1
|
Fawzy El-Sayed KM, Cosgarea R, Sculean A, Doerfer C. Can vitamins improve periodontal wound healing/regeneration? Periodontol 2000 2024; 94:539-602. [PMID: 37592831 DOI: 10.1111/prd.12513] [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: 05/12/2023] [Revised: 07/13/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Periodontitis is a complex inflammatory disorder of the tooth supporting structures, associated with microbial dysbiosis, and linked to a number if systemic conditions. Untreated it can result in an irreversible damage to the periodontal structures and eventually teeth loss. Regeneration of the lost periodontium requires an orchestration of a number of biological events on cellular and molecular level. In this context, a set of vitamins have been advocated, relying their beneficial physiological effects, to endorse the biological regenerative events of the periodontium on cellular and molecular levels. The aim of the present article is to elaborate on the question whether or not vitamins improve wound healing/regeneration, summarizing the current evidence from in vitro, animal and clinical studies, thereby shedding light on the knowledge gap in this field and highlighting future research needs. Although the present review demonstrates the current heterogeneity in the available evidence and knowledge gaps, findings suggest that vitamins, especially A, B, E, and CoQ10, as well as vitamin combinations, could exert positive attributes on the periodontal outcomes in adjunct to surgical or nonsurgical periodontal therapy.
Collapse
Affiliation(s)
- Karim M Fawzy El-Sayed
- Oral Medicine and Periodontology Department, Faculty of Oral and Dental Medicine, Cairo University, Giza, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| | - Raluca Cosgarea
- Department of Periodontology, Operative and Preventive Dentistry, University of Bonn, Bonn, Germany
- Department of Periodontology and Peri-implant Diseases, Philips University Marburg, Marburg, Germany
- Clinic for Prosthetic Dentistry, University Iuliu-Hatieganu, Cluj-Napoca, Romania
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Christof Doerfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany
| |
Collapse
|
2
|
Diederich A, Fründ HJ, Trojanowicz B, Navarrete Santos A, Nguyen AD, Hoang-Vu C, Gernhardt CR. Influence of Ascorbic Acid as a Growth and Differentiation Factor on Dental Stem Cells Used in Regenerative Endodontic Therapies. J Clin Med 2023; 12:jcm12031196. [PMID: 36769844 PMCID: PMC9917775 DOI: 10.3390/jcm12031196] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/23/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Vitamin C is one of the major extracellular nonenzymatic antioxidants involved in the biosynthesis of collagen. It promotes the growth of fibroblasts, wound healing processes, and enhances the survival and differentiation of osteoblasts. The potential effects of ascorbic acid on human dental pulp cells (DPC) and the cells of the apical papilla (CAP) used in actual regenerative endodontic procedures remain largely unknown. In this study, we investigated the possible employment of ascorbic acid in the differentiation and regenerative therapies of DPC and CAP. METHODS Nine extracted human wisdom teeth were selected for this study. Subpopulations of stem cells within DPC and CAP were sorted with the mesenchymal stem cell marker STRO-1, followed by treatments with different concentrations (0 mM, 0.1 mM, 0.5 mM, and 1.0 mM) of ascorbic acid (AA), RT-PCR, and Western blot analysis. RESULTS FACS analysis revealed the presence of cell subpopulations characterized by a strong expression of mesenchymal stem cell marker STRO-1 and dental stem cell markers CD105, CD44, CD146, CD90, and CD29. Treatment of the cells with defined amounts of AA revealed a markedly increased expression of proliferation marker Ki-67, especially in the concentration range between 0.1 mM and 0.5 mM. Further investigations demonstrated that treatment with AA led to significantly increased expression of common stem cell markers OCT4, Nanog, and Sox2. The most potent proliferative and expressional effects of AA were observed in the concentration of 0.1 mM. CONCLUSIONS AA might be a novel and potent growth promoter of human dental cells. Increasing the properties of human dental pulp cells and the cells of the apical papilla using AA could be a useful factor for further clinical developments of regenerative endodontic procedures.
Collapse
Affiliation(s)
- Antje Diederich
- University Outpatient Clinic for Conservative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06112 Halle, Germany
- Correspondence: ; Tel.: +49-345-557-3737
| | - Hanna Juliane Fründ
- Department of Visceral, Vascular and Endocrine Surgery, University Medical Center Halle, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Bogusz Trojanowicz
- Department of Visceral, Vascular and Endocrine Surgery, University Medical Center Halle, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | | | - Anh Duc Nguyen
- University Outpatient Clinic for Conservative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06112 Halle, Germany
- Private Dental Practice, Dr. Juliane Gernhardt, 06120 Halle, Germany
| | - Cuong Hoang-Vu
- Department of Visceral, Vascular and Endocrine Surgery, University Medical Center Halle, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Christian Ralf Gernhardt
- University Outpatient Clinic for Conservative Dentistry and Periodontology, Martin-Luther-University Halle-Wittenberg, 06112 Halle, Germany
| |
Collapse
|
3
|
Lu V, Roy IJ, Teitell MA. Nutrients in the fate of pluripotent stem cells. Cell Metab 2021; 33:2108-2121. [PMID: 34644538 PMCID: PMC8568661 DOI: 10.1016/j.cmet.2021.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/07/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022]
Abstract
Pluripotent stem cells model certain features of early mammalian development ex vivo. Medium-supplied nutrients can influence self-renewal, lineage specification, and earliest differentiation of pluripotent stem cells. However, which specific nutrients support these distinct outcomes, and their mechanisms of action, remain under active investigation. Here, we evaluate the available data on nutrients and their metabolic conversion that influence pluripotent stem cell fates. We also discuss key questions open for investigation in this rapidly expanding area of increasing fundamental and practical importance.
Collapse
Affiliation(s)
- Vivian Lu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Irena J Roy
- Developmental and Stem Cell Biology, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael A Teitell
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
4
|
Cardiac Differentiation of Mesenchymal Stem Cells: Impact of Biological and Chemical Inducers. Stem Cell Rev Rep 2021; 17:1343-1361. [PMID: 33864233 DOI: 10.1007/s12015-021-10165-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular disorders (CVDs) are the leading cause of global death, widely occurs due to irreparable loss of the functional cardiomyocytes. Stem cell-based therapeutic approaches, particularly the use of Mesenchymal Stem Cells (MSCs) is an emerging strategy to regenerate myocardium and thereby improving the cardiac function after myocardial infarction (MI). Most of the current approaches often employ the use of various biological and chemical factors as cues to trigger and modulate the differentiation of MSCs into the cardiac lineage. However, the recent advanced methods of using specific epigenetic modifiers and exosomes to manipulate the epigenome and molecular pathways of MSCs to modify the cardiac gene expression yield better profiled cardiomyocyte like cells in vitro. Hitherto, the role of cardiac specific inducers triggering cardiac differentiation at the cellular and molecular level is not well understood. Therefore, the current review highlights the impact and recent trends in employing biological and chemical inducers on cardiac differentiation of MSCs. Thereby, deciphering the interactions between the cellular microenvironment and the cardiac inducers will help us to understand cardiomyogenesis of MSCs. Additionally, the review also provides an insight on skeptical roles of the cell free biological factors and extracellular scaffold assisted mode for manipulation of native and transplanted stem cells towards translational cardiac research.
Collapse
|
5
|
Elbehwashy MT, Hosny MM, Elfana A, Nawar A, Fawzy El-Sayed K. Clinical and radiographic effects of ascorbic acid-augmented platelet-rich fibrin versus platelet-rich fibrin alone in intra-osseous defects of stage-III periodontitis patients: a randomized controlled clinical trial. Clin Oral Investig 2021; 25:6309-6319. [PMID: 33842996 PMCID: PMC8531044 DOI: 10.1007/s00784-021-03929-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
Aim To assess platelet-rich fibrin (PRF) with ascorbic acid (AA) versus PRF in intra-osseous defects of stage-III periodontitis patients. Methodology Twenty stage-III/grade C periodontitis patients, with ≥ 3 mm intra-osseous defects, were randomized into test (open flap debridement (OFD)+AA/PRF; n = 10) and control (OFD+PRF; n = 10). Clinical attachment level (CAL; primary outcome), probing pocket depth (PPD), gingival recession depth (RD), full-mouth bleeding scores (FMBS), full-mouth plaque scores (FMPS), radiographic linear defect depth (RLDD) and radiographic defect bone density (RDBD) (secondary-outcomes) were examined at baseline, 3 and 6 months post-surgically. Results OFD+AA/PRF and OFD+PRF demonstrated significant intragroup CAL gain and PPD reduction at 3 and 6 months (p < 0.001). OFD+AA/PRF and OFD+PRF showed no differences regarding FMBS or FMPS (p > 0.05). OFD+AA/PRF demonstrated significant RD reduction of 0.90 ± 0.50 mm and 0.80 ± 0.71 mm at 3 and 6 months, while OFD+PRF showed RD reduction of 0.10 ± 0.77 mm at 3 months, with an RD-increase of 0.20 ± 0.82 mm at 6 months (p < 0.05). OFD+AA/PRF and OFD+PRF demonstrated significant RLDD reduction (2.29 ± 0.61 mm and 1.63 ± 0.46 mm; p < 0.05) and RDBD-increase (14.61 ± 5.39% and 12.58 ± 5.03%; p > 0.05). Stepwise linear regression analysis showed that baseline RLDD and FMBS at 6 months were significant predictors of CAL reduction (p < 0.001). Conclusions OFD+PRF with/without AA significantly improved periodontal parameters 6 months post-surgically. Augmenting PRF with AA additionally enhanced gingival tissue gain and radiographic defect fill. Clinical relevance PRF, with or without AA, could significantly improve periodontal parameters. Supplementing PRF with AA could additionally augment radiographic linear defect fill and reduce gingival recession depth. Supplementary Information The online version contains supplementary material available at 10.1007/s00784-021-03929-1.
Collapse
Affiliation(s)
- Mohamed Talaat Elbehwashy
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Al Saraya Str. 11, Manial, Cairo, Egypt
| | - Manal Mohamed Hosny
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Al Saraya Str. 11, Manial, Cairo, Egypt
| | - Ahmed Elfana
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Al Saraya Str. 11, Manial, Cairo, Egypt
| | - Alaa Nawar
- Oral and Maxillofacial Radiology Department, Faculty of Dentistry, Cairo University, Giza, Egypt
| | - Karim Fawzy El-Sayed
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Al Saraya Str. 11, Manial, Cairo, Egypt.
