1
|
Watanabe T, Kawamura T, Harada A, Taira M, Yoshioka D, Shimamura K, Watabe T, Shimosegawa E, Ueno T, Miyagawa S. Human induced pluripotent stem cell-derived cardiomyocyte patches ameliorate right ventricular function in a rat pressure-overloaded right ventricle model. J Artif Organs 2025; 28:234-243. [PMID: 39638947 PMCID: PMC12078445 DOI: 10.1007/s10047-024-01479-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 10/21/2024] [Indexed: 12/07/2024]
Abstract
Right ventricular (RV) failure following surgical repair of congenital heart disease affects survival. Human induced pluripotent stem cell-derived cardiomyocyte (hiPS-CM) sheet transplantation ameliorated left ventricular dysfunction in preclinical studies, indicating its efficacy in RV failure in congenital heart disease. This study aimed to evaluate whether hiPS-CMs could improve RV function in rats with pressure-overloaded RV failure. F344/NJcl-rnu/rnu rats underwent pulmonary artery banding (PAB) via left thoracotomy. Four weeks after PAB, hiPS-CM patch transplantation to the RV was performed in the hiPS-CM group (n = 33), and a sham operation was performed in the sham group (n = 18). We evaluated cardiac catheterization, positron emission tomography data, and pathological results 8 weeks following PAB. RV end-diastolic pressure, the time constant of isovolumic relaxation, and end-diastolic pressure-volume relation were significantly ameliorated in the hiPS-CM group compared with in the sham group. Picrosirius red staining revealed that anti-fibrotic effects were significantly higher in the hiPS-CM group than in the sham group. Von Willebrand factor staining revealed significantly higher myocardial capillary vascular density in the hiPS-CM group than in the sham group. hiPS-CMs were detected in the epicardium 4 weeks after hiPS-CM sheet transplantation. The angiogenic gene expression in the myocardium was significantly higher in the hiPS-CM group than in the sham group. Overall, in rats with pressure-overloaded RV failure, hiPS-CM patch transplantation could improve diastolic function, suppress ventricular fibrosis, and increase capillary density, suggesting that it is a promising treatment for RV failure.
Collapse
Affiliation(s)
- Takuji Watanabe
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Masaki Taira
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Daisuke Yoshioka
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Kazuo Shimamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Eku Shimosegawa
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takayoshi Ueno
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2, Yamada-Oka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
2
|
Ghodrat S, Hoseini SJ, Asadpour S, Nazarnezhad S, Alizadeh Eghtedar F, Kargozar S. Stem cell-based therapies for cardiac diseases: The critical role of angiogenic exosomes. Biofactors 2021; 47:270-291. [PMID: 33606893 DOI: 10.1002/biof.1717] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/25/2021] [Indexed: 12/26/2022]
Abstract
Finding effective treatments for cardiac diseases is among the hottest subjects in medicine; cell-based therapies have brought great promises for managing a broad range of life-threatening heart complications such as myocardial infarction. After clarifying the critical role of angiogenesis in tissue repair and regeneration, various stem/progenitor cell were utilized to accelerate the healing of injured cardiac tissue. Embryonic, fetal, adult, and induced pluripotent stem cells have shown the appropriate proangiogenic potential for tissue repair strategies. The capability of stem cells for differentiating into endothelial lineages was initially introduced as the primary mechanism involved in improving angiogenesis and accelerated heart tissue repair. However, recent studies have demonstrated the leading role of paracrine factors secreted by stem cells in advancing neo-vessel formation. Genetically modified stem cells are also being applied for promoting angiogenesis regarding their ability to considerably overexpress and secrete angiogenic bioactive molecules. Yet, conducting further research seems necessary to precisely identify molecular mechanisms behind the proangiogenic potential of stem cells, including the signaling pathways and regulatory molecules such as microRNAs. In conclusion, stem cells' pivotal roles in promoting angiogenesis and consequent improved cardiac healing and remodeling processes should not be ignored, especially in the case of stem cell-derived extracellular vesicles.
Collapse
Affiliation(s)
- Sara Ghodrat
- Department of Nutrition, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Javad Hoseini
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shiva Asadpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fariba Alizadeh Eghtedar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
3
|
Mazzola M, Di Pasquale E. Toward Cardiac Regeneration: Combination of Pluripotent Stem Cell-Based Therapies and Bioengineering Strategies. Front Bioeng Biotechnol 2020; 8:455. [PMID: 32528940 PMCID: PMC7266938 DOI: 10.3389/fbioe.2020.00455] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases represent the major cause of morbidity and mortality worldwide. Multiple studies have been conducted so far in order to develop treatments able to prevent the progression of these pathologies. Despite progress made in the last decade, current therapies are still hampered by poor translation into actual clinical applications. The major drawback of such strategies is represented by the limited regenerative capacity of the cardiac tissue. Indeed, after an ischaemic insult, the formation of fibrotic scar takes place, interfering with mechanical and electrical functions of the heart. Hence, the ability of the heart to recover after ischaemic injury depends on several molecular and cellular pathways, and the imbalance between them results into adverse remodeling, culminating in heart failure. In this complex scenario, a new chapter of regenerative medicine has been opened over the past 20 years with the discovery of induced pluripotent stem cells (iPSCs). These cells share the same characteristic of embryonic stem cells (ESCs), but are generated from patient-specific somatic cells, overcoming the ethical limitations related to ESC use and providing an autologous source of human cells. Similarly to ESCs, iPSCs are able to efficiently differentiate into cardiomyocytes (CMs), and thus hold a real regenerative potential for future clinical applications. However, cell-based therapies are subjected to poor grafting and may cause adverse effects in the failing heart. Thus, over the last years, bioengineering technologies focused their attention on the improvement of both survival and functionality of iPSC-derived CMs. The combination of these two fields of study has burst the development of cell-based three-dimensional (3D) structures and organoids which mimic, more realistically, the in vivo cell behavior. Toward the same path, the possibility to directly induce conversion of fibroblasts into CMs has recently emerged as a promising area for in situ cardiac regeneration. In this review we provide an up-to-date overview of the latest advancements in the application of pluripotent stem cells and tissue-engineering for therapeutically relevant cardiac regenerative approaches, aiming to highlight outcomes, limitations and future perspectives for their clinical translation.
Collapse
Affiliation(s)
- Marta Mazzola
- Stem Cell Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Elisa Di Pasquale
- Stem Cell Unit, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Institute of Genetic and Biomedical Research (IRGB) - UOS of Milan, National Research Council (CNR), Milan, Italy
| |
Collapse
|
4
|
Albertario A, Swim MM, Ahmed EM, Iacobazzi D, Yeong M, Madeddu P, Ghorbel MT, Caputo M. Successful Reconstruction of the Right Ventricular Outflow Tract by Implantation of Thymus Stem Cell Engineered Graft in Growing Swine. JACC Basic Transl Sci 2019; 4:364-384. [PMID: 31312760 PMCID: PMC6609916 DOI: 10.1016/j.jacbts.2019.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/29/2019] [Accepted: 02/02/2019] [Indexed: 11/29/2022]
Abstract
T-MSCs were isolated from the thymus gland of new born pigs, expanded, characterized and seeded onto a commercially available scaffold. The seeded-grafts were cultured within a bioreactor and then used to reconstruct the RVOT of a growing swine model. Pigs were followed up for 4.5 months; then scanned with a cardiac magnetic resonance and terminated to harvest the implants. By comparing the outcome of the seeded-grafts to the unseeded-ones used as control, we observed a reduced fibrosis and an improved RVOT strain, cardiac remodeling and endothelialization.
Graft cellularization holds great promise in overcoming the limitations associated with prosthetic materials currently used in corrective cardiac surgery. In this study, the authors evaluated the advantages of graft cellularization for right ventricular outflow tract reconstruction in a novel porcine model. After 4.5 months from implantation, improved myocardial strain, better endothelialization and cardiomyocyte incorporation, and reduced fibrosis were observed in the cellularized grafts compared with the acellular grafts. To the authors’ knowledge, this is the first demonstration of successful right ventricular outflow tract correction using bioengineered grafts in a large animal model.
Collapse
Key Words
- CM, cardiomyocyte
- Cx-43, connexin-43
- DMEM, Dulbecco’s modified Eagle’s medium
- EC, endothelial cell
- FBS, fetal bovine serum
- IL, interleukin
- IsoB4, isolectin B4
- MSC, mesenchymal stem cell
- PBS, phosphate-buffered saline
- PS, penicillin/streptomycin
- RT, room temperature
- RV, right ventricular
- RVOT, right ventricular outflow tract
- RVOT-MS, fractional area of change in the right ventricular outflow tract
- SIS-ECM, small intestinal submucosa–derived extracellular matrix
- T-MSC, thymus-derived mesenchymal stem cell
- VMSC, vascular smooth muscle cell
- cMYH, cardiac myosin heavy chain
- congenital heart disease
- reconstruction
- right ventricular outflow swine model
- tissue engineering
- tract stem cells
Collapse
Affiliation(s)
- Ambra Albertario
- University of Bristol, Bristol Heart Institute, Bristol, United Kingdom
| | - Megan M Swim
- University of Bristol, Bristol Heart Institute, Bristol, United Kingdom
| | | | - Dominga Iacobazzi
- University of Bristol, Bristol Heart Institute, Bristol, United Kingdom
| | - Michael Yeong
- University of Bristol, Bristol Heart Institute, Bristol, United Kingdom
| | - Paolo Madeddu
- University of Bristol, Bristol Heart Institute, Bristol, United Kingdom
| | - Mohamed T Ghorbel
- University of Bristol, Bristol Heart Institute, Bristol, United Kingdom
| | - Massimo Caputo
- University of Bristol, Bristol Heart Institute, Bristol, United Kingdom
| |
Collapse
|
5
|
Jin M, Yin Z, Wei K, Xie Y, Bai X, Fu B, Feng Z, Li Q, Chen X. Metanephric mesenchyme-derived Foxd1 + mesangial precursor cells alleviate mesangial proliferative glomerulonephritis. J Mol Med (Berl) 2019; 97:553-561. [PMID: 30810761 DOI: 10.1007/s00109-019-01749-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/16/2019] [Accepted: 01/23/2019] [Indexed: 01/04/2023]
Abstract
Mesangial proliferative glomerulonephritis (MsPGN) is a glomerular disease characterized by the proliferation of mesangial cells and the accumulation of mesangial matrix. No effective treatment is currently able to stop or reverse the disease process. Here, we isolated metanephric mesenchyme-derived Foxd1+ mesangial precursor cells from E13.5 Foxd1Cre; DTRflox double transgenic embryo mice. The Foxd1+ cells showed the cell-specific expression of high levels of Foxd1 without Six2 and manifested specific cell surface markers of mesenchymal stem cells while retaining their differentiation potential. Next, an anti-Thy1 MsPGN rat model was established, and the Foxd1+ cells were injected into the tail veins 24 h later. We found that the Foxd1+ cells could improve the pathological changes to the kidney and significantly reduce proteinuria by inhibiting the sonic hedgehog pathway. Moreover, the Foxd1+ cells could inhibit PDGF-BB-induced activation of mesangial cells by secreting multiple cytokines, including hepatocyte growth factor. Our study uncovered the novel function of Foxd1+ mesangial precursor cells in repairing the damaged mesangium, stabilizing the structure and function of mesangial cells, and restoring their therapeutic effect on the MsPGN. KEY MESSAGES: It is viable to isolate highly purified metanephric mesenchyme-derived Foxd1+ mesangial precursor cells using transgenic mice. Foxd1+ cells could alleviate experimental mesangial proliferative glomerulonephritis and PDGF-induced mesangial cell proliferation through a variety of mechanisms including inhibiting the sonic hedgehog pathway and secreting multiple cytokines. As the progenitor cells of mesangial cells, Foxd1+ cells could stabilize the structure and function of mesangial cells that had undergone pathological changes.
