14 results on '"Shim, Winston"'
Search Results
2. List of Contributors
- Author
-
Abir, Tanvir, primary, Ahmad, Sk Akhtar, additional, Athar, Mohammad, additional, Bailey, Kathryn A., additional, Basha, Riyaz, additional, Calatayud, Marta, additional, Camacho, Lucy Mar, additional, Campos, Isaac, additional, Davis, Thomas A., additional, del Valle, Manel, additional, Deng, Shuguang, additional, Flora, Govinder, additional, Flora, Swaran J.S., additional, Fowler, Bruce A., additional, Fry, Rebecca C., additional, Ghosh, Jyotirmoy, additional, Guan, Huai, additional, Guha Mazumder, D.N., additional, Hossain, AKM Akbar, additional, Hunt, Katherine Marchiony, additional, Islam, Laila N., additional, Islam, Md.Rafiqul, additional, Isokpehi, Raphael D., additional, Kalia, Kiran, additional, Khambholja, Devang B., additional, Khan, Manzurul Haque, additional, Kile, Molly L., additional, Kumar, Sanjay, additional, Laparra Llopis, José Moisés, additional, Lee, Janice S., additional, Li, Shuangyue, additional, Luo, Jiaohua, additional, Maiti, Smarajit, additional, Mandal, Badal Kumar, additional, Mazumdar, Maitreyi, additional, Mehta, Ashish, additional, Milton, Abul Hasnat, additional, Mittal, Megha, additional, Pandey, Sarita, additional, Piao, Fengyuan, additional, Ponnusamy, Sundaravadivelnathan, additional, Rahman, Bayzidur, additional, Rai, Lal Chand, additional, Rai, Rashmi, additional, Ramachandra, Chrishan J.A., additional, Ramasamy, Santhini, additional, Ramsey, Kathryn, additional, Reddy, G.Rajarami, additional, Sabath, Ernesto, additional, Santra, Amal, additional, Shim, Winston, additional, Shu, Weiqun, additional, Sil, Parames C., additional, Singh, Minni, additional, Srivastava, Ritesh Kumar, additional, Tareq, Shafi M., additional, Tchounwou, Paul B., additional, Udensi, Udensi K., additional, and Yedjou, Clement G., additional
- Published
- 2015
- Full Text
- View/download PDF
3. Fatty acid metabolism driven mitochondrial bioenergetics promotes advanced developmental phenotypes in human induced pluripotent stem cell derived cardiomyocytes.
- Author
-
Ramachandra CJA, Mehta A, Wong P, Ja KPMM, Fritsche-Danielson R, Bhat RV, Hausenloy DJ, Kovalik JP, and Shim W
- Subjects
- Cells, Cultured, Humans, Induced Pluripotent Stem Cells ultrastructure, Mitochondria ultrastructure, Myocytes, Cardiac ultrastructure, Phenotype, Energy Metabolism physiology, Fatty Acids metabolism, Induced Pluripotent Stem Cells metabolism, Lipid Metabolism physiology, Mitochondria metabolism, Myocytes, Cardiac metabolism
- Abstract
Background: Preferential utilization of fatty acids for ATP production represents an advanced metabolic phenotype in developing cardiomyocytes. We investigated whether this phenotype could be attained in human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) and assessed its influence on mitochondrial morphology, bioenergetics, respiratory capacity and ultra-structural architecture., Methods and Results: Whole-cell proteome analysis of day 14 and day 30-CMs maintained in glucose media revealed a positive influence of extended culture on mitochondria-related processes that primed the day 30-CMs for fatty acid metabolism. Supplementing the day 30-CMs with palmitate/oleate (fatty acids) significantly enhanced mitochondrial remodeling, oxygen consumption rates and ATP production. Metabolomic analysis upon fatty acid supplementation revealed a β-oxidation fueled ATP elevation that coincided with presence of junctional complexes, intercalated discs, t-tubule-like structures and adult isoform of cardiac troponin T. In contrast, glucose-maintained day 30-CMs continued to harbor underdeveloped ultra-structural architecture and more subdued bioenergetics, constrained by suboptimal mitochondria development., Conclusion: The advanced metabolic phenotype of preferential fatty acid utilization was attained in hiPSC-CMs, whereby fatty acid driven β-oxidation sustained cardiac bioenergetics and respiratory capacity resulting in ultra-structural and functional characteristics similar to those of developmentally advanced cardiomyocytes. Better understanding of mitochondrial bioenergetics and ultra-structural adaptation associated with fatty acid metabolism has important implications in the study of cardiac physiology that are associated with late-onset mitochondrial and metabolic adaptations., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
4. The KCNH2-IVS9-28A/G mutation causes aberrant isoform expression and hERG trafficking defect in cardiomyocytes derived from patients affected by Long QT Syndrome type 2.