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, Kiel, Germany.
| |
Collapse
|
6
|
Bai R, Chang Y, Saleem A, Wu F, Tian L, Zhang S, Li Y, Ma S, Dong T, Guo T, Jiang Y, You Y, Lu WJ, Jiang HF, Lan F. Ascorbic acid can promote the generation and expansion of neuroepithelial-like stem cells derived from hiPS/ES cells under chemically defined conditions through promoting collagen synthesis. Stem Cell Res Ther 2021; 12:48. [PMID: 33422132 PMCID: PMC7796386 DOI: 10.1186/s13287-020-02115-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/22/2020] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Spinal cord injury (SCI) is a neurological, medically incurable disorder. Human pluripotent stem cells (hPSCs) have the potential to generate neural stem/progenitor cells (NS/PCs), which hold promise in the treatment of SCI by transplantation. In our study, we aimed to establish a chemically defined culture system using serum-free medium and ascorbic acid (AA) to generate and expand long-term self-renewing neuroepithelial-like stem cells (lt-NES cells) differentiated from hPSCs effectively and stably. METHODS We induced human embryonic stem cells (hESCs)/induced PSCs (iPSCs) to neurospheres using a newly established in vitro induction system. Moreover, lt-NES cells were derived from hESC/iPSC-neurospheres using two induction systems, i.e., conventional N2 medium with gelatin-coated plates (coated) and N2+AA medium without pre-coated plates (AA), and were characterized by reverse transcription polymerase chain reaction (RT-PCR) analysis and immunocytochemistry staining. Subsequently, lt-NES cells were induced to neurons. A microelectrode array (MEA) recording system was used to evaluate the functionality of the neurons differentiated from lt-NES cells. Finally, the mechanism underlying the induction of lt-NES cells by AA was explored through RNA-seq and the use of inhibitors. RESULTS HESCs/iPSCs were efficiently induced to neurospheres using a newly established induction system in vitro. lt-NES cells derived from hESC/iPSC-neurospheres using the two induction systems (coated vs. AA) both expressed the neural pluripotency-associated genes PAX6, NESTIN, SOX1, and SOX2. After long-term cultivation, we found that they both exhibited long-term expansion for more than a dozen generations while maintaining neuropluripotency. Moreover, the lt-NES cells retained the ability to differentiate into general functional neurons that express β-tubulin at high levels. We also demonstrated that AA promotes the generation and long-term expansion of lt-NES cells by promoting collagen synthesis via the MEK-ERK1/2 pathway. CONCLUSIONS This new chemically defined culture system was stable and effective regarding the generation and culture of lt-NES cells induced from hESCs/iPSCs using serum-free medium combined with AA. The lt-NES cells induced under this culture system maintained their long-term expansion and neural pluripotency, with the potential to differentiate into functional neurons.
Collapse
Affiliation(s)
- Rui Bai
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yun Chang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Amina Saleem
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Fujian Wu
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Lei Tian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Siyao Zhang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Ya'nan Li
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Shuhong Ma
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Tao Dong
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Tianwei Guo
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Youxu Jiang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yi You
- Center for Clinical Translation and Innovation, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.,Shenzhen Bay Laboratory, Shenzhen, 518055, China
| | - Wen-Jing Lu
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Hong Feng Jiang
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China. .,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China. .,Beijing Anzhen Hospital, Research Institute Building, Room 323, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.
| | - Feng Lan
- Beijing Laboratory for Cardiovascular Precision Medicine, MOE Key Laboratory of Medical Engineering for Cardiovascular Diseases, MOE Key Laboratory of Remodeling-Related Cardiovascular Disease, Beijing Collaborative Innovation Center for Cardiovascular Disorders, Anzhen Hospital, Capital Medical University, Beijing, 100029, China. .,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China. .,State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China. .,Beijing Anzhen Hospital, Research Institute Building, Room 319, 2 Anzhen Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
7
|
The Effect of Angiotensin II, Retinoic Acid, EGCG, and Vitamin C on the Cardiomyogenic Differentiation Induction of Human Amniotic Fluid-Derived Mesenchymal Stem Cells. Int J Mol Sci 2020; 21:ijms21228752. [PMID: 33228183 PMCID: PMC7699548 DOI: 10.3390/ijms21228752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 01/07/2023] Open
Abstract
Human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) may be potentially applied in cell therapy or regenerative medicine as a new alternative source of stem cells. They could be particularly valuable in restoring cardiac tissue after myocardial infarction or other cardiovascular diseases. We investigated the potential of biologically active compounds, namely, angiotensin II, retinoic acid (RA), epigallocatechin-3-gallate (EGCG), vitamin C alone, and the combinations of RA, EGCG, and vitamin C with angiotensin II to induce cardiomyogenic differentiation of AF-MSCs. We observed that the upregulated expression of cardiac gene markers (NKX2-5, MYH6, TNNT2, and DES) and cardiac ion channel genes (sodium, calcium, the potassium) also the increased levels of Connexin 43 and Nkx2.5 proteins. Extracellular flux analysis, applied for the first time on AF-MSCs induced with biologically active compounds, revealed the switch in AF-MSCS energetic phenotype and enhanced utilization of oxidative phosphorylation for energy production. Moreover, we demonstrated changes in epigenetic marks associated with transcriptionally active (H3K4me3, H3K9ac, and H4hyperAc) or repressed (H3K27me3) chromatin. All in all, we demonstrated that explored biomolecules were able to induce alterations in AF-MSCs at the phenotypic, genetic, protein, metabolic, and epigenetic levels, leading to the formation of cardiomyocyte progenitors that may become functional heart cells in vitro or in vivo.
Collapse
|
8
|
Wahyuningsih KA, Karina K, Rosadi I, Rosliana I, Subroto WR. Effect of ascorbic acid on morphology of post-thawed human adipose-derived stem cells. Stem Cell Investig 2020; 7:16. [PMID: 33110914 DOI: 10.21037/sci-2020-011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022]
Abstract
Background Ascorbic acid-2-phosphate has been reported to play a role in cell division and to suppress aging of cell. However, post-thawed cell morphology on various concentration of ascorbic acid is still unclear. In this study, we aimed to observe the morphology of post-thawed adipose-derived stem cells (ADSCs) in medium containing L-ascorbic acid-2-phosphate (LAA2P) (50 and 100 µg/mL). Methods The cells were isolated from adipose tissue. Isolated cells then cultured and cryopreserved in liquid nitrogen. We detected mRNA expression of type 1 collagen on day 5. Cell seeded in T25 flask using basal medium [Dulbecco's modified Eagle's medium (DMEM) only] as a control group, DMEM with 10% fetal bovine serum (FBS) and antibiotics as DMFA group, while DMFA with ascorbic acid (50 and 100 µg/mL) as ascorbic acid treatment group. Results The results showed that the cells cultured in DMEM only attached until 96 hours of observation while serum groups with or without ascorbic acid supplementation showed the proliferation until 240 hours of observation. The highest spread size of cell was in a serum group without ascorbic acid supplementation and the highest yield of cells showed in a group with 50 µg/mL of ascorbic acid supplementation. Reduced mRNA expression of type 1 collagen which related to aging was showed in cells cultured without ascorbic acid supplementation. Conclusions These results showed that ascorbic acid increased the cell division and suppressed the aging processes indicated by normal spread cell in size compared to cell cultured in DMFA without ascorbic acid supplementation.
Collapse
Affiliation(s)
- Komang Ardi Wahyuningsih
- Department of Histology, Universitas Katolik Indonesia Atma Jaya, Jakarta, Indonesia.,Klinik Hayandra, Yayasan Hayandra Peduli, Jakarta, Indonesia.,Hayandra Lab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | - Karina Karina
- Klinik Hayandra, Yayasan Hayandra Peduli, Jakarta, Indonesia.,Hayandra Lab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | - Imam Rosadi
- Hayandra Lab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | - Iis Rosliana
- Hayandra Lab, Yayasan Hayandra Peduli, Jakarta, Indonesia
| | | |
Collapse
|
9
|
The Influence of Cell Culture Density on the Cytotoxicity of Adipose-Derived Stem Cells Induced by L-Ascorbic Acid-2-Phosphate. Sci Rep 2020; 10:104. [PMID: 31919399 PMCID: PMC6952413 DOI: 10.1038/s41598-019-56875-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
Ascorbic acid-2-phosphate (A2-P) is an oxidation-resistant derivative of ascorbic acid that has been widely employed in culturing adipose-derived stem cells (ASCs) for faster expansion and cell sheet formation. While high dose ascorbic acid is known to induce cellular apoptosis via metabolic stress and genotoxic effects, potential cytotoxic effects of A2-P at high concentrations has not been explored. In this study, the relationship between ASC seeding density and A2-P-induced cytotoxicity was investigated. Spheroid-derived ASCs with smaller cellular dimensions were generated to investigate the effect of cell-cell contact on the resistance to A2-P-induced cytotoxicity. Decreased viability of ASC, fibroblast, and spheroid-derived ASC was noted at higher A2-P concentration, and it could be reverted with high seeding density. Compared to control ASCs, spheroid-derived ASCs seeded at the same density exhibited decreased viability in the A2-P-supplemented medium. The expression of antioxidant enzymes (catalase, SOD1, and SOD2) was enhanced in ASCs at higher seeding densities. However, their enhanced expression in spheroid-derived ASCs was less evident. Furthermore, we found that co-administration of catalase or N-acetylcysteine nullified the observed cytotoxicity. Collectively, A2-P can induce ASC cytotoxicity at higher concentrations, which can be prevented by seeding ASCs at high density or co-administration of another antioxidant.
Collapse
|
10
|
Hamad S, Derichsweiler D, Papadopoulos S, Nguemo F, Šarić T, Sachinidis A, Brockmeier K, Hescheler J, Boukens BJ, Pfannkuche K. Generation of human induced pluripotent stem cell-derived cardiomyocytes in 2D monolayer and scalable 3D suspension bioreactor cultures with reduced batch-to-batch variations. Theranostics 2019; 9:7222-7238. [PMID: 31695764 PMCID: PMC6831300 DOI: 10.7150/thno.32058] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are promising candidates to treat myocardial infarction and other cardiac diseases. Such treatments require pure cardiomyocytes (CMs) in large quantities. Methods: In the present study we describe an improved protocol for production of hiPSC-CMs in which hiPSCs are first converted into mesodermal cells by stimulation of wingless (Wnt) signaling using CHIR99021, which are then further differentiated into CM progenitors by simultaneous inhibition of porcupine and tankyrase pathways using IWP2 and XAV939 under continuous supplementation of ascorbate during the entire differentiation procedure. Results: The protocol resulted in reproducible generation of >90% cardiac troponin T (TNNT2)-positive cells containing highly organized sarcomeres. In 2D monolayer cultures CM yields amounted to 0.5 million cells per cm2 growth area, and on average 72 million cells per 100 mL bioreactor suspension culture without continuous perfusion. The differentiation efficiency was hardly affected by the initial seeding density of undifferentiated hiPSCs. Furthermore, batch-to-batch variations were reduced by combinatorial use of ascorbate, IWP2, and XAV939. Conclusion: Combined inhibition of porcupine and tankyrase sub-pathways of Wnt signaling and continuous ascorbate supplementation, enable robust and efficient production of hiPSC-CMs.
Collapse
Affiliation(s)
- Sarkawt Hamad
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Biology Department, Faculty of Science, Soran University, Soran, Kurdistan region-Iraq
| | - Daniel Derichsweiler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Symeon Papadopoulos
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Center for Physiology and Pathophysiology, Institute for Vegetative Physiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Agapios Sachinidis
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Konrad Brockmeier
- Department of Pediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Bastiaan J Boukens
- Department of Medical Biology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Kurt Pfannkuche
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty, Cologne, Germany
- Department of Pediatric Cardiology, University Clinics of Cologne, Cologne, Germany
| |
Collapse
|
11
|
Markmee R, Aungsuchawan S, Pothacharoen P, Tancharoen W, Narakornsak S, Laowanitwattana T, Bumroongkit K, Puaninta C, Pangjaidee N. Effect of ascorbic acid on differentiation of human amniotic fluid mesenchymal stem cells into cardiomyocyte-like cells. Heliyon 2019; 5:e02018. [PMID: 31360783 PMCID: PMC6639694 DOI: 10.1016/j.heliyon.2019.e02018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 04/18/2019] [Accepted: 06/27/2019] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to evaluate the efficiency of ascorbic acid (AA) on cell viability, cytotoxicity and the effects on cardiomyogenic differentiation of the human amniotic fluid mesenchymal stem cells (hAF-MSCs). The results of methylthiazole tetrazolium (MTT) assay and cell apoptosis assay indicated that after 24, 48 and 72 h of treatment, AA had no effect on cells viability and cytotoxicity. After treating the hAF-MSCs with 5-azacytidine (5-aza) and a combination of AA and 5-aza, the alamar blue cells proliferation assay showed the normal growth characteristic similar to control group. Especially, the morphological changes were observed between day 0 and day 21, and it was revealed that the hAF-MSCs exhibited myotube-like morphology after 7 days of cell culturing. Moreover, the treatment with a combination of AA and 5-aza was able to up-regulate the cardiomyogenic specific gene levels, which are known to play an important role in cardiomyogenesis. This was specifically notable with the results of immunofluorescence and immunoenzymatic staining in the AA combined with 5-aza treatment group, the highest expression of cardiomyogenic specific proteins was revealed including for GATA4, cTnT, Cx43 and Nkx2.5. It could be concluded that AA may be a good alternative cardiomyogenic inducing factor for hAF-MSCs and may open new insights into future biomedical applications for a clinically treatment.