Collapse
Affiliation(s)
- Meiling Jin
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China
- Department of Nephrology, Beijing Chao-Yang Hospital, Beijing, China
- Medical College, Nankai University, Tianjin, 300071, China
| | - Zhong Yin
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China
| | - Kai Wei
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China
- Medical College, Nankai University, Tianjin, 300071, China
| | - Yuansheng Xie
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China
| | - Xueyuan Bai
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China
| | - Bo Fu
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China
| | - Zhe Feng
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China
| | - Qinggang Li
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China.
| | - Xiangmei Chen
- Department of Nephrology, Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, 28th Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
6
|
Oxygen as a key regulator of cardiomyocyte proliferation: New results about cell culture conditions! BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118460. [PMID: 30885672 DOI: 10.1016/j.bbamcr.2019.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/21/2019] [Accepted: 03/13/2019] [Indexed: 01/16/2023]
Abstract
The goal of the new therapeutically strategies aimed to treat cardiovascular diseases (CVDs) is to enhance the natural ability of the heart to regenerate. This represents a great challenge for the coming years as all the mechanisms underlying the replacement of dying cells by functional cells of the same type are not completely elucidated. Among these, stimulating cardiomyocyte proliferation seems to be crucial for the restoration of normal cardiac function after CVDs. In this review, we summarized the recent advances about the modulation of cardiomyocyte proliferation in physiological (during ageing) and pathological conditions. We highlighted the role of oxygen and we presented new results demonstrating that performing neonatal cardiomyocyte cell cultures in "normoxic" oxygen conditions (i.e. 3% oxygen) increases their proliferation rate, when compared to "hyperoxic" conventional conditions (i.e. 20% oxygen). Thus, oxygen concentration seems to be a key factor in the control of cardiomyocyte proliferation.
Collapse
|
7
|
Swim MM, Albertario A, Iacobazzi D, Caputo M, Ghorbel MT. Amnion-Based Scaffold with Enhanced Strength and Biocompatibility for In Vivo Vascular Repair. Tissue Eng Part A 2018; 25:603-619. [PMID: 30284966 DOI: 10.1089/ten.tea.2018.0175] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
IMPACT STATEMENT This study aimed at developing an amnion-based scaffold suitable for vascular tissue engineering applications and in vivo usage. We successfully produced a multilayered scaffold with improved biomechanical properties and biocompatibility for in vivo vascular implantation. Our approach not only offers an allogeneic "off-the-shelf" solution for clinical use but also it provides the possibility of personalized medicine using a patient's own amnion and stem cells for the production of tissue engineered grafts for reconstructive heart surgery.
Collapse
Affiliation(s)
- Megan M Swim
- Bristol Heart Institute, Bristol Medical School, University of Bristol , Bristol, United Kingdom
| | - Ambra Albertario
- Bristol Heart Institute, Bristol Medical School, University of Bristol , Bristol, United Kingdom
| | - Dominga Iacobazzi
- Bristol Heart Institute, Bristol Medical School, University of Bristol , Bristol, United Kingdom
| | - Massimo Caputo
- Bristol Heart Institute, Bristol Medical School, University of Bristol , Bristol, United Kingdom
| | - Mohamed T Ghorbel
- Bristol Heart Institute, Bristol Medical School, University of Bristol , Bristol, United Kingdom
| |
Collapse
|
8
|
Sugiura T, Hibino N, Breuer CK, Shinoka T. Tissue-engineered cardiac patch seeded with human induced pluripotent stem cell derived cardiomyocytes promoted the regeneration of host cardiomyocytes in a rat model. J Cardiothorac Surg 2016; 11:163. [PMID: 27906085 PMCID: PMC5131419 DOI: 10.1186/s13019-016-0559-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 11/25/2016] [Indexed: 01/15/2023] Open
Abstract
Background Thousands of babies are born with congenital heart defects that require surgical repair involving a prosthetic implant. Lack of growth in prosthetic grafts is especially detrimental in pediatric surgery. Cell seeded biodegradable tissue engineered grafts are a novel solution to this problem. The purpose of the present study is to evaluate the feasibility of seeding human induced pluripotent stem cell derived cardiomyocytes (hiPS-CMs) onto a biodegradable cardiac patch. Methods The hiPS-CMs were cultured on a biodegradable patch composed of a polyglycolic acid (PGA) and a 50:50 poly (l-lactic-co-ε-caprolactone) copolymer (PLCL) for 1 week. Male athymic rats were randomly divided into 2 groups of 10 animals each: 1. hiPS-CM seeded group, and 2. Unseeded group. After culture, the cardiac patch was implanted to repair a defect with a diameter of 2 mm created in the right ventricular outflow tract (RVOT) wall. Hearts were explanted at 4 (n = 2), 8 (n = 2), and 16 (n = 6) weeks after patch implantation. Explanted patches were assessed immunohistochemically. Results Seeded patch explants did not stain positive for α-actinin (marker of cardiomyocytes) at the 4 week time point, suggesting that the cultured hiPS-CMs evacuated the patch in the early phase of tissue remodeling. However, after 16 weeks implantation, the area fraction of positively stained α-actinin cells was significantly higher in the seeded group than in the unseeded group (Seeded group: 6.1 ± 2.8% vs. Unseeded group: 0.95 ± 0.50%, p = 0.004), suggesting cell seeding promoted regenerative proliferation of host cardiomyocytes. Conclusions Seeded hiPS-CMs were not present in the patch after 4 weeks. However, we surmise that they influenced the regeneration of host cardiomyocytes via a paracrine mechanism. Tissue-engineered hiPS-CMs seeded cardiac patches warrant further investigation for use in the repair of congenital heart diseases.
Collapse
Affiliation(s)
- Tadahisa Sugiura
- Tissue Engineering Program and Surgical Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Narutoshi Hibino
- Tissue Engineering Program and Surgical Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Christopher K Breuer
- Tissue Engineering Program and Surgical Research, Nationwide Children's Hospital, Columbus, OH, USA
| | - Toshiharu Shinoka
- Tissue Engineering Program and Surgical Research, Nationwide Children's Hospital, Columbus, OH, USA. .,Department of Cardiothoracic Surgery, The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA. .,Cardiovascular Tissue Engineering Program, Department of Cardiothoracic Surgery, The Heart Center, Nationwide Children's Hospital, 700 Children's Drive, T2294, Columbus, OH, 43205, USA.
| |
Collapse
|
9
|
Peng SY, Yang YS, Chou CJ, Lin KY, Wu SC. Differentiation of Enhanced Green Fluorescent Protein-Labeled Mouse Amniotic Fluid-Derived Stem Cells into Cardiomyocyte-Like Beating Cells. ACTA CARDIOLOGICA SINICA 2016; 31:209-14. [PMID: 27122872 DOI: 10.6515/acs20141027a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Amniotic fluid-derived stem cells (AFSCs) possess optimal differentiation potential and are a promising resource for cell therapy and tissue engineering. Mouse is a good model to be studied for pre-clinical research. METHODS In this study, we successfully established enhanced green fluorescent protein mouse-derived amniotic fluid stem cells (EGFP-mAFSCs) and investigated whether EGFP-mAFSCs possess the ability to differentiate into cardiomyocytes by in vitro culture. We evaluated stem-cell differentiation using immunofluorescence. RESULTS This study showed that EGFP-mAFSCs can give rise to spontaneously beating cardiomyocyte-like cells expressing the specific markers c-kit, myosin heavy chain, and cardiac troponin I. CONCLUSIONS We demonstrated that mAFSCs have the in vitro propensity to acquire a cardiomyogenic phenotype and to a certain extent cardiomyocytes; however the process efficiency which gives rise to cardiomyocyte-like cells remains quite low (2 out of 10 were found). KEY WORDS Amniotic fluid; Cardiomyocytes; In vitro differentiation; Stem cells.