- Author
-
Mura M, Mehta A, Ramachandra CJ, Zappatore R, Pisano F, Ciuffreda MC, Barbaccia V, Crotti L, Schwartz PJ, Shim W, and Gnecchi M
- Subjects
- Female, Gene Expression, Humans, Induced Pluripotent Stem Cells metabolism, Induced Pluripotent Stem Cells pathology, Long QT Syndrome pathology, Male, Myocytes, Cardiac pathology, Pedigree, Protein Transport physiology, ERG1 Potassium Channel biosynthesis, ERG1 Potassium Channel genetics, Long QT Syndrome genetics, Long QT Syndrome metabolism, Mutation genetics, Myocytes, Cardiac metabolism
- Abstract
Background: Long QT Syndrome type 2 (LQT2) is caused by mutations in the KCNH2 gene that encodes for the α-subunit (hERG) of the ion channel conducting the rapid delayed rectifier potassium current (I
Kr ). We have previously identified a disease causing mutation (IVS9-28A/G) in the branch point of the splicing of KCNH2 intron 9. However, the mechanism through which this mutation causes the disease is unknown., Methods and Results: We generated human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) from fibroblasts of two IVS9-28A/G mutation carriers. IVS9-28A/G iPSC-CMs showed prolonged repolarization time, mimicking what observed at the ECG level in the same patients. The expression of the full-length ERG1a isoform resulted reduced, whereas the C-terminally truncated ERG1aUSO isoform was upregulated in mutant iPSC-CMs, with consequent alteration of the physiological ERG1aUSO /ERG1a ratio. Importantly, we observed an impairment of hERG trafficking to the cell membrane. The severity of the alterations in hERG expression and trafficking correlated with the clinical severity of the disease in the two patients under study. Finally, we were able to revert the trafficking defect and reduce the repolarization duration in LQT2 iPSC-CMs using the proteasome inhibitor ALLN., Conclusion: Our results highlight the key role of the KCNH2 intron 9 branch point in the regulation of KCNH2 isoform expression and hERG channel function, and allow to categorize the IVS9-28A/G mutation as LQT2 class 2 mutation. These findings may result in a more personalized clinical management of IVS9-28A/G mutation carriers., (Copyright © 2017 Elsevier B.V. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
5. Molecular pathogenesis of Marfan syndrome.
- Author
-
Ramachandra CJ, Mehta A, Guo KW, Wong P, Tan JL, and Shim W
- Subjects
- Fibrillin-1, Fibrillins, Humans, Induced Pluripotent Stem Cells physiology, Marfan Syndrome genetics, Matrix Metalloproteinases metabolism, Microfilament Proteins genetics, Transforming Growth Factor beta metabolism, Marfan Syndrome etiology
- Abstract
Marfan syndrome (MFS) is a genetic disorder that affects multiple organs. Mortality imposed by aortic aneurysm and dissections represent the most serious clinical manifestation of MFS. Progressive pathological aortic root enlargement as the result of degeneration of microfibril architecture and consequential loss of extracellular matrix integrity due to fibrillin-1 (FBN1) mutations are commonly diagnosed clinical manifestations of MFS. However, overlapping clinical manifestations with other aneurysmal disorders present a significant challenge in early and accurate diagnosis of MFS. While FBN1 mutations, abnormal transforming growth factor-β signaling and dysregulated matrix metalloproteinases have been implicated in MFS, clinically accepted risk-stratifying biomarkers have yet to be reliably identified. In this review, we summarize current consensus and recent insights in the understanding of MFS pathogenesis. Finally, we introduce the application of induced pluripotent stem cells (iPSCs) as cellular models for MFS and its potential as a novel platform into providing better appreciation of mechanisms underlying MFS diverse manifestations in the cardiovascular system., (Copyright © 2015. Published by Elsevier Ireland Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
6. Hydrogen sulfide augments the proliferation and survival of human induced pluripotent stem cell-derived mesenchymal stromal cells through inhibition of BKCa.