Collapse
Affiliation(s)
- Runchana Markmee
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Sirinda Aungsuchawan
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Peraphan Pothacharoen
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Waleephan Tancharoen
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Suteera Narakornsak
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Kanokkan Bumroongkit
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaniporn Puaninta
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nathaporn Pangjaidee
- Department of Anatomy, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
12
|
Devireddy LR, Myers M, Screven R, Liu Z, Boxer L. A serum-free medium formulation efficiently supports isolation and propagation of canine adipose-derived mesenchymal stem/stromal cells. PLoS One 2019; 14:e0210250. [PMID: 30811421 PMCID: PMC6392232 DOI: 10.1371/journal.pone.0210250] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/19/2018] [Indexed: 12/19/2022] Open
Abstract
Medium containing Fetal Bovine Serum (FBS) provides a supportive environment for isolation and expansion of mesenchymal stromal/stem cells (MSCs); however, the inherent variability of FBS may contribute to inconsistencies in cell growth and yield between batches of stem cell products. For this reason, we set out to develop a serum-free medium capable of supporting the in vitro expansion of MSCs. First a naïve serum-free medium was formulated by Sato's approach. Once it was established that the naïve serum-free medium supported the expansion of canine adipose-derived MSCs (Ad-MSCs), the serum-free medium was optimized by addition of growth factors. Combinations of growth factors were chosen and compared by their effect on cell proliferation and colony formation. Growth characteristics of canine adipose-derived MSCs cultured in the serum-free medium were comparable to those cultured in standard FBS containing medium. In addition, cell surface marker expression and differentiation potential of serum-free and FBS-based cultures were also comparable. However, a commercial serum-free medium developed for human MSC culture did not support growth of canine Ad-MSCs. In summary, canine Ad-MSCs isolated and cultured in serum-free medium retained the basic characteristics of MSCs cultured in FBS containing medium.
Collapse
Affiliation(s)
- Laxminarayana R. Devireddy
- Division of Applied Veterinary Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland, United States of America
| | - Michael Myers
- Division of Applied Veterinary Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland, United States of America
| | - Rudell Screven
- Division of Applied Veterinary Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland, United States of America
| | - Zhuoming Liu
- Division of Applied Veterinary Research, Center for Veterinary Medicine, US Food and Drug Administration, Laurel, Maryland, United States of America
| | - Lynne Boxer
- Office of New Animal Drug Evaluation, Center for Veterinary Medicine, US Food and Drug Administration, Rockville, Maryland, United States of America
| |
Collapse
|
13
|
Liu W, Liu H, Wang Y, Zhang L, Wang C, Li H. Ascorbic acid induces cardiac differentiation of white adipose tissue-derived stem cells. Mol Cell Biochem 2018; 450:65-73. [PMID: 29808464 DOI: 10.1007/s11010-018-3373-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/23/2018] [Indexed: 12/27/2022]
Abstract
White adipose tissue (WAT) is the bulk of fatty tissues in humans. Enhancing the potential of WAT-derived stem cells (WATDCs) to generate cardiomyocytes may help supply sufficient number of therapeutically potent cells for heart repair in vivo. Therefore, we investigated whether ascorbic acid (AA) could facilitate the cardiac differentiation of WATDCs and the underlying mechanisms. Our results indicated that AA dose-dependently stimulates the cardiac differentiation of WATDCs, which is supported by the up-regulated expression of cardiac markers and the appearance of myotube-like cell morphologies. Time-course study showed that the front phase (0-4 days) is crucial for the action of AA on cardiac differentiation, which hints that AA may take effect through enhancing the proliferation of cardiac progenitor cells. EdU assay ascertained AA indeed promotes cell growth dose-dependently in the front phase. Further investigation indicated that AA induces the phosphorylation of MEK and ERK, and the synthesis of collagen I (Col I). Interference of MEK/ERK activity or Col I synthesis blocks the cardiomyogenic activity of AA in WATDCs. These findings demonstrated that AA facilitates WATDC cardiogenesis via promoting the proliferation of cardiac progenitor cells through MEK/ERK signaling and collagen synthesis.
Collapse
Affiliation(s)
- Wenhui Liu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Huan Liu
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Yinghui Wang
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Linlin Zhang
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Chunhui Wang
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China
| | - Haifang Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, 271018, China.
| |
Collapse
|
14
|
Fujisawa K, Hara K, Takami T, Okada S, Matsumoto T, Yamamoto N, Sakaida I. Evaluation of the effects of ascorbic acid on metabolism of human mesenchymal stem cells. Stem Cell Res Ther 2018; 9:93. [PMID: 29625581 PMCID: PMC5889584 DOI: 10.1186/s13287-018-0825-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 02/05/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent cells holding much promise for applications in regenerative medicine. However, with problems such as aging, increases in heteroploid cells, genomic instability, and reduced maintenance of stemness, more stable culturing methods and the production of MSCs with an improved therapeutic effect are desired. Ascorbic acid (AsA), which is a cofactor for a variety of enzymes and has an antioxidant effect, cannot be synthesized by certain animals, including humans. Nevertheless, little attention has been paid to AsA when culturing MSCs. METHODS We analyzed the effect of adding AsA to the culture medium on the proliferation and metabolism of human MSCs by serial analysis of gene expression and metabolome analysis. RESULTS We found that AsA promotes MSC proliferation, and is particularly useful when expanding MSCs isolated from bone marrow. Serial analysis of gene expression and metabolome analysis suggested that, due to HIF1α accumulation caused by decreased activity of the enzymes that use AsA as a coenzyme in cultures without AsA, genes downstream of HIF1α are expressed and there is a conversion to a hypoxia-mimetic metabolism. AsA promotes HIF1α breakdown and activates mitochondria, affecting cell proliferation and metabolism. Comprehensive evaluation of the effects of AsA on various metabolic products in MSCs revealed that AsA increases HIF1α hydroxylase activity, suppressing HIF1a transcription and leading to mitochondrial activation. CONCLUSIONS Adding AsA during MSC expansion leads to more efficient preparation of cells. These are expected to be important findings for the future application of MSCs in regenerative medicine.
Collapse
Affiliation(s)
- Koichi Fujisawa
- Center for Regenerative Medicine, Yamaguchi University School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan.,Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Kazusa Hara
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan.
| | - Sae Okada
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Toshihiko Matsumoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Naoki Yamamoto
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| | - Isao Sakaida
- Center for Regenerative Medicine, Yamaguchi University School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan.,Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Minami Kogushi 1-1-1, Ube, Yamaguchi, 755-8505, Japan
| |
Collapse
|
15
|
Dahlmann J, Awad G, Dolny C, Weinert S, Richter K, Fischer KD, Munsch T, Leßmann V, Volleth M, Zenker M, Chen Y, Merkl C, Schnieke A, Baraki H, Kutschka I, Kensah G. Generation of functional cardiomyocytes from rat embryonic and induced pluripotent stem cells using feeder-free expansion and differentiation in suspension culture. PLoS One 2018. [PMID: 29513687 PMCID: PMC5841662 DOI: 10.1371/journal.pone.0192652] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The possibility to generate cardiomyocytes from pluripotent stem cells in vitro has enormous significance for basic research, disease modeling, drug development and heart repair. The concept of heart muscle reconstruction has been studied and optimized in the rat model using rat primary cardiovascular cells or xenogeneic pluripotent stem cell derived-cardiomyocytes for years. However, the lack of rat pluripotent stem cells (rPSCs) and their cardiovascular derivatives prevented the establishment of an authentic clinically relevant syngeneic or allogeneic rat heart regeneration model. In this study, we comparatively explored the potential of recently available rat embryonic stem cells (rESCs) and induced pluripotent stem cells (riPSCs) as a source for cardiomyocytes (CMs). We developed feeder cell-free culture conditions facilitating the expansion of undifferentiated rPSCs and initiated cardiac differentiation by embryoid body (EB)-formation in agarose microwell arrays, which substituted the robust but labor-intensive hanging drop (HD) method. Ascorbic acid was identified as an efficient enhancer of cardiac differentiation in both rPSC types by significantly increasing the number of beating EBs (3.6 ± 1.6-fold for rESCs and 17.6 ± 3.2-fold for riPSCs). These optimizations resulted in a differentiation efficiency of up to 20% cTnTpos rPSC-derived CMs. CMs showed spontaneous contractions, expressed cardiac markers and had typical morphological features. Electrophysiology of riPSC-CMs revealed different cardiac subtypes and physiological responses to cardio-active drugs. In conclusion, we describe rPSCs as a robust source of CMs, which is a prerequisite for detailed preclinical studies of myocardial reconstruction in a physiologically and immunologically relevant small animal model.
Collapse
Affiliation(s)
- Julia Dahlmann
- Clinic of Cardiothoracic Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - George Awad
- Clinic of Cardiothoracic Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Carsten Dolny
- Clinic of Cardiothoracic Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Sönke Weinert
- Clinic of Cardiology and Angiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Karin Richter
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Thomas Munsch
- Institute of Physiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Volkmar Leßmann
- Institute of Physiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Marianne Volleth
- Institute of Human Genetics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Martin Zenker
- Institute of Human Genetics, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Yaoyao Chen
- Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Claudia Merkl
- Chair of Livestock Biotechnology, Technical University Munich, Freising-Weihenstephan, Germany
| | - Angelika Schnieke
- Chair of Livestock Biotechnology, Technical University Munich, Freising-Weihenstephan, Germany
| | - Hassina Baraki
- Clinic of Cardiothoracic Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ingo Kutschka
- Clinic of Cardiothoracic Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - George Kensah
- Clinic of Cardiothoracic Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
- * E-mail:
| |
Collapse
|
16
|
Guo X, Bai Y, Zhang L, Zhang B, Zagidullin N, Carvalho K, Du Z, Cai B. Cardiomyocyte differentiation of mesenchymal stem cells from bone marrow: new regulators and its implications. Stem Cell Res Ther 2018; 9:44. [PMID: 29482607 PMCID: PMC5828435 DOI: 10.1186/s13287-018-0773-9] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In the past years, cardiac mortality has decreased, but cardiac diseases are still responsible for millions of deaths every year worldwide. Bone-marrow mesenchymal stem cells (BMSCs) transplantation may be a promising therapeutic strategy because of its capacity to differentiate into cardiac cells. Current research indicates that chemical substances, microRNAs, and cytokines have biological functions that regulate the cardiomyocytes differentiation of BMSCs. In this review, we chiefly summarize the regulatory factors that induce BMSCs to differentiate into cardiomyocytes.