Collapse
Affiliation(s)
- Shao-Yu Peng
- Institute of Biotechnology, National Taiwan University
| | - Yu-Sheng Yang
- Department of Orthopaedics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Jen Chou
- Institute of Biotechnology, National Taiwan University
| | - Kun-Yi Lin
- Institute of Biotechnology, National Taiwan University; ; Department of Orthopaedics, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shinn-Chih Wu
- Institute of Biotechnology, National Taiwan University
| |
Collapse
|
10
|
Creatine Enhances Transdifferentiation of Bone Marrow Stromal Cell-Derived Neural Stem Cell Into GABAergic Neuron-Like Cells Characterized With Differential Gene Expression. Mol Neurobiol 2016; 54:1978-1991. [PMID: 26910814 DOI: 10.1007/s12035-016-9782-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 02/08/2016] [Indexed: 12/16/2022]
Abstract
Creatine was reported to induce bone marrow stromal cells (BMSC) into GABAergic neuron-like cells (GNLC). In a previous study, creatine was used as a single inducer for BMSC into GNLC with low yield. In this study, BMSC-derived neurospheres (NS) have been used in generating GABAergic phenotype. The BMSC were isolated from adult rats and used in generating neurospheres and used for producing neural stem cells (NSC). A combination of all-trans-retinoic acid (RA), the ciliary neurotrophic factor (CNTF), and creatine was used in order to improve the yield of GNLC. We also used other protocols for the transdifferentiation including RA alone; RA and creatine; RA and CNTF; and RA, CNTF, and creatine. The BMSC, NSC, and GNLC were characterized by specific markers. The activity of the GNLC was evaluated using FM1-43. The isolated BMSC expressed Oct4, fibronectin, and CD44. The NS were immunoreactive to nestin and SOX2, the NSC were immunoreactive to nestin, NF68 and NF160, while the GNLC were immunoreactive to GAD1/2, VGAT, GABA, and synaptophysin. Oct4 and c-MYC, pluripotency genes, were expressed in the BMSC, while SOX2 and c-MYC were expressed in the NSC. The activity of GNLC indicates that the synaptic vesicles were released upon stimulation. The conclusion is that the combination of RA, CNTF, and creatine induced differentiation of neurosphere-derived NSC into GNLC within 1 week. This protocol gives higher yield than the other protocols used in this study. The mechanism of induction was clearly associated with several differential pluripotent genes.
Collapse
|
11
|
Cell Therapy in Ischemic Heart Disease: Interventions That Modulate Cardiac Regeneration. Stem Cells Int 2016; 2016:2171035. [PMID: 26880938 PMCID: PMC4736413 DOI: 10.1155/2016/2171035] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 10/26/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
The incidence of severe ischemic heart disease caused by coronary obstruction has progressively increased. Alternative forms of treatment have been studied in an attempt to regenerate myocardial tissue, induce angiogenesis, and improve clinical conditions. In this context, cell therapy has emerged as a promising alternative using cells with regenerative potential, focusing on the release of paracrine and autocrine factors that contribute to cell survival, angiogenesis, and tissue remodeling. Evidence of the safety, feasibility, and potential effectiveness of cell therapy has emerged from several clinical trials using different lineages of adult stem cells. The clinical benefit, however, is not yet well established. In this review, we discuss the therapeutic potential of cell therapy in terms of regenerative and angiogenic capacity after myocardial ischemia. In addition, we addressed nonpharmacological interventions that may influence this therapeutic practice, such as diet and physical training. This review brings together current data on pharmacological and nonpharmacological approaches to improve cell homing and cardiac repair.
Collapse
|
12
|
Valente M, Araújo A, Esteves T, Laundos TL, Freire AG, Quelhas P, Pinto-do-Ó P, Nascimento DS. Optimized Heart Sampling and Systematic Evaluation of Cardiac Therapies in Mouse Models of Ischemic Injury: Assessment of Cardiac Remodeling and Semi-Automated Quantification of Myocardial Infarct Size. ACTA ACUST UNITED AC 2015; 5:359-391. [PMID: 26629776 DOI: 10.1002/9780470942390.mo140293] [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] [Indexed: 01/15/2023]
Abstract
Cardiac therapies are commonly tested preclinically in small-animal models of myocardial infarction. Following functional evaluation, post-mortem histological analysis is essential to assess morphological and molecular alterations underlying the effectiveness of treatment. However, non-methodical and inadequate sampling of the left ventricle often leads to misinterpretations and variability, making direct study comparisons unreliable. Protocols are provided for representative sampling of the ischemic mouse heart followed by morphometric analysis of the left ventricle. Extending the use of this sampling to other types of in situ analysis is also illustrated through the assessment of neovascularization and cellular engraftment in a cell-based therapy setting. This is of interest to the general cardiovascular research community as it details methods for standardization and simplification of histo-morphometric evaluation of emergent heart therapies. © 2015 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Mariana Valente
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Unit for Lymphopoiesis, Immunology Department, INSERM U668, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur, Paris, France
| | - Ana Araújo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Tiago Esteves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,FEUP - Faculdade de Engenharia da Universidade do Porto, Universidade do Porto, Porto, Portugal
| | - Tiago L Laundos
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ana G Freire
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,FEUP - Faculdade de Engenharia da Universidade do Porto, Universidade do Porto, Porto, Portugal.,Department of Developmental and Regenerative Biology and The Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pedro Quelhas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Perpétua Pinto-do-Ó
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Unit for Lymphopoiesis, Immunology Department, INSERM U668, University Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Institut Pasteur, Paris, France
| | - Diana S Nascimento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
13
|
Yang C, Al-Aama J, Stojkovic M, Keavney B, Trafford A, Lako M, Armstrong L. Concise Review: Cardiac Disease Modeling Using Induced Pluripotent Stem Cells. Stem Cells 2015; 33:2643-51. [PMID: 26033645 DOI: 10.1002/stem.2070] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 05/07/2015] [Accepted: 05/12/2015] [Indexed: 12/16/2022]
Abstract
Genetic cardiac diseases are major causes of morbidity and mortality. Although animal models have been created to provide some useful insights into the pathogenesis of genetic cardiac diseases, the significant species differences and the lack of genetic information for complex genetic diseases markedly attenuate the application values of such data. Generation of induced pluripotent stem cells (iPSCs) from patient-specific specimens and subsequent derivation of cardiomyocytes offer novel avenues to study the mechanisms underlying cardiac diseases, to identify new causative genes, and to provide insights into the disease aetiology. In recent years, the list of human iPSC-based models for genetic cardiac diseases has been expanding rapidly, although there are still remaining concerns on the level of functionality of iPSC-derived cardiomyocytes and their ability to be used for modeling complex cardiac diseases in adults. This review focuses on the development of cardiomyocyte induction from pluripotent stem cells, the recent progress in heart disease modeling using iPSC-derived cardiomyocytes, and the challenges associated with understanding complex genetic diseases. To address these issues, we examine the similarity between iPSC-derived cardiomyocytes and their ex vivo counterparts and how this relates to the method used to differentiate the pluripotent stem cells into a cardiomyocyte phenotype. We progress to examine categories of congenital cardiac abnormalities that are suitable for iPSC-based disease modeling.
Collapse
Affiliation(s)
- Chunbo Yang
- Institute of Genetic Medicine, Newcastle University, The International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Jumana Al-Aama
- Princess Al Jawhara Center of Excellence in Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Miodrag Stojkovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Bernard Keavney
- Institute of Cardiovascular Sciences Core Technology, Manchester University, Manchester, United Kingdom
| | - Andrew Trafford
- Institute of Cardiovascular Sciences Core Technology, Manchester University, Manchester, United Kingdom
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, The International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Lyle Armstrong
- Institute of Genetic Medicine, Newcastle University, The International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
14
|
Achmad N, Götte M. Characteristics and Therapeutic Potential of Menstrual Blood-Derived Stem Cells. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
15
|
N-glycans: phenotypic homology and structural differences between myocardial cells and induced pluripotent stem cell-derived cardiomyocytes. PLoS One 2014; 9:e111064. [PMID: 25357199 PMCID: PMC4214687 DOI: 10.1371/journal.pone.0111064] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 09/19/2014] [Indexed: 11/19/2022] Open
Abstract
Cell surface glycans vary widely, depending on cell properties. We hypothesized that glycan expression on induced pluripotent stem cells (iPSCs) might change during cardiomyogenic differentiation toward the myocardial phenotype. N-glycans were isolated from iPSCs, iPSC-derived cardiomyocytes (iPSC-CM), and original C57BL/6 mouse myocardium (Heart). Their structures were analyzed by a mapping technique based on HPLC elution times and MALDI-TOF/MS spectra. Sixty-eight different N-glycans were isolated; the structures of 60 of these N-glycans were identified. The quantity of high-mannose type (immature) N-glycans on the iPSCs decreased with cardiomyogenic differentiation, but did not reach the low levels observed in the heart. We observed a similar reduction in neutral N-glycans and an increase in fucosylated or sialyl N-glycans. Some structural differences were detected between iPSC-CM and Heart. No N-glycolyl neuraminic acid (NeuGc) structures were detected in iPSC-CM, whereas the heart contained numerous NeuGc structures, corresponding to the expression of cytidine monophosphate-N-acetylneuraminic acid hydroxylase. Furthermore, several glycans containing Galα1-6 Gal, rarely identified in the other cells, were detected in the iPSC-CM. The expression of N-glycan on murine iPSCs changed toward the myocardial phenotype during cardiomyogenic differentiation, leaving the structural differences of NeuGc content or Galα1-6 Gal structures. Further studies will be warranted to reveal the meaning of the difference of N-glycans between the iPSC-CM and the myocardium.
Collapse
|
16
|
Regulation of Cardiac Cell Fate by microRNAs: Implications for Heart Regeneration. Cells 2014; 3:996-1026. [PMID: 25358052 PMCID: PMC4276912 DOI: 10.3390/cells3040996] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/29/2014] [Accepted: 10/10/2014] [Indexed: 01/06/2023] Open
Abstract
microRNAs are post-transcriptional regulators of gene expression that have been shown to be central players in the establishment of cellular programs, often acting as switches that control the choice between proliferation and differentiation during development and in adult tissues. The heart develops from two small patches of cells in the mesoderm, the heart fields, which originate the different cardiac cell types, including cardiomyocytes, vascular smooth muscle and endothelial cells. These progenitors proliferate and differentiate to establish a highly connected three-dimensional structure, involving a robust succession of gene expression programs strongly influenced by microRNAs. Although the mammalian heart has conventionally been viewed as a post-mitotic organ, cardiac cells have recently been shown to display some regenerative potential, which is nonetheless insufficient to regenerate heart lesions, in contrast with other vertebrates like the zebrafish. Both the proliferation of adult cardiac stem cells and the ability of cardiomyocytes to re-enter the cell cycle have been proposed to sustain these regenerative processes. Here we review the role of microRNAs in the control of stem cell and cardiomyocyte dependent cardiac regeneration processes, and discuss potential applications for the treatment of cardiac injury.