- Author
-
Zhao Y, Wei H, Kong G, Shim W, and Zhang G
- Subjects
- Apoptosis drug effects, Caspase 8 biosynthesis, Cell Hypoxia drug effects, Cell Line, Cell Proliferation drug effects, Cell Survival drug effects, Cell- and Tissue-Based Therapy, Humans, Indoles pharmacology, Large-Conductance Calcium-Activated Potassium Channels drug effects, Large-Conductance Calcium-Activated Potassium Channels metabolism, Patch-Clamp Techniques, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation drug effects, Potassium Channel Blockers pharmacology, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction drug effects, bcl-2-Associated X Protein biosynthesis, Hydrogen Sulfide pharmacology, Induced Pluripotent Stem Cells drug effects, Large-Conductance Calcium-Activated Potassium Channels antagonists & inhibitors, Mesenchymal Stem Cells drug effects
- Abstract
Background: Hydrogen sulfide (H2S) is an endogenously generated gaseous transmitter known for its cytoprotective effect mediated by the PI3K-Akt signaling pathway. Human induced pluripotent stem cell (hiPSC)-derived mesenchymal stromal cells (MSCs), or hiPSC-MSCs, represent an alternative source of MSCs for autologous cell therapy. The big-conductance Ca(2+)-activated outward K(+) currents (BKCa), known to mediate cell proliferation, have been detected in >80% of hiPSC-MSCs. The present study aimed to explore the effect of H2S on survival and proliferation of hiPSC-MSCs and investigate the mediatory role of BKCa., Methods: Effects of H2S on proliferation and survival of hiPSC-MSCs were measured by 5-bromo-2-deoxyuridine incorporation, population doubling and cell cycle assays, and by 3-(4,5)-dimethylthiahiazo(-z-y1)-3,5-di-phenytetrazoliumromide assay and 4'-6-diamidino-2-phenylindole staining, respectively. BKCa was recorded by means of the whole-cell patch-clamp technique. The expressions of KCa 1.1 (encoding BKCa) and apoptosis-related genes were measured by reverse transcriptase-polymerase chain reaction. The phosphorylation of Akt was assessed by Western blot analysis., Results: Exogenously administered NaHS (an H2S donor, 50-300 μmol/L) significantly promoted proliferation of hiPSC-MSCs. NaHS prevented the hypoxia-induced apoptosis and suppressed BKCa currents without altering the expression levels of α- and β-KCa 1.1. In addition, NaHS increased the phosphorylation of Akt and decreased the expression of Caspase 8 and Bax in hiPSC-MSCs. Paxilline (1 μmol/L), a BKCa blocker, showed similar effects on promoting cell proliferation and phosphorylation of Akt and suppression of apoptotic genes in hiPSC-MSCs., Conclusions: Our data confirmed that H2S arguments the proliferation and survival of hiPSC-MSCs through activation of the PI3K-Akt pathway and that such effects could be mediated through inhibition of BKCa., (Copyright © 2013 International Society for Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