Collapse
Affiliation(s)
- Xiaofei Guo
- Department of Pharmacy, the Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Yan Bai
- Department of Pharmacy, the Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Li Zhang
- Department of Pharmacy, the Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Bo Zhang
- Department of Pharmacy, the Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Naufal Zagidullin
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Russia
| | - Katherine Carvalho
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pequeno Príncipe Faculty, Pelé Pequeno Príncipe Institute, Curitiba, Brazil
| | - Zhimin Du
- Department of Pharmacy, the Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, Heilongjiang Province, 150081, People's Republic of China
| | - Benzhi Cai
- Department of Pharmacy, the Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Harbin, Heilongjiang Province, 150081, People's Republic of China.
| |
Collapse
|
17
|
Ascorbic acid promotes 3T3-L1 cells adipogenesis by attenuating ERK signaling to upregulate the collagen VI. Nutr Metab (Lond) 2017; 14:79. [PMID: 29299041 PMCID: PMC5745638 DOI: 10.1186/s12986-017-0234-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 12/07/2017] [Indexed: 12/11/2022] Open
Abstract
Background Type VI collagen is supposed to be a regulation factor in adipogenesis. This study aimed to assess the promoting effect of vitamin C (VC) on adipogenic differentiation of preadipocytes as well as its mechanism. Methods Five sets of different combinations of chemicals were used to inhibit synthesis of type I to VI collagens, blocking ERK1/2 phosphorylation during adipogenesis of 3T3-L1 preadipocytes. Furthermore, to explore whether collagen VI plays a critical role during adipogenesis, specific knockdown of collagen VI was performed by using RNA interference. The morphology and expression patterns of several target factors involved in adipogenesis were assessed at various time points. Results A reduction in ERK1/2 phosphorylation and an increase in collagen VI and adipogenic-specific factors, such as C/EBPβ, PPARγ and C/EBPα, were observed after treating adipogenic 3T3-L1 cells with AA2P, a stable derivative of VC. Inhibition of collagen synthesis by ethyl-3, 4-dihydroxybenzoate (EDHB) or by specific knockdown of collagen VI by RNAi could promote ERK1/2 phosphorylation. The ERK1/2 phosphorylation in both cases could be attenuated by AA2P treatment. In addition, the inhibition of ERK1/2 phosphorylation by U0126, a highly selective inhibitor of both MEK1 and MEK2 and a type of MAPK/ERK kinase, up-regulated the expression of collagen VI, while it down-regulated the adipogenic-specific factors. Conclusion AA2P could up-regulate the expression of collagen VI by attenuating ERK1/2 phosphorylation, further up-regulating adipocyte-specific factors, thus finally promoting the adipogenesis of 3T3-L1 preadipocytes. Electronic supplementary material The online version of this article (10.1186/s12986-017-0234-y) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Perino MG, Yamanaka S, Riordon DR, Tarasova Y, Boheler KR. Ascorbic acid promotes cardiomyogenesis through SMAD1 signaling in differentiating mouse embryonic stem cells. PLoS One 2017; 12:e0188569. [PMID: 29232368 PMCID: PMC5726630 DOI: 10.1371/journal.pone.0188569] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 11/09/2017] [Indexed: 12/03/2022] Open
Abstract
Numerous groups have documented that Ascorbic Acid (AA) promotes cardiomyocyte differentiation from both mouse and human ESCs and iPSCs. AA is now considered indispensable for the routine production of hPSC-cardiomyocytes (CMs) using defined media; however, the mechanisms involved with the inductive process are poorly understood. Using a genetically modified mouse embryonic stem cell (mESC) line containing a dsRED transgene driven by the cardiac-restricted portion of the ncx1 promoter, we show that AA promoted differentiation of mESCs to CMs in a dose- and time-dependent manner. Treatment of mPSCs with AA did not modulate total SMAD content; however, the phosphorylated/active forms of SMAD2 and SMAD1/5/8 were significantly elevated. Co-administration of the SMAD2/3 activator Activin A with AA had no significant effect, but the addition of the nodal co-receptor TDGF1 (Cripto) antagonized AA’s cardiomyogenic-promoting ability. AA could also reverse some of the inhibitory effects on cardiomyogenesis of ALK/SMAD2 inhibition by SB431542, a TGFβ pathway inhibitor. Treatment with BMP2 and AA strongly amplified the positive cardiomyogenic effects of SMAD1/5/8 in a dose-dependent manner. AA could not, however, rescue dorsomorphin-mediated inhibition of ALK/SMAD1 activity. Using an inducible model system, we found that SMAD1, but not SMAD2, was essential for AA to promote the formation of TNNT2+-CMs. These data firmly demonstrate that BMP receptor-activated SMADs, preferential to TGFβ receptor-activated SMADs, are necessary to promote AA stimulated cardiomyogenesis. AA-enhanced cardiomyogenesis thus relies on the ability of AA to modulate the ratio of SMAD signaling among the TGFβ-superfamily receptor signaling pathways.
Collapse
Affiliation(s)
- Maria Grazia Perino
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Satoshi Yamanaka
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Daniel R. Riordon
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Yelena Tarasova
- Laboratory of Cardiovascular Science, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Kenneth R. Boheler
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, LKS Faculty of Medicine, University of Hong Kong, Hong Kong, SAR China
- Division of Cardiology, Johns Hopkins Medical Institute, Baltimore, Maryland, United States of America
| |
Collapse
|
19
|
Abbey D, Seshagiri PB. Ascorbic acid-mediated enhanced cardiomyocyte differentiation of mouse ES-cells involves interplay of DNA methylation and multiple-signals. Differentiation 2017; 96:1-14. [PMID: 28554048 DOI: 10.1016/j.diff.2017.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 02/23/2017] [Accepted: 04/25/2017] [Indexed: 01/10/2023]
Abstract
Embryonic stem cells (ES-cells) provide a good model system to study lineage-specific differentiation. Though, the differentiation of ES-cells to cardiomyocytes is documented, a clear understanding of the molecular mechanism of differentiation and improved functional-differentiation efficiency are yet to be achieved. In this regard, ascorbic acid (Aa) is shown to be one of the effective cardiac inducers in ES-cells. But, its mechanism is poorly understood. We therefore, investigated the mechanism of Aa-mediated cardiomyocyte differentiation of ES-cells. Here, we describe the potential involvement of epigenetic (DNA methylation) as well as integrin- and Erk- signaling systems during cardiomyocyte differentiation. Transgenic GS-2 ES-cells and wild-type D3 ES-cells were differentiated to cardiomyocytes, in the presence or absence of Aa and with or without inhibitors of Erk-, collagen- and integrin- pathways. At specific time points, differentiated states of ES-cells were scored by gene expression analyses and the proportion of functional cTnI+ cardiomyocytes. DNA methylation changes of Isl-1, BMP-2, GATA-4 and α-MHC in cardiogenic cells, following stimulation with Aa, were analyzed by using methylation specific PCR (MSP). We observed that Aa, when applied in initial phase of ES-cell differentiation, consistently enhanced cardiac differentiation (99%) over that observed during spontaneous differentiation (70%). This was associated with enhanced expressions of cardiogenesis-associated genes. A two-fold increase in cTnI+ cells was observed, with appropriate myofibril arrangement. The observed effect of Aa was due to enhanced collagen and integrin signaling, coupled with a high p-ERK1/2 expression, downstream. Besides, the involvement of DNA methylation in regulating the expression of cardiac genes i.e., Isl-1 and α-MHC was also observed. Overall, this study, for the first time, demonstrates that Aa-mediated cardiac enhancement is brought about, mechanistically, through the interplay of epigenetic changes in DNA methylation of cardiac genes (Isl-1 and α-MHC) and integrin signaling system.
Collapse
Affiliation(s)
- Deepti Abbey
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India
| | - Polani B Seshagiri
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore 560012, India.
| |
Collapse
|
20
|
Vitamin C in Stem Cell Biology: Impact on Extracellular Matrix Homeostasis and Epigenetics. Stem Cells Int 2017; 2017:8936156. [PMID: 28512473 PMCID: PMC5415867 DOI: 10.1155/2017/8936156] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/05/2017] [Indexed: 12/30/2022] Open
Abstract
Transcription factors and signaling molecules are well-known regulators of stem cell identity and behavior; however, increasing evidence indicates that environmental cues contribute to this complex network of stimuli, acting as crucial determinants of stem cell fate. l-Ascorbic acid (vitamin C (VitC)) has gained growing interest for its multiple functions and mechanisms of action, contributing to the homeostasis of normal tissues and organs as well as to tissue regeneration. Here, we review the main functions of VitC and its effects on stem cells, focusing on its activity as cofactor of Fe+2/αKG dioxygenases, which regulate the epigenetic signatures, the redox status, and the extracellular matrix (ECM) composition, depending on the enzymes' subcellular localization. Acting as cofactor of collagen prolyl hydroxylases in the endoplasmic reticulum, VitC regulates ECM/collagen homeostasis and plays a key role in the differentiation of mesenchymal stem cells towards osteoblasts, chondrocytes, and tendons. In the nucleus, VitC enhances the activity of DNA and histone demethylases, improving somatic cell reprogramming and pushing embryonic stem cell towards the naive pluripotent state. The broad spectrum of actions of VitC highlights its relevance for stem cell biology in both physiology and disease.
Collapse
|
21
|
Tohyama S, Tanosaki S, Someya S, Fujita J, Fukuda K. Manipulation of Pluripotent Stem Cell Metabolism for Clinical Application. CURRENT STEM CELL REPORTS 2017; 3:28-34. [PMID: 28261548 PMCID: PMC5315714 DOI: 10.1007/s40778-017-0073-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Pluripotent stem cells (PSCs) have the capacity to differentiate into various types of cells, and are promising cell sources for regenerative therapy and drug screening. However, to realize the clinical application of PSCs, a large number of highly qualified target cells must be stably prepared with low cost. To achieve this, great improvements in the reprogramming, differentiation, and elimination of residual PSCs will be necessary. In this review, we summarize the updated knowledge about metabolism in PSCs and its application. RECENT FINDINGS Recent studies have shown that PSCs have distinct metabolic profiles compared to differentiated cells. The metabolic profiles of PSCs are indispensable for the maintenance of pluripotency, self-renewal, differentiation capacity, and cell survival. SUMMARY Metabolic approaches show improved simplicity, scalability, and lower cost than conventional methods for differentiation and elimination of residual PSCs. Thus, manipulation of PSC metabolism will lead to new technologies to improve their efficiencies.
Collapse
Affiliation(s)
- Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582 Japan
- Department of Organ Fabrication, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Sho Tanosaki
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Shota Someya
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Jun Fujita
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582 Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, 35 Shinanomachi Shinjuku-ku, Tokyo, 160-8582 Japan
| |
Collapse
|
22
|
Kucinska-Lipka J, Janik H, Gubanska I. Ascorbic Acid in Polyurethane Systems for Tissue Engineering. CHEMISTRY & CHEMICAL TECHNOLOGY 2016. [DOI: 10.23939/chcht10.04si.607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The introduction of the paper was devoted to the main items of tissue engineering (TE) and the way of porous structure obtaining as scaffolds. Furthermore, the significant role of the scaffold design in TE was described. It was shown, that properly designed polyurethanes (PURs) find application in TE due to the proper physicochemical, mechanical and biological properties. Then the use of L-ascorbic acid (L-AA) in PUR systems for TE was described. L-AA has been applied in this area due to its suitable biological characteristics and antioxidative properties. Moreover, L-AA influences tissue regeneration due to improving collagen synthesis, which is a primary component of the extracellular matrix (ECM). Modification of PUR with L-AA leads to the materials with higher biocompatibility and such system is promising for TE applications.
