeling and morphogenesis. Embryonic and adult stem cells may have different life spans, as being programmed genetically to apoptosis. The endogenous and environmental factors play important roles in regulation of stem cells, including inflammatory cytokines, growth factors, surface receptors, proteolytic enzymes, mitochondrial respiration, nuclear proteins, telomerase activities, hypoxia-responding proteins, and stem cell-host cell interaction. Clarification of the molecular mechanisms may help us understand and design stem cell therapies.
Collapse
Affiliation(s)
- Yong-Jian Geng
- Center for Cardiovascular Biology and Atherosclerosis, Department of Internal Medicine, The University of Texas, Health Science Center at Houston, Medical School, Texas Heart Institute, Houston, Texas 77030, USA.
| |
Collapse
|
18
|
Abstract
Hematopoietic stem cell (HSC) plasticity and its clinical application have been studied profoundly in the past few years. Recent investigations indicate that HSC and other bone marrow stem cells can develop into other tissues. Because of the high morbidity and mortality of myocardial infarction and other heart disorders, myocardial regeneration is a good example of the clinical application of HSC plasticity in regenerative medicine. Preclinical studies in animals suggest that the use of this kind of treatment can reconstruct heart blood vessels, muscle, and function. Some clinical study results have been reported in the past 2 years. In 2003, reports of myocardial regeneration treatment increased significantly. Other studies include observations on the cell surface markers of transplanted cells and treatment efficacy. Some investigations, such as HSC testing, have focused on clinical applications using HSC plasticity and bone marrow transplantation to treat different types of disorders. In this review, we focus on the clinical application of bone marrow cells for myocardial regeneration.
Collapse
Affiliation(s)
- Fu-Sheng Wang
- R&D Department, Sysmex America, Mundelein, Illinois 60060, USA.
| | | |
Collapse
|
19
|
Abstract
Atherosclerotic vascular disease becomes a clinical problem when there is sufficient atherosclerotic plaque burden and/or endothelial dysfunction to cause a limitation of nutrient blood flow to tissues. However, once myocardial infarction has occurred, there is little, if any, way to stimulate the growth of new blood vessels or cardiac muscle to replace that which has been lost. The potential use of hematopoietic stem cells (HSCs) to treat cardiovascular disease has recently been suggested from preclinical and clinical studies. HSCs are precursors of all the blood cells, but they may also give rise to cells of the vascular system, endothelial cells in the form of endothelial progenitor cells (EPCs). Clinical trials have been conducted in patients with either acute myocardial infarction or limb ischemia to determine the initial effectiveness and safety of this treatment approach. These studies demonstrated the potential clinical effectiveness of this stem cell approach to the treatment of patients with acute myocardial ischemia and limb ischemia. Today, more preclinical studies are planned to elucidate the mechanism by which transplanted stem cells can home and differentiate into these endothelial cells and cardiac muscle cells. At the same time, new clinical trials are planned to evaluate both chronic, stable as well as acute myocardial ischemia and limb ischemia with CD34+ and CD133+ stem cells, as well as with further selected EPCs and mesenchymal stem cells.
Collapse
Affiliation(s)
- David L Amrani
- Baxter Healthcare, RLT-12 Route 120 and Wilson Rd, Round Lake, IL 60073, USA.
| | | |
Collapse
|
20
|
Abstract
The death of highly vulnerable cardiomyocytes during ischemia leads to cardiac dysfunction, including heart failure. Due to limited proliferation of adult mammalian cardiomyocytes, the dead myocardium is replaced by noncontractile fibrotic tissue. Introducing exogenous cells to participate in the regeneration of infarcted myocardium has thus been proposed as a novel therapeutic approach. In view of the availability of various xenogeneic cells and fewer ethical and political concerns that surround human embryonic stem cells and fetal cardiomyocytes, cellular xenotransplantation may be a potential alternative approach for cardiac repair in humans. However, one of the most daunting challenges of xenotransplantation is immunorejection. This article summarizes the progress in cellular xenotransplantation for cardiac repair in experimental settings and the current understanding of possible immune responses following the engraftment of xenogeneic cells. The public attitude towards xenotransplantation is reportedly more favorable to receiving cells or tissues than a whole organ, but many scientific obstacles need to be overcome before the utilization of xenogeneic cells for cardiac repair in patients with heart disease becomes applicable to clinical practice.
Collapse
Affiliation(s)
- Yong-Fu Xiao
- Stem Cell Research Laboratory, The Charles A. Dana Research Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| | | | | |
Collapse
|
21
|
|