7. Modeling type 3 long QT syndrome with cardiomyocytes derived from patient-specific induced pluripotent stem cells.
- Author
-
Ma D, Wei H, Zhao Y, Lu J, Li G, Sahib NB, Tan TH, Wong KY, Shim W, Wong P, Cook SA, and Liew R
- Subjects
- Action Potentials physiology, Child, Child, Preschool, Dermis cytology, Electrocardiography, Female, Fibroblasts cytology, Fibroblasts physiology, Genotype, Heart Arrest genetics, Heart Arrest physiopathology, Humans, Long QT Syndrome genetics, Membrane Potentials physiology, Mexiletine pharmacology, Myocytes, Cardiac drug effects, NAV1.5 Voltage-Gated Sodium Channel genetics, Patch-Clamp Techniques, Voltage-Gated Sodium Channel Blockers pharmacology, Chromosomes, Human, Pair 3, Long QT Syndrome physiopathology, Myocytes, Cardiac cytology, Myocytes, Cardiac physiology, Pluripotent Stem Cells cytology
- Abstract
Background: Type 3 long QT syndrome (LQT3) is the third most common form of LQT syndrome and is characterized by QT-interval prolongation resulting from a gain-of-function mutation in SCN5A. We aimed to establish a patient-specific human induced pluripotent stem cell (hiPSC) model of LQT3, which could be used for future drug testing and development of novel treatments for this inherited disorder., Methods and Results: Dermal fibroblasts obtained from a patient with LQT3 harboring a SCN5A mutation (c.5287G>A; p.V1763M) were reprogrammed to hiPSCs via repeated transfection of mRNA encoding OCT-4, SOX-2, KLF-4, C-MYC and LIN-28. hiPSC-derived cardiomyocytes (hiPSC-CMs) were obtained via cardiac differentiation. hiPSC-CMs derived from the patient's healthy sister were used as a control. Compared to the control, patient hiPSC-CMs exhibited dominant mutant SCN5A allele gene expression, significantly prolonged action potential duration or APD (paced CMs of control vs. patient: 226.50 ± 17.89 ms vs. 536.59 ± 37.1 ms; mean ± SEM, p < 0.005), an increased tetrodotoxin (TTX)-sensitive late or persistent Na(+) current (control vs. patient: 0.65 ± 0.11 vs. 3.16 ± 0.27 pA/pF; n = 9, p < 0.01), a positive shift of steady state inactivation and a faster recovery from inactivation. Mexiletine, a NaV1.5 blocker, reversed the elevated late Na(+) current and prolonged APD in LQT3 hiPSC-CMs., Conclusions: We demonstrate that hiPSC-CMs derived from a LQT3 patient recapitulate the biophysical abnormalities that define LQT3. The clinical significance of such an in vitro model is in the development of novel therapeutic strategies and a more personalized approach in testing drugs on patients with LQT3., (© 2013.)
- Published
- 2013
- Full Text
- View/download PDF
8. Critical path in cardiac stem cell therapy: an update on cell delivery.
- Author
-
Shim W, Mehta A, Wong P, Chua T, and Koh TH
- Subjects
- Administration, Intravenous, Heart, Heart Failure therapy, Humans, Myocardial Infarction therapy, Regenerative Medicine, Cardiomyopathies therapy, Regeneration, Stem Cell Transplantation methods, Stem Cells
- Abstract
Despite optimal medical therapy, cardiovascular disease remains a leading cause of morbidity and mortality worldwide. One emerging therapeutic approach for treatment of cardiomyopathies is stem cell therapy. Use of stem cells for regenerative medicine has quickly evolved over the last decade. On the basis of encouraging pre-clinical results, stem cell therapy has developed rapidly into a potentially promising treatment for ischemic heart disease, myocardial infarction and congestive heart failure. In this review, we summarize the current state-of-the-art of stem cell therapy and compare collective experiences gleaned from trials using intravenous, intra-coronary and intra-myocardial delivery in exacting credible benefits. We discuss implications of clinical outcomes reported in relation to the delivered stem cells as probable destiny therapy for cardiovascular repair., (Copyright © 2013 International Society for Cellular Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
9. Ontogenic development of cardiomyocytes derived from transgene-free human induced pluripotent stem cells and its homology with human heart.
- Author
-
Sequiera GL, Mehta A, Ooi TH, and Shim W
- Subjects
- Animals, Biomarkers metabolism, Humans, Isoproterenol pharmacology, Mice, Mice, SCID, Microelectrodes, Myocytes, Cardiac drug effects, Quinidine pharmacology, Real-Time Polymerase Chain Reaction, Sodium metabolism, Verapamil pharmacology, Cell Differentiation physiology, Gene Expression Regulation physiology, Induced Pluripotent Stem Cells metabolism, Myocytes, Cardiac metabolism
- Abstract
Aim: Reprogramming of somatic cells utilizing viral free methods provide a remarkable method to generate human induced pluripotent stem cells (hiPSCs) for regenerative medicine. In this study, we evaluate developmental ontogeny of cardiomyocytes following induced differentiation of hiPSCs., Main Methods: Fibroblasts were reprogrammed with episomal vectors to generate hiPSC and were subsequently differentiated to cardiomyocytes. Ontogenic development of cardiomyocytes was studied by real-time PCR., Key Findings: Human iPSCs derived from episomal based vectors maintain classical pluripotency markers, generate teratomas and spontaneously differentiate into three germ layers in vitro. Cardiomyogenic induction of these hiPSCs efficiently generated cardiomyocytes. Ontogenic gene expression studies demonstrated that differentiation of cardiomyocytes was initiated by increased expression of mesodermal markers, followed by early cardiac committed markers, structural and ion channel genes. Furthermore, our correlation analysis of gene expression studies with human heart demonstrated that pivotal structural genes like cardiac troponin, actinin, myosin light chain maintained a high correlation with ion channel genes indicating coordinated activation of cardiac transcriptional machinery. Finally, microelectrode recordings show that these cardiomyocytes could respond aptly to pharmacologically active drugs. Cardiomyocytes showed a chronotropic response to isoproterenol, reduced Na(+) influx with quinidine, prolongation of beating rate corrected field potential duration (cFPD) with E-4031 and reduced beating frequency and shortened cFPD with verapamil., Significance: Our study shows that viral free hiPSCs efficiently differentiate into cardiomyocytes with cardiac-specific molecular, structural, and functional properties that recapitulate developmental ontogeny of cardiogenesis. These results, coupled with the potential to generate patient-specific hiPSC lines hold great promise for the development of in vitro platform for drug pharmacogenomics; disease modeling and regenerative medicine., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