Collapse
|
23
|
Li XL, Zeng D, Chen Y, Ding L, Li WJ, Wei T, Ou DB, Yan S, Wang B, Zheng QS. Role of alpha- and beta-adrenergic receptors in cardiomyocyte differentiation from murine-induced pluripotent stem cells. Cell Prolif 2016; 50. [PMID: 27790820 DOI: 10.1111/cpr.12310] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/13/2016] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a promising source of cells for regenerative heart disease therapies, but progress towards their use has been limited by their low differentiation efficiency and high cellular heterogeneity. Previous studies have demonstrated expression of adrenergic receptors (ARs) in stem cells after differentiation; however, roles of ARs in fate specification of stem cells, particularly in cardiomyocyte differentiation and development, have not been characterized. MATERIALS AND METHODS Murine-induced pluripotent stem cells (miPSCs) were cultured in hanging drops to form embryoid bodies, cells of which were then differentiated into cardiomyocytes. To determine whether ARs regulated miPSC differentiation into cardiac lineages, effects of the AR agonist, epinephrine (EPI), on miPSC differentiation and underlying signalling mechanisms, were evaluated. RESULTS Treatment with EPI, robustly enhanced miPSC cardiac differentiation, as indicated by increased expression levels of cardiac-specific markers, GATA4, Nkx2.5 and Tnnt2. Although β-AR signalling is the foremost signalling pathway in cardiomyocytes, EPI-enhanced cardiac differentiation depended more on α-AR signalling than β-AR signalling. In addition, selective activation of α1 -AR signalling with specific agonists induced vigorous cardiomyocyte differentiation, whereas selective activation of α2 - or β-AR signalling induced no or less differentiation, respectively. EPI- and α1 -AR-dependent cardiomyocyte differentiation from miPSCs occurred through specific promotion of CPC proliferation via the MEK-ERK1/2 pathway and regulation of miPS cell-cycle progression. CONCLUSIONS These results demonstrate that activation of ARs, particularly of α1 -ARs, promoted miPSC differentiation into cardiac lineages via MEK-ERK1/2 signalling.
Collapse
Affiliation(s)
- Xiao-Li Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Yan Chen
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China.,Department of Emergency, Chinese PLA No.401 Hospital, Qingdao, 266071, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Wen-Ju Li
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Dong-Bo Ou
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Song Yan
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Bin Wang
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| | - Qiang-Sun Zheng
- Department of Cardiology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, China
| |
Collapse
|
24
|
Aguado T, Gutiérrez FJ, Aix E, Schneider RP, Giovinazzo G, Blasco MA, Flores I. Telomere Length Defines the Cardiomyocyte Differentiation Potency of Mouse Induced Pluripotent Stem Cells. Stem Cells 2016; 35:362-373. [PMID: 27612935 DOI: 10.1002/stem.2497] [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: 03/02/2016] [Revised: 07/26/2016] [Accepted: 08/16/2016] [Indexed: 12/21/2022]
Abstract
Induced pluripotent stem cells (iPSCs) can be differentiated in vitro and in vivo to all cardiovascular lineages and are therefore a promising cell source for cardiac regenerative therapy. However, iPSC lines do not all differentiate into cardiomyocytes (CMs) with the same efficiency. Here, we show that telomerase-competent iPSCs with relatively long telomeres and high expression of the shelterin-complex protein TRF1 (iPSChighT ) differentiate sooner and more efficiently into CMs than those with relatively short telomeres and low TRF1 expression (iPSClowT ). Ascorbic acid, an enhancer of cardiomyocyte differentiation, further increases the cardiomyocyte yield from iPSChighT but does not rescue the cardiomyogenic potential of iPSClowT . Interestingly, although iPSCslowT differentiate very poorly to the mesoderm and endoderm lineages, they differentiate very efficiently to the ectoderm lineage, indicating that cell fate can be determined by in vitro selection of iPSCs with different telomere content. Our findings highlight the importance of selecting iPSCs with ample telomere reserves in order to generate high numbers of CMs in a fast, reliable, and efficient way. Stem Cells 2017;35:362-373.
Collapse
Affiliation(s)
- Tania Aguado
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Francisco J Gutiérrez
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Esther Aix
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - Ralph P Schneider
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Giovanna Giovinazzo
- Pluripotent Cell Technology Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| | - María A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Ignacio Flores
- Regeneration and Aging Group, Centro Nacional de Investigaciones Cardiovasculares (CNIC-ISCIII), Madrid, Spain
| |
Collapse
|
25
|
Kilberg MS, Terada N, Shan J. Influence of Amino Acid Metabolism on Embryonic Stem Cell Function and Differentiation. Adv Nutr 2016; 7:780S-9S. [PMID: 27422515 PMCID: PMC4942862 DOI: 10.3945/an.115.011031] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) have promise in regenerative medicine because of their ability to differentiate into all 3 primary germ layers. This review describes recent advances in the understanding of the link between the metabolism of ESCs/iPSCs and their maintenance/differentiation in the cell culture setting, with particular emphasis on amino acid (AA) metabolism. ESCs are endowed with unique metabolic features with regard to energy consumption, metabolite flux through particular pathways, and macromolecular synthesis. Therefore, nutrient availability has a strong influence on stem cell growth, self-renewal, and lineage specification, both in vivo and in vitro. Evidence from several laboratories has documented that self-renewal and differentiation of mouse ESCs are critically dependent on proline metabolism, with downstream metabolites possibly serving as signal molecules. Likewise, catabolism of either threonine (mouse) or methionine (human) is required for growth and differentiation of ESCs because these AAs serve as precursors for donor molecules used in histone methylation and acetylation. Epigenetic mechanisms are recognized as critical steps in differentiation, and AA metabolism in ESCs appears to modulate these epigenetic processes. Recent reports also document that, in vitro, the nutrient composition of the culture medium in which ESCs are differentiated into embryoid bodies can influence lineage specification, leading to enrichment of a specific cell type. Although research designed to direct tissue specification of differentiating embryoid bodies in culture is still in its infancy, early results indicate that manipulation of the nutrient milieu can promote or suppress the formation of specific cell lineages.
Collapse
Affiliation(s)
| | - Naohiro Terada
- Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Jixiu Shan
- Departments of Biochemistry and Molecular Biology and
| |
Collapse
|
26
|
Kucinska-Lipka J, Gubanska I, Janik H, Pokrywczynska M, Drewa T. l-ascorbic acid modified poly(ester urethane)s as a suitable candidates for soft tissue engineering applications. REACT FUNCT POLYM 2015. [DOI: 10.1016/j.reactfunctpolym.2015.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
27
|
Jiang B, Xiang Z, Ai Z, Wang H, Li Y, Ji W, Li T. Generation of cardiac spheres from primate pluripotent stem cells in a small molecule-based 3D system. Biomaterials 2015; 65:103-14. [DOI: 10.1016/j.biomaterials.2015.06.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/08/2015] [Accepted: 06/12/2015] [Indexed: 12/21/2022]
|
28
|
Affiliation(s)
- Dennis Schade
- Department
of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse
6, 44227 Dortmund, Germany
| | - Alleyn T. Plowright
- Department
of Medicinal Chemistry, Cardiovascular and Metabolic Diseases Innovative
Medicines, AstraZeneca, Pepparedsleden 1, Mölndal, 43183, Sweden
| |
Collapse
|
29
|
Shabani P, Ghazizadeh Z, Pahlavan S, Hashemizadeh S, Baharvand H, Aghdami N, Doosti M. Exogenous treatment with eicosapentaenoic acid supports maturation of cardiomyocytes derived from embryonic stem cells. Biochem Biophys Res Commun 2015; 461:281-6. [PMID: 25871791 DOI: 10.1016/j.bbrc.2015.04.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/03/2015] [Indexed: 11/29/2022]
Abstract
Embryonic stem cells offer multiple advantages over adult stem cells in terms of achieving acceptable number of functional cardiomyocytes to be exploited in cell therapy. However, differentiation efficacy is still a major issue to be solved before moving to regenerative medicine. Although a vast number of chemical compounds have been tested on efficiency of cardiac differentiation, the effect of fish oil components, such as eicosapentaenoic acid (EPA) on developmental bioenergetics, and hence cardiac differentiation, remained unstudied. EPA has been reported to have several cardioprotective effects, but there is no study addressing its role in cardiac differentiation. After mesoderm induction of embryoid bodies (EBs) derived from mouse embryonic stem cells (mESCs) in hanging drops initiated by ascorbic acid, they were treated with various concentrations of EPA. Gene and protein expression and functional properties of cardiomyocytes derived from ESCs were evaluated following treatment with various concentrations of EPA. Exposure to low concentrations of EPA (10 μM) increased percentage of beating colonies and beating area. This treatment also resulted in up to 3 fold increase in expression of NKX2-5, MEF2C, MYH6, TNNT2 and CX43. FACS analysis confirmed gene expression analysis with increased percentage of MYH6 positive cells in EPA-treated group compared to the control group. In contrast, the expression of genes coding for cardiac differentiation, remained constant or even declined with higher concentrations of EPA. In conclusion, we have demonstrated that treatment of mESCs undergoing cardiac differentiation with low concentration, but not high concentration of EPA up-regulate transcription of genes associated with cardiac development.
Collapse
Affiliation(s)
- Parisa Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zaniar Ghazizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shiva Hashemizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mahmood Doosti
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
30
|
Gandhimathi C, Venugopal JR, Tham AY, Ramakrishna S, Kumar SD. Biomimetic hybrid nanofibrous substrates for mesenchymal stem cells differentiation into osteogenic cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2015; 49:776-785. [DOI: 10.1016/j.msec.2015.01.075] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 12/16/2014] [Accepted: 01/23/2015] [Indexed: 12/16/2022]
|
31
|
Liu Y, Li P, Liu K, He Q, Han S, Sun X, Li T, Shen L. Timely inhibition of Notch signaling by DAPT promotes cardiac differentiation of murine pluripotent stem cells. PLoS One 2014; 9:e109588. [PMID: 25313563 PMCID: PMC4196912 DOI: 10.1371/journal.pone.0109588] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/02/2014] [Indexed: 12/21/2022] Open
Abstract
The Notch signaling pathway plays versatile roles during heart development. However, there is contradictory evidence that Notch pathway either facilitates or impairs cardiomyogenesis in vitro. In this study, we developed iPSCs by reprogramming of murine fibroblasts with GFP expression governed by Oct4 promoter, and identified an effective strategy to enhance cardiac differentiation through timely modulation of Notch signaling. The Notch inhibitor DAPT (N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester) alone drove the iPSCs to a neuronal fate. After mesoderm induction of embryoid bodies initiated by ascorbic acid (AA), the subsequent treatment of DAPT accelerated the generation of spontaneously beating cardiomyocytes. The timed synergy of AA and DAPT yielded an optimal efficiency of cardiac differentiation. Mechanistic studies showed that Notch pathway plays a biphasic role in cardiomyogenesis. It favors the early–stage cardiac differentiation, but exerts negative effects on the late-stage differentiation. Therefore, DAPT administration at the late stage enforced the inhibition of endogenous Notch activity, thereby enhancing cardiomyogenesis. In parallel, DAPT dramatically augmented the expression of Wnt3a, Wnt11, BMP2, and BMP4. In conclusion, our results highlight a practicable approach to generate cardiomyocytes from iPSCs based on the stage-specific biphasic roles of Notch signaling in cardiomyogenesis.