Collapse
|
17
|
Valente M, Nascimento DS, Cumano A, Pinto-do-Ó P. Sca-1+ cardiac progenitor cells and heart-making: a critical synopsis. Stem Cells Dev 2014; 23:2263-73. [PMID: 24926741 DOI: 10.1089/scd.2014.0197] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The identification, in the adult, of cardiomyocyte turnover events and of cardiac progenitor cells (CPCs) has revolutionized the field of cardiovascular medicine. However, the low rate of CPCs differentiation events reported both in vitro and in vivo, even after injury, raised concerns on the biological significance of these subsets. In this Comprehensive Review, we discuss the current understanding of cardiac Lin(-)Sca-1(+) cells in light of what is also known for cellular compartments with similar phenotypes in other organs. The Lin(-)Sca-1(+) heart subset is heterogeneous and displays a mesenchymal profile, characterized by a limited ability to generate cardiomyocytes in vitro and in vivo, even after injury. There is no evidence for Sca-1 expression in embryonic cardiovascular progenitors. In other organs, Sca-1 expression is mainly observed on mesoderm-derived cells, although it is not restricted to stem/progenitor cell populations. It is urgent to determine, at a single cell level, to which extent cardiac Lin(-)Sca-1(+) cells overlap with the fibroblast compartment.
Collapse
Affiliation(s)
- Mariana Valente
- 1 Stem-Cell Microenvironments in Repair/Regeneration Team, Microenvironments for NewTherapies Group, INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto , Porto, Portugal
| | | | | | | |
Collapse
|
18
|
Xie HL, Zhang Y, Huang YZ, Li S, Wu CG, Jiao XF, Tan MY, Huang YC, Deng L. Regulation of high mobility group box 1 and hypoxia in the migration of mesenchymal stem cells. Cell Biol Int 2014; 38:892-897. [PMID: 24687575 DOI: 10.1002/cbin.10279] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 02/20/2014] [Indexed: 02/05/2023]
Abstract
Mesenchymal stem cells (MSCs) have been increasingly offered for tissue regeneration with the premise that they can survive and thrive amidst the microenvironment of injured or degenerate tissues. The role of high mobility group box 1 (HMGB1) and hypoxia in the proliferation and migration of rat bone marrow MSCs (rBM-MSCs) has been investigated. First, the effect of HMGB1 on the proliferation of rBM-MSCs was determined. Second, to evaluate the regulation of hypoxia and HMGB1 in the migration of rBM-MSCs, cells in the wound healing model were exposed to four conditions: normoxia (20% O2) and complete medium, normoxia and HMGB1, hypoxia (1% O2) and complete medium, hypoxia and HMGB1. RT-PCR and Western blotting were used to measure the expression of migration-related genes and proteins. HMGB1 inhibited the proliferation of rBM-MSCs; HMGB1 alone or together with hypoxia and promoted the migration of MSCs and upregulated the expression of HIF-1α and SDF-1. These results demonstrated that HMGB1 arrested the proliferation of rBM-MSCs, but enhanced the migration of rBM-MSCs which could be further improved by hypoxia. This study strengthens current understanding of the interaction between MSCs and the microenvironment of damaged tissues.
Collapse
Affiliation(s)
- Hong-Lei Xie
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kim S, Chaudhry A, Lee I, Frank JA. Effects of long-term hypoxia and pro-survival cocktail in bone marrow-derived stromal cell survival. Stem Cells Dev 2014; 23:530-40. [PMID: 24147599 PMCID: PMC3928716 DOI: 10.1089/scd.2013.0297] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 10/22/2013] [Indexed: 12/31/2022] Open
Abstract
The goal of this study was to determine whether a pro-survival cocktail (PSC, consisting of IGF-1, Bcl-XL, and Caspase-I Inhibitor) and long-term hypoxia (LTH) enhance survival and functional properties of bone marrow-derived stromal stem cells (BMSCs), in response to stress conditions. PSC-treated cells retained BMSC surface markers and protected cells from apoptosis under serum starvation and ischemic (1% O2 and 100 μM H2O2) conditions. LTH promoted osteogenesis, while suppressing adipogenesis. LTH alone did not result in an improvement in the apoptosis rate; however, PSC conferred significant protection regardless of the oxygenation status. One of the possible mechanisms of PSC protection was due to the elevated phospho-AKT in treated groups. PSC treatment or LTH did not alter migration toward stem cell-derived factor-1 alpha (SDF-1α) or fetal bovine serum, nor did they enhance cell motility during wound healing. There was no difference in the secreted cytokine profiles of BMSCs treated with PSC after stress when grown in normoxic or LTH. However, LTH did upregulate the vascular endothelial growth factor, hepatocyte growth factor, and SDF-1α, while it downregulated other anti- and proinflammatory cytokines and chemokines. We also observed a high degree of interdonor BMSC variability in response to pretreatment with PSC and LTH, confounding the functional results, underscoring the observation that not all donor-derived BMSCs will respond similarly.
Collapse
Affiliation(s)
- Saejeong Kim
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Aneeka Chaudhry
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Inae Lee
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Joseph A. Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
20
|
|
21
|
Leonardini A, Avogaro A. Abnormalities of the cardiac stem and progenitor cell compartment in experimental and human diabetes. Arch Physiol Biochem 2013; 119:179-87. [PMID: 23772700 DOI: 10.3109/13813455.2013.798334] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Diabetic cardiomyopathy consists of a series of structural and functional changes. Accumulating evidence supports the concept that a "cardiac stem cell compartment disease" plays an important role in the pathophysiology of diabetic cardiomyopathy. In diabetic hearts, human cardiac stem/progenitor cells (CSPC) are reduced and manifest defective proliferative capacity. Hyperglycaemia, hyperlipidemia, inflammation, and the consequent oxidative stress are enhanced in diabetes: these conditions can induce defects in both growth and survival of these cells with an imbalance between cell death and cell replacement, thus favouring the onset of diabetic cardiomyopathy and its progression towards heart failure. The preservation of CSPC compartment can contribute to counteract the negative impact of diabetes on the myocardium. The recent studies summarized in this review have improved our understanding of the development and stem cell biology within the cardiovascular system. However, several issues remain unsolved before cell therapy can become a clinical therapeutically relevant strategy.
Collapse
Affiliation(s)
- Anna Leonardini
- Department of Emergency and Organ Transplantation - Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro , Bari , Italy and
| | | |
Collapse
|
22
|
Abstract
Traumatic brain injury (TBI) has been associated with various neurological disorders. However, the role of cerebrovascular dysfunction and its mechanisms associated with TBI are still not well understood. Inflammation is the main cause of vascular dysfunction. It affects properties of blood components and the vascular wall leading to changes in blood flow and in interaction of blood components and vascular endothelium exacerbating microcirculatory complications during inflammatory diseases. One of the markers of inflammation is a plasma adhesion protein, fibrinogen (Fg). At elevated levels, Fg can also cause inflammatory responses. One of the manifestations of inflammatory responses is an increase in microvascular permeability leading to accumulation of plasma proteins in the subendothelial matrix and causing vascular remodelling. This has a most devastating effect on cerebral circulation after TBI that is accompanied with an elevation of plasma level of Fg and with an increased cerebrovascular permeability in injury penumbra impairing the normal healing process. This study reviews cerebrovascular alterations after TBI, considers the consequences of increased blood-brain barrier permeability, defines the role of elevated level of Fg and discusses the potential mechanisms of its action leading to vascular dysfunction, which subsequently can cause impairment in neuronal function. Thus, possible mechanisms of vasculo-neuronal dysfunction after TBI are considered.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine , Louisville, KY , USA
| | | |
Collapse
|
23
|
Castells-Sala C, Sanchez B, Recha-Sancho L, Puig V, Bragos R, Semino CE. Influence of electrical stimulation on 3D-cultures of adipose tissue derived progenitor cells (ATDPCs) behavior. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2013; 2012:5658-61. [PMID: 23367213 DOI: 10.1109/embc.2012.6347278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Tissue engineering has a fundamental role in regenerative medicine. Still today, the major motivation for cardiac regeneration is to design a platform that enables the complete tissue structure and physiological function regeneration of injured myocardium areas. Although tissue engineering approaches have been generally developed for two-dimensional (2D) culture systems, three-dimensional (3D) systems are being spotlighted as the means to mimic better in vivo cellular conditions. This manuscript examines the influence of electrical stimulation on 3D cultures of adipose tissue-derived progenitor cells (ATDPCs). ATDPCs cells were encapsulated into a self-assembling peptide nanoscaffold (RAD16-I) and continuously electro stimulated during 14-20 days with 2-ms pulses of 50mV/cm at a frequency of 1 Hz. Good cellular network formation and construct diameter reduction was observed in electro stimulated samples. Importantly, the process of electro stimulation does not disrupt cell viability or connectivity. As a future outlook, differentiation studies to cardiomyocytes-like cells will be performed analyzing gene profile and protein expression.
Collapse
Affiliation(s)
- C Castells-Sala
- Tissue Engineering Laboratory, Bioengineering Department, Institut Quimic de Sarria, Universidad Ramon Llull, Barcelona, SPAIN.
| | | | | | | | | | | |
Collapse
|
24
|
Role of Prokineticin Receptor-1 in Epicardial Progenitor Cells. J Dev Biol 2013; 1:20-31. [DOI: 10.3390/jdb1010020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 06/05/2013] [Accepted: 06/08/2013] [Indexed: 11/16/2022] Open
Abstract
G protein-coupled receptors (GPCRs) form a large class of seven transmembrane (TM) domain receptors. The use of endogenous GPCR ligands to activate the stem cell maintenance or to direct cell differentiation would overcome many of the problems currently encountered in the use of stem cells, such as rapid in vitro differentiation and expansion or rejection in clinical applications. This review focuses on the definition of a new GPCR signaling pathway activated by peptide hormones, called “prokineticins”, in epicardium-derived cells (EPDCs). Signaling via prokineticin-2 and its receptor, PKR1, is required for cardiomyocyte survival during hypoxic stress. The binding of prokineticin-2 to PKR1 induces proliferation, migration and angiogenesis in endothelial cells. The expression of prokineticin and PKR1 increases during cardiac remodeling after myocardial infarction. Gain of function of PKR1 in the adult mouse heart revealed that cardiomyocyte-PKR1 signaling activates EPDCs in a paracrine fashion, thereby promoting de novo vasculogenesis. Transient PKR1 gene therapy after myocardial infarction in mice decreases mortality and improves heart function by promoting neovascularization, protecting cardiomyocytes and mobilizing WT1+ cells. Furthermore, PKR1 signaling promotes adult EPDC proliferation and differentiation to adopt endothelial and smooth muscle cell fate, for the induction of de novo vasculogenesis. PKR1 is expressed in the proepicardium and epicardial cells derived from mice kidneys. Loss of PKR1 causes deficits in EPDCs in the neonatal mice hearts and kidneys and impairs vascularization and heart and kidney function. Taken together, these data indicate a novel role for PKR1 in heart-kidney complex via EPDCs.