10. One-step derivation of cardiomyocytes and mesenchymal stem cells from human pluripotent stem cells.
- Author
-
Wei H, Tan G, Manasi, Qiu S, Kong G, Yong P, Koh C, Ooi TH, Lim SY, Wong P, Gan SU, and Shim W
- Subjects
- Animals, Biomarkers metabolism, Cell Differentiation, Cell Line, Cell Lineage, Cell Membrane metabolism, Cell Proliferation, Cell Transformation, Neoplastic pathology, Chromosome Aberrations, Female, Flow Cytometry, Humans, Karyotyping, Kinetics, Mesenchymal Stem Cells metabolism, Mice, Mice, SCID, Multipotent Stem Cells cytology, Myocytes, Cardiac physiology, Neovascularization, Physiologic, Telomerase metabolism, Wound Healing, Cell Culture Techniques methods, Induced Pluripotent Stem Cells cytology, Mesenchymal Stem Cells cytology, Myocytes, Cardiac cytology
- Abstract
Cardiomyocytes (CMs) and mesenchymal stem cells (MSCs) are important cell types for cardiac repair post myocardial infarction. Here we proved that both CMs and MSCs can be simultaneously generated from human induced pluripotent stem cells (hiPSCs) via a pro-mesoderm differentiation strategy. Two hiPSC lines, hiPSC (1) and hiPSC (2) were generated from human dermal fibroblasts using OCT-4, SOX-2, KLF-4, c-Myc via retroviral-based reprogramming. H9 human embryonic stem cells (hESCs) served as control. CMs and MSCs were co-generated from hiPSCs and hESCs via embryoid body-dependent cardiac differentiation protocol involving a serum-free and insulin-depleted medium containing a p38 MAPK inhibitor, SB 203580. Comparing to bone marrow and umbilical cord blood-derived MSCs, hiPSC-derived MSCs (iMSCs) expressed common MSC markers and were capable of adipogenesis, osteogenesis and chondrogenesis. Moreover, iMSCs continuously proliferated for more than 32 population doublings without cellular senescence and showed superior pro-angiogenic and wound healing properties. In summary, we generated a large number of homogenous MSCs in conjunction with CMs in a low-cost and efficient one step manner. Functionally competent CMs and MSCs co-generated from hiPSCs may be useful for autologous cardiac repair., (Copyright © 2012 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