Collapse
Affiliation(s)
- Yinan Liu
- Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University, Haidian District, Beijing, China
| | - Peng Li
- Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University, Haidian District, Beijing, China
| | - Kaiyu Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University, Haidian District, Beijing, China
| | - Qihua He
- Center of Medical and Health Analysis, School of Basic Medical Sciences, Peking University, Haidian District, Beijing, China
| | - Shuo Han
- Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University, Haidian District, Beijing, China
| | - Xiaofeng Sun
- Department of histology and embryology, Institute of Chinese Medicine, Hunan University of Chinese Medicine, Science Garden District of Hanpu, Changsha, Hunan, China
| | - Tao Li
- Department of Biology, College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- * E-mail: (TL); (LS)
| | - Li Shen
- Stem Cell Research Center, Department of Cell Biology, School of Basic Medical Sciences, Peking University, Haidian District, Beijing, China
- * E-mail: (TL); (LS)
| |
Collapse
|
32
|
Shen X, Yang Q, Jin P, Li X. Alpha-lipoic acid enhances DMSO-induced cardiomyogenic differentiation of P19 cells. Acta Biochim Biophys Sin (Shanghai) 2014; 46:766-73. [PMID: 25112287 DOI: 10.1093/abbs/gmu057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alpha-lipoic acid (α-LA) is a potent antioxidant that acts as an essential cofactor in mitochondrial dehydrogenase reactions. α-LA has been shown to possess anti-inflammatory and cytoprotective properties, and is used to improve symptoms of diabetic neuropathy. However, the role of α-LA in stem cell differentiation and the underlying molecular mechanisms remain unknown. In the present study, we showed that α-LA significantly promoted dimethyl sulfoxide (DMSO)-induced cardiomyogenic differentiation of mouse embryonic carcinoma P19 cells. α-LA dose dependently increased beating embryonic body (EB) percentages of DMSO-differentiated P19 cells. The expressions of cardiac specific genes TNNT2, Nkx2.5, GATA4, MEF2C, and MLC2V and cardiac isoform of troponin T (cTnT)-positively stained cell population were significantly up-regulated by the addition of α-LA. We also demonstrated that the differentiation time after EB formation was critical for α-LA to take effect. Interestingly, without DMSO treatment, α-LA did not stimulate the cardiomyogenic differentiation of P19 cells. Further investigation indicated that collagen synthesis-enhancing activity, instead of the antioxidative property, plays a significant role in the cardiomyogenic differentiation-promoting function of α-LA. These findings highlight the potential use of α-LA for regenerative therapies in heart diseases.
Collapse
Affiliation(s)
- Xinghua Shen
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Qinghui Yang
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Peng Jin
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Xueqi Li
- Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| |
Collapse
|
33
|
Yu J, Tu YK, Tang YB, Cheng NC. Stemness and transdifferentiation of adipose-derived stem cells using l-ascorbic acid 2-phosphate-induced cell sheet formation. Biomaterials 2014; 35:3516-26. [DOI: 10.1016/j.biomaterials.2014.01.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/08/2014] [Indexed: 12/29/2022]
|
34
|
Aikawa H, Tamai M, Mitamura K, Itmainati F, Barber GN, Tagawa YI. Innate immunity in an in vitro murine blastocyst model using embryonic and trophoblast stem cells. J Biosci Bioeng 2014; 117:358-65. [DOI: 10.1016/j.jbiosc.2013.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/02/2013] [Accepted: 09/02/2013] [Indexed: 01/16/2023]
|
35
|
Promotion of cardiac differentiation of brown adipose derived stem cells by chitosan hydrogel for repair after myocardial infarction. Biomaterials 2014; 35:3986-98. [PMID: 24508080 DOI: 10.1016/j.biomaterials.2014.01.021] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/08/2014] [Indexed: 02/03/2023]
Abstract
The ability to restore heart function by replacement of diseased myocardium is one of the great challenges in biomaterials and regenerative medicine. Brown adipose derived stem cells (BADSCs) present a new source of cardiomyocytes to regenerate the myocardium after infarction. In this study, we explored an injectable tissue engineering strategy to repair damaged myocardium, in which chitosan hydrogels were investigated as a carrier for BADSCs. In vitro, the effect and mechanism of chitosan components on the cardiac differentiation of BADSCs were investigated. In vivo, BADSCs carrying double-fusion reporter gene (firefly luciferase and monomeric red fluorescent protein (fluc-mRFP)) were transplanted into infarcted rat hearts with or without chitosan hydrogel. Multi-techniques were used to assess the effects of treatments. We observed that chitosan components significantly enhanced cardiac differentiation of BADSCs, which was assessed by percentages of cTnT(+) cells and expression of cardiac-specific markers, including GATA-4, Nkx2.5, Myl7, Myh6, cTnI, and Cacna1a. Treatment with collagen synthesis inhibitors, cis-4-hydroxy-D-proline (CIS), significantly inhibited the chitosan-enhanced cardiac differentiation, indicating that the enhanced collagen synthesis by chitosan accounts for its promotive role in cardiac differentiation of BADSCs. Longitudinal in vivo bioluminescence imaging and histological staining revealed that chitosan enhanced the survival of engrafted BADSCs and significantly increased the differentiation rate of BADSCs into cardiomyocytes in vivo. Furthermore, BADSCs delivered by chitosan hydrogel prevented adverse matrix remodeling, increased angiogenesis, and preserved heart function. These results suggested that the injectable cardiac tissue engineering based on chitosan hydrogel and BADSCs is a useful strategy for myocardium regeneration.
Collapse
|
36
|
Koike K, Ishigami A, Sato Y, Hirai T, Yuan Y, Kobayashi E, Tobino K, Sato T, Sekiya M, Takahashi K, Fukuchi Y, Maruyama N, Seyama K. Vitamin C prevents cigarette smoke-induced pulmonary emphysema in mice and provides pulmonary restoration. Am J Respir Cell Mol Biol 2014; 50:347-57. [PMID: 24032444 DOI: 10.1165/rcmb.2013-0121oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Vitamin C (VC) is a potent antioxidant and is essential for collagen synthesis. We investigated whether VC treatment prevents and cures smoke-induced emphysema in senescence marker protein-30 knockout (SMP30-KO) mice, which cannot synthesize VC. Two smoke-exposure experiments using SMP30-KO mice were conducted. In the first one (a preventive study), 4-month-old mice received minimal VC (0.0375 g/l) [VC(L)] or physiologically sufficient VC (1.5 g/l) [VC(S)] and exposed to cigarette smoke or smoke-free air for 2 months. Pulmonary evaluations followed when the mice were 6 months of age. The second study began after the establishment of smoke-induced emphysema (a treatment study). These mice no longer underwent smoke exposure but received VC(S) or VC(L) treatment for 2 months. Morphometric analysis was performed, and measurements of oxidative stress, collagen synthesis, and vascular endothelial growth factor in the lungs were evaluated. Chronic smoke exposure caused emphysema (29.6% increases of mean linear intercepts [MLI] and 106.5% increases of destructive index compared with the air-only group) in 6-month-old SMP30-KO mice, and this emphysema closely resembled human chronic obstructive pulmonary disease. Smoke-induced emphysema persisted in the VC(L) group after smoking cessation, whereas VC treatment provided pulmonary restoration (18.5% decrease of MLI and 41.3% decrease of destructive index compared with VC(L) group). VC treatment diminished oxidative stress, increased collagen synthesis, and improved vascular endothelial growth factor levels in the lungs. Our results suggest that VC not only prevents smoke-induced emphysema in SMP30-KO mice but also restores emphysematous lungs. Therefore, VC may provide a new therapeutic strategy for treating chronic obstructive pulmonary disease in humans.
Collapse
Affiliation(s)
- Kengo Koike
- 1 Division of Respiratory Medicine, Juntendo University Faculty of Medicine and Graduate School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Park J, Park S, Ryu S, Bhang SH, Kim J, Yoon JK, Park YH, Cho SP, Lee S, Hong BH, Kim BS. Graphene-regulated cardiomyogenic differentiation process of mesenchymal stem cells by enhancing the expression of extracellular matrix proteins and cell signaling molecules. Adv Healthc Mater 2014; 3:176-81. [PMID: 23949999 DOI: 10.1002/adhm.201300177] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 06/28/2013] [Indexed: 12/26/2022]
Abstract
The potential of graphene as a mesenchymal stem cell (MSC) culture substrate to promote cardiomyogenic differentiation is demonstrated. Graphene exhibits no sign of cytotoxicity for stem cell culture. MSCs are committed toward cardiomyogenic lineage by simply culturing them on graphene. This may be attributed, at least partially, to the regulation of expression levels of extracellular matrix and signaling molecules.
Collapse
Affiliation(s)
- Jooyeon Park
- School of Chemical and Biological Engineering, Seoul National University, Seoul 151-744, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Wasik AM, Grabarek J, Pantovic A, Cieślar-Pobuda A, Asgari HR, Bundgaard-Nielsen C, Rafat M, Dixon IMC, Ghavami S, Łos MJ. Reprogramming and carcinogenesis--parallels and distinctions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 308:167-203. [PMID: 24411172 DOI: 10.1016/b978-0-12-800097-7.00005-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Rapid progress made in various areas of regenerative medicine in recent years occurred both at the cellular level, with the Nobel prize-winning discovery of reprogramming (generation of induced pluripotent stem (iPS) cells) and also at the biomaterial level. The use of four transcription factors, Oct3/4, Sox2, c-Myc, and Klf4 (called commonly "Yamanaka factors") for the conversion of differentiated cells, back to the pluripotent/embryonic stage, has opened virtually endless and ethically acceptable source of stem cells for medical use. Various types of stem cells are becoming increasingly popular as starting components for the development of replacement tissues, or artificial organs. Interestingly, many of the transcription factors, key to the maintenance of stemness phenotype in various cells, are also overexpressed in cancer (stem) cells, and some of them may find the use as prognostic factors. In this review, we describe various methods of iPS creation, followed by overview of factors known to interfere with the efficiency of reprogramming. Next, we discuss similarities between cancer stem cells and various stem cell types. Final paragraphs are dedicated to interaction of biomaterials with tissues, various adverse reactions generated as a result of such interactions, and measures available, that allow for mitigation of such negative effects.
Collapse
Affiliation(s)
- Agata M Wasik
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Jerzy Grabarek
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Aleksandar Pantovic
- Institute of Microbiology and Immunology, School of Medicine, University of Belgrade, and Clinic of Neurology, Military Medical Academy, Belgrade, Serbia
| | - Artur Cieślar-Pobuda
- Department of Clinical and Experimental Medicine (IKE), Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University, Linköping, Sweden; Biosystems Group, Institute of Automatic Control, Silesian University of Technology, Gliwice, Poland
| | | | - Caspar Bundgaard-Nielsen
- Department of Clinical and Experimental Medicine (IKE), Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University, Linköping, Sweden; Laboratory for Stem Cell Research, Aalborg University, Aalborg, Denmark
| | - Mehrdad Rafat
- Department of Clinical and Experimental Medicine (IKE), Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University, Linköping, Sweden; Department of Biomedical Engineering (IMT), Linköping University, Linköping, Sweden
| | - Ian M C Dixon
- Department of Physiology, St. Boniface Research Centre, and Manitoba Institute of Child Health, University of Manitoba, Winnipeg, Canada
| | - Saeid Ghavami
- Department of Physiology, St. Boniface Research Centre, and Manitoba Institute of Child Health, University of Manitoba, Winnipeg, Canada
| | - Marek J Łos
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland; Department of Clinical and Experimental Medicine (IKE), Division of Cell Biology, and Integrative Regenerative Medicine Center (IGEN), Linköping University, Linköping, Sweden; BioApplications Enterprises, Winnipeg, Manitoba, Canada.
| |
Collapse
|
39
|
Matrix identity and tractional forces influence indirect cardiac reprogramming. Sci Rep 2013; 3:3474. [PMID: 24326998 PMCID: PMC3858798 DOI: 10.1038/srep03474] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/25/2013] [Indexed: 12/22/2022] Open
Abstract
Heart regeneration through in vivo cardiac reprogramming has been demonstrated as a possible regenerative strategy. While it has been reported that cardiac reprogramming in vivo is more efficient than in vitro, the influence of the extracellular microenvironment on cardiac reprogramming remains incompletely understood. This understanding is necessary to improve the efficiency of cardiac reprogramming in order to implement this strategy successfully. Here we have identified matrix identity and cell-generated tractional forces as key determinants of the dedifferentiation and differentiation stages during reprogramming. Cell proliferation, matrix mechanics, and matrix microstructure are also important, but play lesser roles. Our results suggest that the extracellular microenvironment can be optimized to enhance cardiac reprogramming.