Collapse
|
25
|
Liu J, Wang Y, Du W, Liu W, Liu F, Zhang L, Zhang M, Hou M, Liu K, Zhang S, Yu B. Wnt1 inhibits hydrogen peroxide-induced apoptosis in mouse cardiac stem cells. PLoS One 2013; 8:e58883. [PMID: 23533594 PMCID: PMC3606408 DOI: 10.1371/journal.pone.0058883] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 02/07/2013] [Indexed: 01/04/2023] Open
Abstract
Background Because of their regenerative and paracrine abilities, cardiac stem cells (CSCs) are the most appropriate, optimal and promising candidates for the development of cardiac regenerative medicine strategies. However, native and exogenous CSCs in ischemic hearts are exposed to various pro-apoptotic or cytotoxic factors preventing their regenerative and paracrine abilities. Methods and Results We examined the effects of H2O2 on mouse CSCs (mCSCs), and observed that hydrogen peroxide (H2O2) treatment induces mCSCs apoptosis via the caspase 3 pathway, in a dose-dependent manner. We then examined the effects of Wnt1 over-expression on H2O2-induced apoptosis in mCSCs and observed that Wnt1 significantly decreased H2O2-induced apoptosis in mCSCs. On the other hand, inhibition of the canonical Wnt pathway by the secreted frizzled related protein 2 (SFRP2) or knockdown of β-catenin in mCSCs reduced cells resistance to H2O2-induced apoptosis, suggesting that Wnt1 predominantly prevents H2O2-induced apoptosis through the canonical Wnt pathway. Conclusions Our results provide the first evidences that Wnt1 plays an important role in CSCs’ defenses against H2O2-induced apoptosis through the canonical Wnt1/GSK3β/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Jingjin Liu
- Cardiology Department, Second Affiliated Hospital of Harbin Medical University, Harbin, Province Heilongjiang, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Zhang W, Yan H. Dysfunction of circulating endothelial progenitor cells in type 1 diabetic rats with diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2013; 251:1123-31. [PMID: 23385543 DOI: 10.1007/s00417-013-2267-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 12/27/2012] [Accepted: 01/15/2013] [Indexed: 12/27/2022] Open
Abstract
PURPOSE To investigate the role of endothelial progenitor cell (EPC) in the pathogenesis of diabetic retinopathy (DR) in streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM) rats. METHODS A total of 160 male Wistar rats (12 weeks old; 250-350 g) were randomly assigned into four groups (n = 40 for each), including control (group 1, no treatment), T1DM1 (group 2, 1 month after 50 mg/kg of STZ, single i.p.), T1DM3 (group 3, 3 months after 50 mg/kg of STZ, single i.p.), T1DM6 (group 4, 6 months after 50 mg/kg of STZ, single i.p.). Enumeration of circulating EPC from peripheral blood was measured by flow cytometry. EPC from bone marrow of rats was cultured in vitro to evaluate its function of proliferation, adhesion, and migration activities. Plasma levels of vascular endothelial growth factor (VEGF) and nitric oxide (NO) were measured by enzyme-linked immunosorbent assay (ELISA). Retinal sections were imaged by light microscopy and a transmission electron microscope (TEM). RESULTS The numbers of circulating EPC were significantly decreased in diabetic groups compared with the control group. Impaired proliferation, adhesion, and migratory activities of cultured EPC were observed in diabetic groups. There were significantly higher levels of plasma VEGF but lower levels of plasma NO in diabetic groups than those in non-diabetic controls. The significantly reduced thickness and obvious disorganized retinal cell layers were seen in T1DM DR rats. In the diabetic groups, we also found that T1DM rats developed telangiectatic vessels, vacuolar degeneration of ganglion cells, and thickened capillary basement membrane with capillary lumen stenosis in the retina. Significantly raised EPC numbers during DR formation and progression were also found. CONCLUSIONS The reduced numbers and impaired function of circulating EPC may contribute to the pathogenesis of DR in T1DM rats.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, No. 154, Anshan Road, Tianjin 300052, China
| | | |
Collapse
|
27
|
Kawamura M, Miyagawa S, Miki K, Saito A, Fukushima S, Higuchi T, Kawamura T, Kuratani T, Daimon T, Shimizu T, Okano T, Sawa Y. Feasibility, safety, and therapeutic efficacy of human induced pluripotent stem cell-derived cardiomyocyte sheets in a porcine ischemic cardiomyopathy model. Circulation 2012; 126:S29-37. [PMID: 22965990 DOI: 10.1161/circulationaha.111.084343] [Citation(s) in RCA: 345] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) are a promising source of cells for regenerating myocardium. However, several issues, especially the large-scale preparation of hiPS-CMs and elimination of undifferentiated iPS cells, must be resolved before hiPS cells can be used clinically. The cell-sheet technique is one of the useful methods for transplanting large numbers of cells. We hypothesized that hiPS-CM-sheet transplantation would be feasible, safe, and therapeutically effective for the treatment of ischemic cardiomyopathy. METHODS AND RESULTS Human iPS cells were established by infecting human dermal fibroblasts with a retrovirus carrying Oct3/4, Sox2, Klf4, and c-Myc. Cardiomyogenic differentiation was induced by WNT signaling molecules, yielding hiPS-CMs that were almost 90% positive for α-actinin, Nkx2.5, and cardiac troponin T. hiPS-CM sheets were created using thermoresponsive dishes and transplanted over the myocardial infarcts in a porcine model of ischemic cardiomyopathy induced by ameroid constriction of the left anterior descending coronary artery (n=6 for the iPS group receiving sheet transplantation and the sham-operated group; both groups received tacrolimus daily). Transplantation significantly improved cardiac performance and attenuated left ventricular remodeling. hiPS-CMs were detectable 8 weeks after transplantation, but very few survived long term. No teratoma formation was observed in animals that received hiPS-CM sheets. CONCLUSIONS The culture system used yields a large number of highly pure hiPS-CMs, and hiPS-CM sheets could improve cardiac function after ischemic cardiomyopathy. This newly developed culture system and the hiPS-CM sheets may provide a basis for the clinical use of hiPS cells in cardiac regeneration therapy.
Collapse
Affiliation(s)
- Masashi Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Li Y, Yang CM, Xi Y, Wu G, Shelat H, Gao S, Cheng J, Geng YJ. MicroRNA-1/133 targeted dysfunction of potassium channels KCNE1 and KCNQ1 in human cardiac progenitor cells with simulated hyperglycemia. Int J Cardiol 2012; 167:1076-8. [PMID: 23157812 DOI: 10.1016/j.ijcard.2012.10.060] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 10/28/2012] [Indexed: 10/27/2022]
|
29
|
Smart N, Dubé KN, Riley PR. Epicardial progenitor cells in cardiac regeneration and neovascularisation. Vascul Pharmacol 2012; 58:164-73. [PMID: 22902355 DOI: 10.1016/j.vph.2012.08.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 07/31/2012] [Accepted: 08/06/2012] [Indexed: 01/14/2023]
Abstract
While cardiovascular diseases remain the major worldwide cause of mortality and morbidity, there is an urgent need to tackle the clinical and economic burden of heart failure. Since the mammalian heart is unable to adequately regenerate beyond early postnatal stages, individuals surviving acute myocardial infarction are at risk of heart failure. Understanding the embryonic mechanisms of vasculogenesis and cardiogenesis, as well as the mechanisms retained for regeneration in species such as the zebrafish, will inform on strategies for human myocardial repair. Due to their fundamental role in heart development, epicardium-derived cells (EPDCs) have emerged as a population with potential to restore myocardium and coronary vasculature. The ability to revive ordinarily dormant EPDCs lies in the identification of key molecular cues used in the embryo to orchestrate cardiovascular development. One such stimulatory factor, Thymosin β4 (Tβ4), restores the quiescent adult epicardium to its pluripotent embryonic state. Tβ4 treatment of infarcted hearts induces dramatic EPDC proliferation and formation of a network of perfused, functional vessels to enhance blood flow to the ischaemic myocardium. Moreover, Tβ4 facilitates an epicardial contribution of mature de novo cardiomyocytes, structurally and functionally coupled with resident myocardium, which may contribute towards the functional improvement of Tβ4-treated hearts post-MI.
Collapse
Affiliation(s)
- Nicola Smart
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK.
| | | | | |
Collapse
|
30
|
Exposure to supernatants of macrophages that phagocytized dead mesenchymal stem cells improves hypoxic cardiomyocytes survival. Int J Cardiol 2012; 165:333-40. [PMID: 22475845 DOI: 10.1016/j.ijcard.2012.03.088] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 01/28/2012] [Accepted: 03/03/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To observe the impact of supernatants from macrophages that phagocytized dead MSCs (pMΦ) on the survival of hypoxic cardiomyocytes. METHODS MSCs were isolated from bone marrow of mice and dead MSCs were harvested after 6h hypoxia. Macrophages were obtained from thioglycolate-elicited peritoneal cavity. Macrophages and dead MSCs were co-cultured for 2 days in the presence or absence of LPS (1 μg/ml). Cardiomyocytes obtained from neonatal mice were exposed to various medium including supernatants from pMΦ. MTT cell proliferation assay and mitochondria membrane potential were used to evaluate the viability of cardiomyocytes. Cytokines and chemokines (TNF-α, IFN-γ, IL-6, IL-12, PGE2, VEGF-α, Ang-1, KGF, IGF-1, PDGF-BB, and EPO) in culture medium of macrophages, MSCs and pMΦ were detected by ELISA and Real-Time-PCR. RESULTS phagocytic activity of macrophages to dMSC was significantly enhanced by LPS. PGE2, VEGF-α, Ang-1, KGF, IGF-1, PDGF-BB, and EPO levels were significantly increased in supernatants of pMΦ. Exposure to supernatants of pMΦ significantly improved viability and survival time of hypoxic cardiomyocytes. CONCLUSION Exposure to supernatants of pMΦ significantly improved viability and survival time of hypoxic cardiomyocytes, which might be linked to increased cytokines and chemokines secretion by pMΦ.