11. Dose-dependent systolic contribution of differentiated stem cells in post-infarct ventricular function.
- Author
-
Shim WS, Tan G, Gu Y, Qian L, Li S, Chung YY, Lim SY, Sim E, Chuah SC, and Wong P
- Subjects
- Animals, Cells, Cultured, Disease Models, Animal, Female, Humans, Mesenchymal Stem Cells, Myocytes, Cardiac cytology, Rats, Rats, Wistar, Systole, Cell Differentiation, Mesenchymal Stem Cell Transplantation, Myocardial Infarction surgery, Myocytes, Cardiac transplantation, Ventricular Function, Left
- Abstract
Background: Differentiation of bone marrow stem cells toward cardiomyocytes has been widely reported in vitro. However, optimum cell types and mechanisms leading to functional improvement in cardiac cell therapy remain unresolved. There is limited evidence showing a dose-dependent effect of transplanted cells in contributing to functional recovery. This study showed that cell transplantation of differentiated cardiomyocyte-like cells (CLCs) and undifferentiated mesenchymal stem cells (MSCs) dose-dependently improved left ventricular function in a rat myocardial infarction model., Methods: At 1 week after infarction in Wistar rats, 1 × 10(6) MSCs (n = 9) or CLCs (n = 9) and 5 × 10(6) MSCs (n = 18) or CLCs (n = 15) were injected into peri-infarcted myocardium to study their effect after 6 weeks., Results: High-dose CLCs exhibited a dose-response that was significantly more effective than MSCs in recovering cardiac contractility. Superiority of CLCs over MSCs was demonstrated in load-independent measurement of the end-systolic pressure-volume relationship and pre-load recruitable stroke work, but not in the end-diastolic pressure-volume relationship. These findings showed a unique systolic role of CLCs in contractility recovery. Functional improvement mediated by MSCs was mainly derived from preservation of endogenous myocyte function and restriction of chamber dilatation by enhancing intramyocardial angiogenesis during post-infarct ventricular remodeling. Engrafted CLCs showed better survival, were strategically integrated into myofiber-associated collagen V matrix, and exhibited mature sarcomeric cross-striations. Vascular differentiation, but not cardiac, was observed with MSCs., Conclusion: These cell type-specific effects suggest that committing stem cells to a cardiac phenotype ex vivo promoted mechanical and functional integration of CLCs into the myofibrillar syncytium of infarcted myocardium., (Crown Copyright © 2010. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
12. Differential effect of myocardial matrix and integrins on cardiac differentiation of human mesenchymal stem cells.
- Author
-
Tan G, Shim W, Gu Y, Qian L, Chung YY, Lim SY, Yong P, Sim E, and Wong P
- Subjects
- Animals, Bone Marrow Cells cytology, Bone Marrow Cells physiology, Cells, Cultured, Collagen metabolism, Female, Gene Expression Profiling, Hemodynamics, Humans, Mesenchymal Stem Cell Transplantation, Mesenchymal Stem Cells cytology, Myocardium cytology, Rats, Rats, Wistar, Cell Differentiation physiology, Extracellular Matrix metabolism, Integrins metabolism, Mesenchymal Stem Cells physiology, Myocardium metabolism
- Abstract
Dysregulation of matrix synthesis during myocardial fibrosis in post-infarct ventricular remodeling contributes to ventricular dysfunction. Bone marrow stem cell transplantation prevents functional deterioration following myocardial infarction. However, effect of myocardial extracellular matrix (ECM) on stem cell differentiation is poorly understood. We investigate the role of collagen matrices and integrin system in cardiac differentiation and engraftment of stem cells in infarcted myocardium. Sternum-derived bone marrow mesenchymal stem cells (MSCs) were differentiated into cardiomyocyte-like cells (CLCs). They were characterized using RT-PCR, immunofluorescence, flow cytometry and functional integrin neutralization assays. CLCs were injected into peri-infarct borders of injured myocardium of Wistar rats one week following left anterior descending (LAD) artery ligation. Cardiac function was analyzed via pressure-volume relationships. Cardiac differentiated CLCs displayed collagen V specificity, which was absent in undifferentiated MSCs. Collagen V, but not collagen I matrix, promoted attachment, proliferation and cardiac differentiation of CLCs. In contrast to beta(1), alpha(v) integrin contributed minimally in the attachment of CLCs on collagen matrices. However, inhibition of alpha(v)beta(3,) but not alpha(2)beta(1) integrin, selectively attenuated troponin T, sarcomeric alpha-actin and ryanodine 2 receptor gene expression in CLCs. Both MSC and CLC transplantation prevented chamber dilatation and improved contractile function. However, systolic activity in MSC transplanted animals was accompanied by heightened wall stress as demonstrated by elevated myocardial end-diastolic pressure and prolonged tissue relaxation time. Localization of CLCs in the vicinity of collagen V-expressing myofibers promoted their integration into cardiac syncytium. CLCs may facilitate hemodynamic recovery by preserving tissue elasticity in the peri-infarct borders that sustains contractile efficiency for functional recovery in an actively remodeling infarcted myocardium.