Collapse
|
40
|
Wang J, Cao H, Xue X, Fan C, Fang F, Zhou J, Zhang Y, Zhang X. Effect of vitamin C on growth of caprine spermatogonial stem cells in vitro. Theriogenology 2013; 81:545-55. [PMID: 24368149 DOI: 10.1016/j.theriogenology.2013.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 11/02/2013] [Accepted: 11/08/2013] [Indexed: 01/15/2023]
Abstract
The genetic manipulation of spermatogonial stem cells (SSCs) can be used for the production of transgenic animals in a wide range of species. However, this technology is limited by the absence of an ideal culture system in which SSCs can be maintained and proliferated, especially in domestic animals like the goat. The aim of this study therefore was to investigate whether the addition of vitamin C (Vc) in cell culture influences the growth of caprine SSCs. Various concentrations of Vc (0, 5, 10, 25, 40, and 50 μg/mL(-1)) were added to SSC culture media, and their effect on morphology and alkaline phosphatase activity was studied. The number of caprine SSC colonies and area covered by them were measured at 10 days of culture. The expression of various germ cell and somatic cell markers such as VASA, integrins, Oct-4, GATA-4, α-SMA, vimentin, and Thy-1 was studied to identify the proliferated cells using immunostaining analyses. Further, the intracellular reactive oxygen species (ROS) level was measured at the 3rd, 6th, and 9th day after culture, and expression of Bax, Bcl-2, and P53, factors involved in the regulation of apoptosis, were analyzed on the 7th day after culture using reverse transcription polymerase chain reaction and quantitative real-time polymerase chain reaction. The results showed that the SSCs formed compact colonies and had unclear borders in the different Vc-supplemented groups at 10 days, and there were no major morphologic differences between the groups. The number and area of colonies were both the highest in the 40 μg/mL(-1) Vc group. Differential expression of markers for germ cells, undifferentiated spermatogonia, and testis somatic cells was observed. Cultured germ cell clumps were found to have alkaline phosphatase activity regardless of the Vc dose. The number of Thy-1- and Oct-4-positive cells was the most in the 40 μg/mL(-1) Vc group. Moreover, the level of ROS was dependent on the Vc dose and culture time. The Vc dose 40 μg/mL(-1) was found to be optimum with regard to decreasing ROS generation, and increasing the expression of the antiapoptotic gene Bcl-2 and decreasing the expression of the proapoptotic genes Bax and P53. In conclusion, the addition of 40 μg/mL(-1) Vc can maintain a certain physiological level of ROS, trigger the expression of the antiapoptosis gene Bcl-2, suppress the proapoptotic gene P53 and Bax pathway, and further promote the proliferation of caprine SSCs in vitro.
Collapse
Affiliation(s)
- Juhua Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Hongguo Cao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Xiuheng Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Caiyun Fan
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Fugui Fang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Jie Zhou
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China
| | - Yunhai Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China; Anhui Provincial Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Hefei, Anhui, China
| | - Xiaorong Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, China; Anhui Provincial Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-Breeding, Hefei, Anhui, China; Engineering Research Center of Reproduction and breeding in Sheep of Anhui Province, Hefei, Anhui, China.
| |
Collapse
|
41
|
Date Y, Hasegawa S, Yamada T, Inoue Y, Mizutani H, Nakata S, Akamatsu H. Major amino acids in collagen hydrolysate regulate the differentiation of mouse embryoid bodies. J Biosci Bioeng 2013; 116:386-90. [PMID: 23623897 DOI: 10.1016/j.jbiosc.2013.03.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/14/2013] [Accepted: 03/21/2013] [Indexed: 10/26/2022]
Abstract
To take advantage of the therapeutic potential of embryonic stem cells (ESCs), it is necessary to regulate their differentiation in response to defined factors. In this study, in order to explore novel molecules that regulate the differentiation of ESCs, we investigated whether collagen hydrolysate, collagen-characteristic amino acids, glycine (Gly), l-proline and trans-4-hydroxy-l-proline (l-Hyp); or dipeptides, proline-hydroxyproline and hydroxyproline-glycine regulate the differentiation of mouse embryoid bodies (EBs). We identified that treatment with collagen hydrolysate or Gly repressed the expression of the mesendodermal markers, Brachyury and Foxa2 in EBs and maintained the undifferentiated state of mESCs in a feeder-free monolayer culture. In contrast, l-Hyp promoted the expression of Brachyury, Mixl1, Gsc and Foxa2 in EBs. And the treatment with l-Hyp promoted cardiac differentiation within EBs, which was proven by the spontaneous contraction of cardiomyocytes and the expression of the cardiac markers, α-MHC, MLC-2v and Nkx2.5. Results suggest that l-Hyp is a promising new inducer for reproducible and efficient differentiation of mesendoderm lineages.
Collapse
Affiliation(s)
- Yasushi Date
- Research Laboratories, Nippon Menard Cosmetic Co., Ltd., 2-7 Torimicho, Nishi-Ku, Nagoya, Aichi 451-0071, Japan.
| | | | | | | | | | | | | |
Collapse
|
42
|
Zeng D, Ou DB, Wei T, Ding L, Liu XT, Hu XL, Li X, Zheng QS. Collagen/β(1) integrin interaction is required for embryoid body formation during cardiogenesis from murine induced pluripotent stem cells. BMC Cell Biol 2013; 14:5. [PMID: 23350814 PMCID: PMC3562267 DOI: 10.1186/1471-2121-14-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Accepted: 01/23/2013] [Indexed: 01/16/2023] Open
Abstract
Background The interactions between stem cells and extracellular matrix (ECM) mediated by integrins play important roles in the processes that determine stem cell fate. However, the role of ECM/integrin interaction in the formation of embryoid bodies (EBs) during cardiogenesis from murine induced pluripotent stem cells (miPSCs) remains unclear. Results In the present study, collagen type I and β1 integrin were expressed and upregulated synergistically during the formation of miPSC-derived EBs, with a peak expression at day 3 of differentiation. The blockage of collagen/β1 integrin interaction by β1 integrin blocking antibody resulted in the production of defective EBs that were characterized by decreased size and the absence of a shell-like layer composed of primitive endoderm cells. The quantification of spontaneous beating activity, cardiac-specific gene expression and cardiac troponin T (cTnT) immunostaining showed that the cardiac differentiation of these defective miPSC-derived EBs was lower than that of control EBs. Conclusions These findings indicate that collagen/β1 integrin interaction is required for the growth and cardiac differentiation of miPSC-derived EBs and will be helpful in future engineering of the matrix microenvironment within EBs to efficiently direct the cardiac fate of pluripotent stem cells to promote cardiovascular regeneration.
Collapse
Affiliation(s)
- Di Zeng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Zainal Ariffin SH, Kermani S, Megat Abdul Wahab R, Senafi S, Zainal Ariffin Z, Abdul Razak M. In vitro chondrogenesis transformation study of mouse dental pulp stem cells. ScientificWorldJournal 2012; 2012:827149. [PMID: 22919354 PMCID: PMC3419417 DOI: 10.1100/2012/827149] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 06/13/2012] [Indexed: 12/18/2022] Open
Abstract
A major challenge in the application of mesenchymal stem cells in cartilage reconstruction is that whether the cells are able to differentiate into fully mature chondrocytes before grafting. The aim of this study was to isolate mouse dental pulp stem cells (DPSC) and differentiate them into chondrocytes. For this investigation, morphological, molecular, and biochemical analyses for differentiated cells were used. To induce the chondrocyte differentiation, DPSC were cultured in chondrogenic medium (Zen-Bio, Inc.). Based on morphological analyses using toluidine blue staining, proteoglycan products appear in DPSC after 21 days of chondrocyte induction. Biochemical analyses in differentiated group showed that alkaline phosphatase activity was significantly increased at day 14 as compared to control (P < 0.05). Cell viability analyses during the differentiation to chondrocytes also showed that these cells were viable during differentiation. However, after the 14th day of differentiation, there was a significant decrease (P < 0.05) in the viability proportion among differentiated cells as compared to the control cells. In RT-PCR molecular analyses, mouse DPSC expressed Cd146 and Cd166 which indicated that these cells belong to mesenchymal stem cells. Coll I and Coll II markers showed high expression after 14 and 21 days, respectively. In conclusion, this study showed that DPSC successfully differentiated into chondrocytes.
Collapse
Affiliation(s)
- Shahrul Hisham Zainal Ariffin
- School of Biosciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia.
| | | | | | | | | | | |
Collapse
|
44
|
Bax NAM, van Marion MH, Shah B, Goumans MJ, Bouten CVC, van der Schaft DWJ. Matrix production and remodeling capacity of cardiomyocyte progenitor cells during in vitro differentiation. J Mol Cell Cardiol 2012; 53:497-508. [PMID: 22820459 DOI: 10.1016/j.yjmcc.2012.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 07/02/2012] [Accepted: 07/11/2012] [Indexed: 11/25/2022]
Abstract
Cell-based therapy has emerged as a treatment modality for myocardial repair. Especially cardiac resident stem cells are considered a potential cell source since they are able to differentiate into cardiomyocytes and have improved heart function after injury in a preclinical model for myocardial infarction. To avoid or repair myocardial damage it is important not only to replace the lost cardiomyocytes, but also to remodel and replace the scar tissue by "healthy" extracellular matrix (ECM). Interestingly, the role of cardiac stem cells in this facet of cardiac repair is largely unknown. Therefore, we investigated the expression and production of ECM proteins, matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) in human cardiomyocyte progenitor cells (CMPCs) undergoing differentiation towards the cardiomyogenic lineage. Our data suggest that CMPCs have the capacity to synthesize and modulate their own matrix environment, especially during differentiation towards the cardiomyogenic lineage. While undifferentiated CMPCs expressed collagen I, III, IV and fibronectin, but no elastin, during the process of differentiation the expression of collagen I, III, IV and fibronectin increased and interestingly also elastin expression was induced. Furthermore, undifferentiated CMPCs express MMP-1 -2 and -9 and upon differentiation the expression of MMP-1 decreased, while the expression of MMP-2 and MMP-9, although the latter only in the early stage of differentiation, increased. Additionally, the expression of TIMP-1, -2 and -4 was induced during differentiation. This study provides new insights into the matrix production and remodeling capacity of human CMPCs, with potential beneficial effects for the treatment of cardiac injury.
Collapse
Affiliation(s)
- Noortje A M Bax
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | | | | | | | | | | |
Collapse
|
45
|
Synergistic effects of hypoxia and extracellular matrix cues in cardiomyogenesis. Biomaterials 2012; 33:6313-9. [PMID: 22717366 DOI: 10.1016/j.biomaterials.2012.05.063] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 05/27/2012] [Indexed: 11/23/2022]
Abstract
Limited characterization of how the stem cell niche evolves has hindered our ability to mimic the physiological environment. In this paper, we hypothesized that hypoxia-induced extracellular matrix (ECM) cues may facilitate cardiomyogenesis. We evaluated the expression of four ECM proteins - fibronectin, collagen I, collagen IV, and laminin - over a period of 20 days in H1 and H9 human embryonic stem cell-derived embryoid bodies (EBs) under hypoxic (5% oxygen) and normoxic (21% oxygen) conditions. Hypoxic EBs exhibited increased collagen I, collagen IV and fibronectin expression relative to normoxic EBs between days 9-13, which coincided with increased expression of mesoderm genes. The effect of ECM cues was confirmed by plating day 9 EBs on collagen IV, gelatin, and fibronectin-rich substrates for 11 days. Hypoxia/gelatin cultures synergistically increased the cardiomyocyte yield by 1.7 and 5.5 fold relative to normoxia/gelatin and normoxia/collagen IV cultures, respectively. Current differentiation protocols may underestimate the contribution of hypoxia and ECM cues that evolve during EB maturation.