Collapse
|
31
|
Pillekamp F, Haustein M, Khalil M, Emmelheinz M, Nazzal R, Adelmann R, Nguemo F, Rubenchyk O, Pfannkuche K, Matzkies M, Reppel M, Bloch W, Brockmeier K, Hescheler J. Contractile properties of early human embryonic stem cell-derived cardiomyocytes: beta-adrenergic stimulation induces positive chronotropy and lusitropy but not inotropy. Stem Cells Dev 2012; 21:2111-21. [PMID: 22268955 DOI: 10.1089/scd.2011.0312] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CMs) provide the unique opportunity to study the very early development of the human heart. The aim of this study was to investigate the effect of calcium and beta-adrenergic stimulation on the contractile properties of early hESC-CMs. Beating clusters containing hESC-CMs were co-cultured in vitro with noncontractile slices of neonatal murine ventricles. After 5-7 days, when beating clusters had integrated morphologically into the damaged tissue, isometric force measurements were performed during spontaneous beating as well as during electrical field stimulation. Spontaneous beating stopped when extracellular calcium ([Ca²⁺](ec)) was removed or after administration of the Ca²⁺ channel blocker nifedipine. During field stimulation at a constant rate, the developed force increased with incremental concentrations of [Ca²⁺](ec). During spontaneous beating, rising [Ca²⁺](ec) increased beating rate and developed force up to a [Ca²⁺](ec) of 2.5 mM. When [Ca²⁺](ec) was increased further, spontaneous beating rate decreased, whereas the developed force continued to increase. The beta-adrenergic agonist isoproterenol induced a dose-dependent increase of the frequency of spontaneous beating; however, it did not significantly change the developed force during spontaneous contractions or during electrical stimulation at a constant rate. Force developed by early hESC-CMs depends on [Ca²⁺](ec) and on the L-type Ca²⁺ channel. The lack of an inotropic reaction despite a pronounced chronotropic response after beta-adrenergic stimulation most likely indicates immaturity of the sarcoplasmic reticulum. For cell-replacement strategies, further maturation of cardiac cells has to be achieved either in vitro before or in vivo after transplantation.
Collapse
Affiliation(s)
- Frank Pillekamp
- Pediatric Cardiology, Heinrich-Heine-University of Duesseldorf, Duesseldorf, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Park JH, Yoon JY, Ko SM, Jin SA, Kim JH, Cho CH, Kim JM, Lee JH, Choi SW, Seong IW, Jeong JO. Endothelial progenitor cell transplantation decreases lymphangiogenesis and adverse myocardial remodeling in a mouse model of acute myocardial infarction. Exp Mol Med 2012; 43:479-85. [PMID: 21694495 DOI: 10.3858/emm.2011.43.8.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cardiac lymphatic system in the remodeling after acute myocardial infarction (AMI) has been overlooked. We wanted to investigate the role of bone marrow-derived endothelial progenitor cells (EPCs) and their contribution to lymphatic distribution in myocardial remodeling after AMI. Mouse (C57bl/6J) MI models were created by ligation of the left anterior descending coronary artery and were treated with phosphate buffered saline (PBS) or EPCs. Real-time RT-PCR with 2- to 4-week myocardial tissue samples revealed that lymphangiogenetic factors such as vascular endothelial growth factor (VEGF)-C (8.5 fold, P < 0.05), VEGF-D (6.1 fold, P < 0.05), Lyve-1 (15 fold, P < 0.05), and Prox-1 (11 fold, P < 0.05) were expressed at significantly higher levels in the PBS group than the EPC group. The PBS group also showed a significantly higher density of lymphatic vessels in the peri-infarction area. Echocardiography showed that from 2 weeks after the treatment, left ventricle (LV) dimensions at both systole and diastole were significantly smaller in the EPC group than in the PBS group (P < 0.01) and LV fractional shortening was higher in the EPC group accordingly (P < 0.01). Lymphangiogenic markers increased in a mouse MI model. EPC transplantation decreased lymphangiogenesis and adverse ventricular remodeling after AMI. These novel findings suggest that new lymphatic vessels may be formed in severely damaged myocardium, and may be involved in adverse myocardial remodeling after AMI.
Collapse
Affiliation(s)
- Jae-Hyeong Park
- Division of Cardiology, Chungnam National University, Daejeon 301-721, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Irradiation induces homing of donor endothelial progenitor cells in allogeneic hematopoietic stem cell transplantation. Int J Hematol 2012; 95:189-97. [DOI: 10.1007/s12185-011-1000-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Revised: 12/13/2011] [Accepted: 12/13/2011] [Indexed: 10/14/2022]
|
34
|
Hang R, Zhang M, Ma S, Chu PK. Biological response of endothelial cells to diamond-like carbon-coated NiTi alloy. J Biomed Mater Res A 2011; 100:496-506. [PMID: 22125203 DOI: 10.1002/jbm.a.33295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 07/11/2011] [Accepted: 09/26/2011] [Indexed: 11/08/2022]
Abstract
Diamond-like carbon (DLC) coatings were deposited on nearly equiatomic nickel-titanium (NiTi) alloy by arc-enhanced magnetron sputtering. The microstructure, surface morphology, chemical composition, surface free energy, protein adsorbance, and leach amount of Ni ions were assessed by Raman spectroscopy, high-resolution transmission electron microscopy (HR-TEM), atomic force microscopy (AFM), X-ray photoelectron spectroscopy (XPS), contact angle measurements, micro BCA™ protein assay kit, and inductively coupled plasma mass spectrometry (ICP-MS). The biological response of the endothelial cells (ECs) was evaluated by cell adhesion, morphology, viability, and expression levels of thrombogenicity-related genes. Our results show that the DLC coatings inhibit the release of Ni ions from the NiTi substrate effectively thus enhancing its biosafety. The easy adhesion, elongated morphology, and high viability of ECs on the DLC coatings suggest fast endothelialization after implantation and so application of DLC coatings improves the surface properties of NiTi in cardiovascular applications. The relationship between the surface characteristics, Ni leaching, and concomitant biological response are discussed in details.
Collapse
Affiliation(s)
- Ruiqiang Hang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | | | | | | |
Collapse
|
35
|
Hu S, Huang M, Nguyen PK, Gong Y, Li Z, Jia F, Lan F, Liu J, Nag D, Robbins RC, Wu JC. Novel microRNA prosurvival cocktail for improving engraftment and function of cardiac progenitor cell transplantation. Circulation 2011; 124:S27-34. [PMID: 21911815 DOI: 10.1161/circulationaha.111.017954] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although stem cell therapy has provided a promising treatment for myocardial infarction, the low survival of the transplanted cells in the infarcted myocardium is possibly a primary reason for failure of long-term improvement. Therefore, the development of novel prosurvival strategies to boost stem cell survival will be of significant benefit to this field. METHODS AND RESULTS Cardiac progenitor cells (CPCs) were isolated from transgenic mice, which constitutively express firefly luciferase and green fluorescent protein. The CPCs were transduced with individual lentivirus carrying the precursor of miR-21, miR-24, and miR-221, a cocktail of these 3 microRNA precursors, or green fluorescent protein as a control. After challenge in serum free medium, CPCs treated with the 3 microRNA cocktail showed significantly higher viability compared with untreated CPCs. After intramuscular and intramyocardial injections, in vivo bioluminescence imaging showed that microRNA cocktail-treated CPCs survived significantly longer after transplantation. After left anterior descending artery ligation, microRNA cocktail-treated CPCs boost the therapeutic efficacy in terms of functional recovery. Histological analysis confirmed increased myocardial wall thickness and CPC engraftment in the myocardium with the microRNA cocktail. Finally, we used bioinformatics analysis and experimental validation assays to show that Bim, a critical apoptotic activator, is an important target gene of the microRNA cocktail, which collectively can bind to the 3'UTR region of Bim and suppress its expression. CONCLUSIONS We have demonstrated that a microRNA prosurvival cocktail (miR-21, miR-24, and miR-221) can improve the engraftment of transplanted cardiac progenitor cells and therapeutic efficacy for treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Shijun Hu
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305-5454, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Nascimento DS, Valente M, Esteves T, de Pina MDF, Guedes JG, Freire A, Quelhas P, Pinto-do-Ó P. MIQuant--semi-automation of infarct size assessment in models of cardiac ischemic injury. PLoS One 2011; 6:e25045. [PMID: 21980376 PMCID: PMC3184116 DOI: 10.1371/journal.pone.0025045] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 08/23/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The cardiac regenerative potential of newly developed therapies is traditionally evaluated in rodent models of surgically induced myocardial ischemia. A generally accepted key parameter for determining the success of the applied therapy is the infarct size. Although regarded as a gold standard method for infarct size estimation in heart ischemia, histological planimetry is time-consuming and highly variable amongst studies. The purpose of this work is to contribute towards the standardization and simplification of infarct size assessment by providing free access to a novel semi-automated software tool. The acronym MIQuant was attributed to this application. METHODOLOGY/PRINCIPAL FINDINGS Mice were subject to permanent coronary artery ligation and the size of chronic infarcts was estimated by area and midline-length methods using manual planimetry and with MIQuant. Repeatability and reproducibility of MIQuant scores were verified. The validation showed high correlation (r(midline length) = 0.981; r(area) = 0.970 ) and agreement (Bland-Altman analysis), free from bias for midline length and negligible bias of 1.21% to 3.72% for area quantification. Further analysis demonstrated that MIQuant reduced by 4.5-fold the time spent on the analysis and, importantly, MIQuant effectiveness is independent of user proficiency. The results indicate that MIQuant can be regarded as a better alternative to manual measurement. CONCLUSIONS We conclude that MIQuant is a reliable and an easy-to-use software for infarct size quantification. The widespread use of MIQuant will contribute towards the standardization of infarct size assessment across studies and, therefore, to the systematization of the evaluation of cardiac regenerative potential of emerging therapies.