- Published
- 2010
- Full Text
- View/download PDF
13. Myoblast transplantation for cardiac repair: from automyoblast to allomyoblast transplantation.
- Author
-
Guo C, Haider HKh, Wang C, Tan RS, Shim WS, Wong P, and Sim EK
- Subjects
- Animals, Disease Models, Animal, Female, Graft Rejection, Graft Survival, Immunohistochemistry, Immunosuppressive Agents pharmacology, Male, Myoblasts, Skeletal immunology, Myocardial Infarction pathology, Probability, Random Allocation, Rats, Rats, Wistar, Regeneration physiology, Risk Factors, Sensitivity and Specificity, Transplantation, Autologous, Transplantation, Homologous, Ventricular Remodeling physiology, Cell Transplantation methods, Cyclosporins pharmacology, Myoblasts, Skeletal transplantation, Myocardial Infarction surgery
- Abstract
Background: We sought to compare host immune cell kinetics, survival profile of donor skeletal myoblasts, and skeletal myoblast graft efficacy after autologous and allogeneic skeletal myoblast transplantation into a rat model of myocardial infarction., Methods: One week after myocardial infarction, 128 animals were divided into four groups: group 1 (n = 24, receiving medium only), group 2 (n = 24, receiving medium and cyclosporine), group 3 (n = 40, autologous skeletal myoblast transplantation), and group 4 (n = 40, allogeneic skeletal myoblast transplantation with cyclosporine treatment). Rats were euthanized 10 minutes, 1 day, and 4, 7, and 28 days later. Host immune cell kinetics were assessed by immunohistochemical studies for macrophages, and CD4+ and CD8+ lymphocytes. Donor skeletal myoblast survival was confirmed by tracking prelabeled signals, and quantified by beta-gal assay. Heart function was evaluated by echocardiography., Results: A transient immune cell infiltration was demonstrated in group 3, with macrophage infiltration on day 1 and day 4, CD8+ cell infiltration on day 4 and day 7, and CD4+ cell infiltration on day 4. In group 4, immunocyte infiltration was slightly more severe than that in group 3. Automyoblasts and allomyoblasts showed no significant difference of survival from day 1 to day 7 (p > 0.10); however, on day 28, automyoblasts showed better survival than allomyoblasts (p < 0.05). Transplantation of allomyoblasts increased systolic heart function and limited heart dilation after myocardial injury to a similar degree as automyoblasts (p > 0.10)., Conclusions: The use of allomyoblasts is feasible and effective for cardiac repair with immunosuppressive treatment as compared with automyoblasts.
- Published
- 2008
- Full Text
- View/download PDF
14. Ex vivo differentiation of human adult bone marrow stem cells into cardiomyocyte-like cells.
- Author
-
Shim WS, Jiang S, Wong P, Tan J, Chua YL, Tan YS, Sin YK, Lim CH, Chua T, Teh M, Liu TC, and Sim E
- Subjects
- Adipocytes metabolism, Aged, Ascorbic Acid pharmacology, Cell Differentiation, Cell Lineage, Cell Proliferation, Cells, Cultured, Coculture Techniques, Culture Media pharmacology, DNA Primers chemistry, Dexamethasone pharmacology, Female, Humans, Male, Mesenchymal Stem Cells cytology, Mesoderm cytology, Middle Aged, Muscle, Skeletal cytology, Myocardium metabolism, Phenotype, RNA chemistry, Reverse Transcriptase Polymerase Chain Reaction, Stem Cells cytology, Time Factors, Bone Marrow Cells cytology, Cell Culture Techniques methods, Myocytes, Cardiac metabolism
- Abstract
Bone marrow mesenchymal stem cells have been shown to transdifferentiate into cardiomyocytes after 5-azacytidine treatment or co-culturing with rodent cardiomyocytes. We investigate if adult human bone marrow stem cells can be differentiated ex vivo into cardiomyocyte-like cells (CLCs) independent of cytotoxic agents or co-culturing technique. Sternal bone marrow was collected from 16 patients undergoing coronary artery bypass surgery. Mesenchymal stem cells were differentiated in a cardiomyogenic differentiation medium containing insulin, dexamethasone, and ascorbic acid. Differentiation towards CLCs was determined by induced expression of cardiomyocyte-specific proteins. Differentiated CLCs expressed multiple structural and contractile proteins that are associated with cardiomyocytes. Thin filament associated myofibrillar proteins were detected early in the cells, with cardiac troponin I, sarcomeric tropomyosin, and cardiac titin among the first expressed. Some CLCs were found to develop into a nascent cardiomyocyte phenotype with cross-striated myofibrils characterized by alpha-actinin-positive Z bands after 4-5 passages in differentiated culture. These lineage-defined CLCs may be potentially useful for repairing damaged myocardium.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.