Collapse
|
46
|
Liu J, Zhang Z, Liu Y, Guo C, Gong Y, Yang S, Ma M, Li Z, Gao WQ, He Z. Generation, characterization, and potential therapeutic applications of cardiomyocytes from various stem cells. Stem Cells Dev 2012; 21:2095-110. [PMID: 22428725 DOI: 10.1089/scd.2012.0031] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Heart failure is one of the leading causes of death worldwide. Myocardial cell transplantation emerges as a novel therapeutic strategy for heart failure, but this approach has been hampered by severe shortage of human cardiomyocytes. We have recently induced mouse embryonic stem cells to differentiate into embryoid bodies and eventually, cardiomyocytes. Here, we address recent advancements in cardiomyocyte differentiation from cardiac stem cells and pluripotent stem cells. We highlight the methodologies, using growth factors, endoderm-like cell cocultures, small molecules, and biomaterials, in directing the differentiation of pluripotent stem cells into cardiomyocytes. The characterization and identification of pluripotent stem cell-derived cardiomyocytes by morphological, phenotypic, and functional features are also discussed. Notably, increasing evidence demonstrates that cardiomyocytes may be generated from the stem cells of several tissues outside the cardiovascular system, including skeletal muscles, bone marrow, testes, placenta, amniotic fluid, and adipose tissues. We further address the potential applications of cardiomyocytes derived from various kinds of stem cells. The differentiation of stem cells into functional cardiomyocytes, especially from an extra-cardiac stem cell source, would circumvent the scarcity of heart donors and human cardiomyocytes, and, most importantly, it would offer an ideal and promising cardiomyocyte source for cell therapy and tissue engineering in treating heart failure.
Collapse
Affiliation(s)
- Jianfang Liu
- Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
When considering the history of vitamin C, and the names given to this molecule in early days, the Latin proverb nomen est omen suddenly comes to mind. Around 1920, when Casimir Funk introduced the term Vitamin C to indicate the nutritional factor necessary to prevent the pathological state known as scurvy, the nature of the active molecule was still unknown (Davies MB, Austin J, Partridge DA (1991) Vitamin C: Its chemistry and biochemistry. The Royal Society of Chemistry, Cambridge UK). Almost in the same years, Albert Szent-Giörgyi was striving to identify a new 6-carbon sugar he had obtained in crystal form from oranges, lemons, cabbage and adrenal glands. As humorously described by Szent-Giörgyi himself (Szent-Giörgyi A (1963) Lost in the twentieth century. Annu Rev Biochem 36:1-15), he intended to name this yet unknown carbohydrate "ignose". When this name was rejected by Sir Arthur Harden, editor of the Biochemical Journal, he suggested to name it "godnose", meaning that only God could know the real identity of the molecule. Obviously, also this choice was considered inappropriate by Harden, who suggested the plain name "hexuronic acid". Only later, when the structure of "hexuronic acid" had been completely elucidated, and biological tests performed by Swirbely identified this molecule as the anti-scurvy factor vitamin C, Szent-Giörgyi and Walter Norman Haworth decided to eventually name it ascorbic acid (Szent-Giörgyi A (1963) Lost in the twentieth century. Annu Rev Biochem 36:1-15). "Ascorbic" literally means "against scurvy", and scurvy is known to be mainly due to the inactivation of some important dioxygenases involved in the synthesis of a few key molecules, including different collagen forms (De Tullio MC (2004) How does ascorbic acid prevent scurvy? A survey of the nonantioxidant functions of vitamin C. In: Asard H, May J, Smirnoff N (eds) Vitamin C, its functions and biochemistry in animals and plants. Bios Scientific Publishers, Oxford, UK, pp. 159-172). All this has very little to do with the celebrated role of ascorbic acid (ASC) as an antioxidant. So, if the fate of ASC had to be found in its name, its role in the prevention of scurvy (i.e. beyond the antioxidant function) should be considered its main feature. But, in spite of more than 80 years of extensive research (34,424 hits in a PubMed query on January 6 2007), an unprecedented popularity among the general public, an estimated market of several billion dollars (Hancock RD, Viola R (2005) Improving the nutritional value of crops through enhancement of l-ascorbic acid (vitamin C) content: Rationale and biotechnological opportunities. J Agr Food Chem 53:5248-5257), we should honestly conclude that the fate of vitamin C is still in the first name it received, many years ago: we still ignore much of its actual relevance in cell metabolism, although we are progressively getting aware of the many facets of this fascinating molecule, and its direct involvement in the regulation of apparently unrelated pathways (Arrigoni O, De Tullio MC (2002) Ascorbic acid, much more than just an antioxidant. Biochim Biophys Acta 1569:1-9; De Tullio MC, Arrigoni O (2004) Hopes, disillusions and more hopes from vitamin C. Cell Mol Life Sci 61:209-219; Duarte TL, Lunec J (2005) When is an antioxidant not an antioxidant? A review of novel actions and reactions of vitamin C. Free Rad Res 39:671-686). Recent data on ASC involvement in cell signalling and gene expression open new perspectives, that will be presented and discussed in this chapter.
Collapse
Affiliation(s)
- Mario C De Tullio
- Dipartimento di Biologia e Patologia Vegetale, Universita' di Bari, 70125, Bari, Italia,
| |
Collapse
|
48
|
Shafa M, Krawetz R, Zhang Y, Rattner JB, Godollei A, Duff HJ, Rancourt DE. Impact of stirred suspension bioreactor culture on the differentiation of murine embryonic stem cells into cardiomyocytes. BMC Cell Biol 2011; 12:53. [PMID: 22168552 PMCID: PMC3260255 DOI: 10.1186/1471-2121-12-53] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 12/14/2011] [Indexed: 12/11/2022] Open
Abstract
Background Embryonic stem cells (ESCs) can proliferate endlessly and are able to differentiate into all cell lineages that make up the adult organism. Under particular in vitro culture conditions, ESCs can be expanded and induced to differentiate into cardiomyocytes in stirred suspension bioreactors (SSBs). However, in using these systems we must be cognizant of the mechanical forces acting upon the cells. The effect of mechanical forces and shear stress on ESC pluripotency and differentiation has yet to be clarified. The purpose of this study was to investigate the impact of the suspension culture environment on ESC pluripotency during cardiomyocyte differentiation. Results Murine D3-MHC-neor ESCs formed embyroid bodies (EBs) and differentiated into cardiomyocytes over 25 days in static culture and suspension bioreactors. G418 (Geneticin) was used in both systems from day 10 to enrich for cardiomyocytes by eliminating non-resistant, undifferentiated cells. Treatment of EBs with 1 mM ascorbic acid and 0.5% dimethyl sulfoxide from day 3 markedly increased the number of beating EBs, which displayed spontaneous and cadenced contractile beating on day 11 in the bioreactor. Our results showed that the bioreactor differentiated cells displayed the characteristics of fully functional cardiomyocytes. Remarkably, however, our results demonstrated that the bioreactor differentiated ESCs retained their ability to express pluripotency markers, to form ESC-like colonies, and to generate teratomas upon transplantation, whereas the cells differentiated in adherent culture lost these characteristics. Conclusions This study demonstrates that although cardiomyocyte differentiation can be achieved in stirred suspension bioreactors, the addition of medium enhancers is not adequate to force complete differentiation as fluid shear forces appear to maintain a subpopulation of cells in a transient pluripotent state. The development of successful ESC differentiation protocols within suspension bioreactors demands a more complete understanding of the impacts of shear forces on the regulation of pluripotency and differentiation in pluripotent stem cells.
Collapse
Affiliation(s)
- Mehdi Shafa
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Calgary, AB, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Cao N, Liu Z, Chen Z, Wang J, Chen T, Zhao X, Ma Y, Qin L, Kang J, Wei B, Wang L, Jin Y, Yang HT. Ascorbic acid enhances the cardiac differentiation of induced pluripotent stem cells through promoting the proliferation of cardiac progenitor cells. Cell Res 2011; 22:219-36. [PMID: 22143566 DOI: 10.1038/cr.2011.195] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Generation of induced pluripotent stem cells (iPSCs) has opened new avenues for the investigation of heart diseases, drug screening and potential autologous cardiac regeneration. However, their application is hampered by inefficient cardiac differentiation, high interline variability, and poor maturation of iPSC-derived cardiomyocytes (iPS-CMs). To identify efficient inducers for cardiac differentiation and maturation of iPSCs and elucidate the mechanisms, we systematically screened sixteen cardiomyocyte inducers on various murine (m) iPSCs and found that only ascorbic acid (AA) consistently and robustly enhanced the cardiac differentiation of eleven lines including eight without spontaneous cardiogenic potential. We then optimized the treatment conditions and demonstrated that differentiation day 2-6, a period for the specification of cardiac progenitor cells (CPCs), was a critical time for AA to take effect. This was further confirmed by the fact that AA increased the expression of cardiovascular but not mesodermal markers. Noteworthily, AA treatment led to approximately 7.3-fold (miPSCs) and 30.2-fold (human iPSCs) augment in the yield of iPS-CMs. Such effect was attributed to a specific increase in the proliferation of CPCs via the MEK-ERK1/2 pathway by through promoting collagen synthesis. In addition, AA-induced cardiomyocytes showed better sarcomeric organization and enhanced responses of action potentials and calcium transients to β-adrenergic and muscarinic stimulations. These findings demonstrate that AA is a suitable cardiomyocyte inducer for iPSCs to improve cardiac differentiation and maturation simply, universally, and efficiently. These findings also highlight the importance of stimulating CPC proliferation by manipulating extracellular microenvironment in guiding cardiac differentiation of the pluripotent stem cells.
Collapse
Affiliation(s)
- Nan Cao
- Key Laboratory of Stem Cell Biology, Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) & Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Chemical compound 31002 stimulates cardiomyogenic differentiation of embryonic stem cells. Lab Anim Res 2011; 27:205-12. [PMID: 21998609 PMCID: PMC3188727 DOI: 10.5625/lar.2011.27.3.205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 08/27/2011] [Accepted: 08/28/2011] [Indexed: 11/21/2022] Open
Abstract
Embryonic stem cells (ESCs) are an emerging source for cell-based therapies aimed at repairing damaged organ tissues; however, the efficiency of directed differentiation is low and refinement of differentiation protocols is hampered by incomplete understanding of the mechanisms involved in this process. To find new compounds which can improve the efficiency of directed differentiation of ESCs to cardiomyocytes, we screened several thousand chemical compounds and identified a promising group. All of the compounds found have a common structure of 1H-pyrrole,2,2'-(phenylmethylene)bis. Here we report the potential mechanism of action for 31002 which showed the strongest activity among the compounds selected. In the presence of 31002, 15 times more cardiomyocytes differentiated from ESCs, i.e., 3.5% to 52% of total differentiated cells. Moreover, the cardiomyocytes showed functional characteristics including rhythmic beating and marker gene expression. 31002 inhibited the down-regulation of genes related to the three germ layers in the late stage of ESCs differentiation, implying that 31002 supports a continuous fate commitment of undifferentiated ESCs to the cardiac lineage by prolonging the three germ layer stages. Therefore, compounds in this group, including 31002, might be useful as directed cardiomyogenic differentiation-inducers to produce cells for use in cell therapy aimed at restoring damaged heart tissue.
Collapse
|