Collapse
Affiliation(s)
- Diana S. Nascimento
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Mariana Valente
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Tiago Esteves
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Maria de Fátima de Pina
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Departamento de Epidemiologia Clínica, Medicina Preditiva e Saúde Pública, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Joana G. Guedes
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Ana Freire
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Pedro Quelhas
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Perpétua Pinto-do-Ó
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
37
|
Bollini S, Pozzobon M, Nobles M, Riegler J, Dong X, Piccoli M, Chiavegato A, Price AN, Ghionzoli M, Cheung KK, Cabrelle A, O'Mahoney PR, Cozzi E, Sartore S, Tinker A, Lythgoe MF, De Coppi P. In vitro and in vivo cardiomyogenic differentiation of amniotic fluid stem cells. Stem Cell Rev Rep 2011; 7:364-80. [PMID: 21120638 DOI: 10.1007/s12015-010-9200-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cell therapy has developed as a complementary treatment for myocardial regeneration. While both autologous and allogeneic uses have been advocated, the ideal candidate has not been identified yet. Amniotic fluid-derived stem (AFS) cells are potentially a promising resource for cell therapy and tissue engineering of myocardial injuries. However, no information is available regarding their use in an allogeneic context. c-kit-sorted, GFP-positive rat AFS (GFP-rAFS) cells and neonatal rat cardiomyocytes (rCMs) were characterized by cytocentrifugation and flow cytometry for the expression of mesenchymal, embryonic and cell lineage-specific antigens. The activation of the myocardial gene program in GFP-rAFS cells was induced by co-culture with rCMs. The stem cell differentiation was evaluated using immunofluorescence, RT-PCR and single cell electrophysiology. The in vivo potential of Endorem-labeled GFP-rAFS cells for myocardial repair was studied by transplantation in the heart of animals with ischemia/reperfusion injury (I/R), monitored by magnetic resonance imaging (MRI). Three weeks after injection a small number of GFP-rAFS cells acquired an endothelial or smooth muscle phenotype and to a lesser extent CMs. Despite the low GFP-rAFS cells count in the heart, there was still an improvement of ejection fraction as measured by MRI. rAFS cells have the in vitro propensity to acquire a cardiomyogenic phenotype and to preserve cardiac function, even if their potential may be limited by poor survival in an allogeneic setting.
Collapse
Affiliation(s)
- Sveva Bollini
- Stem Cell Processing Laboratory-Fondazione Città della Speranza, Venetian Institute of Molecular Medicine (VIMM), University of Padua, Via G. Orus, 2, 35129, Padua, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Rosenblatt-Velin N, Ogay S, Felley A, Stanford WL, Pedrazzini T. Cardiac dysfunction and impaired compensatory response to pressure overload in mice deficient in stem cell antigen-1. FASEB J 2011; 26:229-39. [PMID: 21957128 DOI: 10.1096/fj.11-189605] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Stem cell antigen-1 (Sca-1) has been used to identify cardiac stem cells in the mouse heart. To investigate the function of Sca-1 in aging and during the cardiac adaptation to stress, we used Sca-1-deficient mice. These mice developed dilated cardiomyopathy [end-diastolic left ventricular diameter at 18 wk of age: wild-type (WT) mice, 4.2 mm ± 0.3; Sca-1-knockout (Sca-1-KO) mice, 4.6 mm ± 0.1; ejection fraction: WT mice, 51.1 ± 2.7%; Sca-1-KO mice, 42.9 ± 2.7%]. Furthermore, the hearts of mice lacking Sca-1 demonstrated exacerbated susceptibility to pressure overload [ejection fraction after transaortic constriction (TAC): WT mice, 43.5 ± 3.2%; Sca-1-KO mice, 30.8% ± 4.0] and increased apoptosis, as shown by the 2.5-fold increase in TUNEL(+) cells in Sca-1-deficient hearts under stress. Sca-1 deficiency affected primarily the nonmyocyte cell fraction. Indeed, the number of Nkx2.5(+) nonmyocyte cells, which represent a population of cardiac precursor cells (CPCs), was 2-fold smaller in Sca-1 deficient neonatal hearts. In vitro, the ability of CPCs to differentiate into cardiomyocytes was not affected by Sca-1 deletion. In contrast, these cells demonstrated unrestricted differentiation into cardiomyocytes. Interestingly, proliferation of cardiac nonmyocyte cells in response to stress, as judged by BrdU incorporation, was higher in mice lacking Sca-1 (percentages of BrdU(+) cells in the heart after TAC: WT mice, 4.4 ± 2.1%; Sca-1-KO mice, 19.3 ± 4.2%). These data demonstrate the crucial role of Sca-1 in the maintenance of cardiac integrity and suggest that Sca-1 restrains spontaneous differentiation in the precursor population. The absence of Sca-1 results in uncontrolled precursor recruitment, exhaustion of the precursor pool, and cardiac dysfunction.
Collapse
|
39
|
Gonzales C, Ullrich ND, Gerber S, Berthonneche C, Niggli E, Pedrazzini T. Isolation of cardiovascular precursor cells from the human fetal heart. Tissue Eng Part A 2011; 18:198-207. [PMID: 21902604 DOI: 10.1089/ten.tea.2011.0022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Weakening of cardiac function in patients with heart failure results from a loss of cardiomyocytes in the damaged heart. Cell replacement therapies as a way to induce myocardial regeneration in humans could represent attractive alternatives to classical drug-based approaches. However, a suitable source of precursor cells, which could produce a functional myocardium after transplantation, remains to be identified. In the present study, we isolated cardiovascular precursor cells from ventricles of human fetal hearts at 12 weeks of gestation. These cells expressed Nkx2.5 but not late cardiac markers such as α-actinin and troponin I. In addition, proliferating cells expressed the mesenchymal stem cell markers CD73, CD90, and CD105. Evidence for functional cardiogenic differentiation in vitro was demonstrated by the upregulation of cardiac gene expression as well as the appearance of cells with organized sarcomeric structures. Importantly, differentiated cells presented spontaneous and triggered calcium signals. Differentiation into smooth muscle cells was also detected. In contrast, precursor cells did not produce endothelial cells. The engraftment and differentiation capacity of green fluorescent protein (GFP)-labeled cardiac precursor cells were then tested in vivo after transfer into the heart of immunodeficient severe combined immunodeficient mice. Engrafted human cells were readily detected in the mouse myocardium. These cells retained their cardiac commitment and differentiated into α-actinin-positive cardiomyocytes. Expression of connexin-43 at the interface between GFP-labeled and endogenous cardiomyocytes indicated that precursor-derived cells connected to the mouse myocardium. Together, these results suggest that human ventricular nonmyocyte cells isolated from fetal hearts represent a suitable source of precursors for cell replacement therapies.
Collapse
Affiliation(s)
- Christine Gonzales
- Experimental Cardiology Unit, Department of Medicine, University of Lausanne Medical School, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
40
|
Wu Z, Li H, Rao L, He L, Bao L, Liao J, Cui C, Zuo Z, Li Q, Dai H, Qian L, Tian Q, Xiao L, Tan X. Derivation and characterization of human embryonic stem cell lines from the Chinese population. J Genet Genomics 2011; 38:13-20. [PMID: 21338948 DOI: 10.1016/j.jcg.2010.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Revised: 11/01/2010] [Accepted: 11/05/2010] [Indexed: 12/31/2022]
Abstract
Human embryonic stem cells (hESCs) can self-renew indefinitely and differentiate into all cell types in the human body. Therefore, they are valuable in regenerative medicine, human developmental biology and drug discovery. A number of hESC lines have been derived from the Chinese population, but limited of them are available for research purposes. Here we report the derivation and characterization of two hESC lines derived from human blastocysts of Chinese origin. These hESCs express alkaline phosphatase and hESC-specific markers, including Oct4, Nanog, SSEA-3, SSEA-4, TRA-1-60 and TRA-1-81. They also have high levels of telomerase activity and normal karyotypes. These cells can form embryoid body in vitro and can be differentiated into all three germ layers in vivo by teratoma formation. The newly established hESCs will be distributed for research purposes. The availability of hESC lines from the Chinese population will facilitate studies on the differences in hESCs from different ethnic groups.
Collapse
Affiliation(s)
- Zhao Wu
- Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Cell Bank, Stem Cell Bank, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ye Z, Zhou Y, Cai H, Tan W. Myocardial regeneration: Roles of stem cells and hydrogels. Adv Drug Deliv Rev 2011; 63:688-97. [PMID: 21371512 DOI: 10.1016/j.addr.2011.02.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 02/23/2011] [Accepted: 02/23/2011] [Indexed: 01/19/2023]
Abstract
Heart failure remains the leading cause of morbidity and mortality. Recently, it was reported that the adult heart has intrinsic regenerative capabilities, prompting a great wave of research into applying cell-based therapies, especially with skeletal myoblasts and bone marrow-derived cells, to regenerate heart tissues. While the mechanism of action for the observed beneficial effects of bone marrow-derived cells remains unclear, new cell candidates are emerging, including embryonic stem (ES) and introduced pluripotent stem (iPS) cells, as well as cardiac stem cells (CSCs) from adult hearts. However, the very low engraftment efficiency and survival of implanted cells prevent cell therapy from turning into a clinical reality. Injectable hydrogel biomaterials based on hydrophilic, biocompatible polymers and peptides have great potential for addressing many of these issues by serving as cell/drug delivery vehicles and as a platform for cardiac tissue engineering. In this review, we will discuss the application of stem cells and hydrogels in myocardial regeneration.
Collapse
Affiliation(s)
- Zhaoyang Ye
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | | | | | | |
Collapse
|
42
|
van Weerd JH, Koshiba-Takeuchi K, Kwon C, Takeuchi JK. Epigenetic factors and cardiac development. Cardiovasc Res 2011; 91:203-11. [PMID: 21606181 DOI: 10.1093/cvr/cvr138] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Congenital heart malformations remain the leading cause of death related to birth defects. Recent advances in developmental and regenerative cardiology have shed light on a mechanistic understanding of heart development that is controlled by a transcriptional network of genetic and epigenetic factors. This article reviews the roles of chromatin remodelling factors important for cardiac development with the current knowledge of cardiac morphogenesis, regeneration, and direct cardiac differentiation. In the last 5 years, critical roles of epigenetic factors have been revealed in the cardiac research field.
Collapse
Affiliation(s)
- Jan Hendrick van Weerd
- Cardiovascular Regeneration, Institute of Molecular and Cellular Biosciences, University of Tokyo, Tokyo, Japan
| | | | | | | |
Collapse
|
43
|
Fagoonee S, Pellicano R, Silengo L, Altruda F. Potential applications of germline cell-derived pluripotent stem cells in organ regeneration. Organogenesis 2011; 7:116-122. [PMID: 21593601 PMCID: PMC3142448 DOI: 10.4161/org.7.2.16284] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 04/24/2011] [Accepted: 05/02/2011] [Indexed: 01/01/2023] Open
Abstract
Impressive progress has been made since the turn of the century in the field of stem cells. Different types of stem cells have now been isolated from different types of tissues. Pluripotent stem cells are the most promising cell source for organ regeneration. One such cell type is the germline cell-derived pluripotent cell, which is derived from adult spermatogonial stem cells. The germline cell-derived pluripotent stem cells have been obtained from both human and mouse and, importantly, are adult stem cells with embryonic stem cell-like properties that do not require specific manipulations for pluripotency acquisition, hence bypassing problems related to induced pluripotent stem cells and embryonic stem cells. The germline cell-derived pluripotent stem cells have been induced to differentiate into cells deriving from the three germ layers and shown to be functional in vitro. This review will discuss the plasticity of the germline cell-derived pluripotent stem cells and their potential applications in human organ regeneration, with special emphasis on liver regeneration. Potential problems related to their use are also highlighted.
Collapse
|
44
|
Bollini S, Smart N, Riley PR. Resident cardiac progenitor cells: at the heart of regeneration. J Mol Cell Cardiol 2011; 50:296-303. [PMID: 20643135 DOI: 10.1016/j.yjmcc.2010.07.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2010] [Revised: 07/02/2010] [Accepted: 07/08/2010] [Indexed: 10/19/2022]
Abstract
Stem cell therapy has recently emerged as an innovative strategy over conventional cardiovascular treatments to restore cardiac function in patients affected by ischemic heart disease. Various stem cell populations have been tested and their potential for cardiac repair has been analyzed. Embryonic stem cells retain the greatest differentiation potential, but concerns persist with regard to their immunogenic and teratogenic effects. Although adult somatic stem cells are not tumourigenic and easier to use in an autologous setting, they exist in small numbers and possess reduced differentiation potential. Traditionally the heart was considered to be a post-mitotic organ; however, this dogma has recently been challenged with the identification of a reservoir of resident stem cells, defined as cardiac progenitor cells (CPCs). These endogenous progenitors may represent the best candidates for cardiovascular cell therapy, as they are tissue-specific, often pre-committed to a cardiac fate, and display a greater propensity to differentiate towards cardiovascular lineages. This review will focus on current research into the biology of CPCs and their regenerative potential. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".
Collapse
Affiliation(s)
- Sveva Bollini
- Molecular Medicine Unit, University College London-Institute of Child Health, London, UK
| | | | | |
Collapse
|
45
|
Bone marrow support of the heart in pressure overload is lost with aging. PLoS One 2010; 5:e15187. [PMID: 21203577 PMCID: PMC3006343 DOI: 10.1371/journal.pone.0015187] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Accepted: 10/30/2010] [Indexed: 01/01/2023] Open
Abstract
RATIONALE Exogenous stem cell delivery is under investigation to prevent and treat cardiac dysfunction. It is less studied as to the extent endogenous bone marrow derived stem cells contribute to cardiac homeostais in response to stress and the affects of aging on this stress response. OBJECTIVE To determine the role of bone marrow (BM) derived stem cells on cardiac homeostasis in response to pressure overload (PO) and how this response is altered by aging. METHODS AND RESULTS Young (8 weeks) and old (>40 weeks) C57/b6 mice underwent homo- and heterochronic BM transplantation prior to transverse aortic constriction (TAC). We found that older BM is associated with decreased cardiac function following TAC. This decreased function is associated with decrease in BM cell engraftment, increased myocyte apoptosis, decreased myocyte hypertrophy, increased myocardial fibrosis and decreased cardiac function. Additionally, there is a decrease in activation of resident cells within the heart in response to PO in old mice. Interestingly, these effects are not due to alterations in vascular density or inflammation in response to PO or differences in ex vivo stem cell migration between young and old mice. CONCLUSIONS BM derived stem cells are activated in response to cardiac PO, and the recruitment of BM derived cells are involved in cardiac myocyte hypertrophy and maintenance of function in response to PO which is lost with aging.
Collapse
|
46
|
Abstract
Traditionally, nuclear reprogramming of cells has been performed by transferring somatic cell nuclei into oocytes, by combining somatic and pluripotent cells together through cell fusion and through genetic integration of factors through somatic cell chromatin. All of these techniques changes gene expression which further leads to a change in cell fate. Here we discuss recent advances in generating induced pluripotent stem cells, different reprogramming methods and clinical applications of iPS cells. Viral vectors have been used to transfer transcription factors (Oct4, Sox2, c-myc, Klf4, and nanog) to induce reprogramming of mouse fibroblasts, neural stem cells, neural progenitor cells, keratinocytes, B lymphocytes and meningeal membrane cells towards pluripotency. Human fibroblasts, neural cells, blood and keratinocytes have also been reprogrammed towards pluripotency. In this review we have discussed the use of viral vectors for reprogramming both animal and human stem cells. Currently, many studies are also involved in finding alternatives to using viral vectors carrying transcription factors for reprogramming cells. These include using plasmid transfection, piggyback transposon system and piggyback transposon system combined with a non viral vector system. Applications of these techniques have been discussed in detail including its advantages and disadvantages. Finally, current clinical applications of induced pluripotent stem cells and its limitations have also been reviewed. Thus, this review is a summary of current research advances in reprogramming cells into induced pluripotent stem cells.
Collapse
Affiliation(s)
- Minal Patel
- Department of Oral Biology, School of Dental Medicine, The State University of New York at Buffalo, Buffalo, NY, 14214
| | - Shuying Yang
- Department of Oral Biology, School of Dental Medicine, The State University of New York at Buffalo, Buffalo, NY, 14214
| |
Collapse
|
47
|
Abstract
The differentiation of embryonic stem cells along the endothelial cell lineage requires a tightly coordinated sequence of events that are regulated in both space and time. Although significant gaps remain in this process, major strides have been made over the past 10 years in identifying the growth factors, signal transduction pathways, and transcription factors that function together as critical mediators of this process. Examples of some of the signal transduction pathways include the hedgehog (HH), WNT, BMP, and Notch pathways. A complex interplay between growth factors, and activation of a variety of signal transduction pathways leads to the induction of transcriptional programs that promote the differentiation of embryonic stem cells along the endothelial lineage and ultimately into arterial, venous, and lymphatic endothelial cells. The purpose of this review is to summarize the recent advances in our understanding of the molecular mechanisms underlying endothelial differentiation.
Collapse
Affiliation(s)
- Alex Le Bras
- Division of Cardiology, and Molecular and Vascular Biology, Department of Medicine and the Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | | |
Collapse
|
48
|
Bonsignore MR, Morici G, Riccioni R, Huertas A, Petrucci E, Veca M, Mariani G, Bonanno A, Chimenti L, Gioia M, Palange P, Testa U. Hemopoietic and angiogenetic progenitors in healthy athletes: different responses to endurance and maximal exercise. J Appl Physiol (1985) 2010; 109:60-7. [PMID: 20448032 DOI: 10.1152/japplphysiol.01344.2009] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The effects of endurance or maximal exercise on mobilization of bone marrow-derived hemopoietic and angiogenetic progenitors in healthy subjects are poorly defined. In 10 healthy amateur runners, we collected venous blood before, at the end of, and the day after a marathon race (n = 9), and before and at the end of a 1.5-km field test (n = 8), and measured hemopoietic and angiogenetic progenitors by flow cytometry and culture assays, as well as plasma or serum concentrations of several cytokines/growth factors. After the marathon, CD34(+) cells were unchanged, whereas clonogenetic assays showed decreased number of colonies for both erythropoietic (BFU-E) and granulocyte-monocyte (CFU-GM) series, returning to baseline the morning post-race. Conversely, CD34(+) cells, BFU-E, and CFU-GM increased after the field test. Angiogenetic progenitors, assessed as CD34(+)KDR(+) and CD133(+)VE-cadherin(+) cells or as adherent cells in culture expressing endothelial markers, increased after both endurance and maximal exercise but showed a different pattern between protocols. Interleukin-6 increased more after the marathon than after the field test, whereas hepatocyte growth factor and stem cell factor increased similarly in both protocols. Plasma levels of angiopoietin (Ang) 1 and 2 increased after both types of exercise, whereas the Ang-1-to-Ang-2 ratio or vascular endothelial growth factor-A were little affected. These data suggest that circulating hemopoietic progenitors may be utilized in peripheral tissues during prolonged endurance exercise. Endothelial progenitor mobilization after exercise in healthy trained subjects appears modulated by the type of exercise. Exercise-induced increase in growth factors suggests a physiological trophic effect of exercise on the bone marrow.
Collapse
Affiliation(s)
- Maria R Bonsignore
- Biomedical Department, Internal and Specialistic Medicine (DIBIMIS), Section of Pneumology, University of Palermo, Via Trabucco, 180, 90146 Palermo, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Yamashita JK. ES and iPS cell research for cardiovascular regeneration. Exp Cell Res 2010; 316:2555-9. [PMID: 20385126 DOI: 10.1016/j.yexcr.2010.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Accepted: 04/04/2010] [Indexed: 01/08/2023]
Abstract
Embryonic stem (ES) cells and induced pluripotent stem (iPS) cells, which are ES-like stem cells induced from adult tissues, are twin stem cells with currently (with the exception of fertilized eggs) the broadest differentiation potentials. These two stem cells show various similarities in appearance, maintenance methods, growth and differentiation potentials, i.e. theoretically, those cells can give rise to all kinds of cells including germ-line cells. Generation of human ES and iPS cells is further facilitating the researches towards the realization of regenerative medicine. The following three issues are important purposes of ES and iPS cell researches for regenerative medicine: (1) dissection of differentiation mechanisms, (2) application to cell transplantation, and (3) drug discovery. In this review, the current status of cardiovascular regenerative trials using ES and iPS cells is briefly discussed.
Collapse
Affiliation(s)
- Jun K Yamashita
- Department of Cell Growth and Differentiation, Center for iPS Cell Research and Application, Kyoto University, Japan.
| |
Collapse
|