1
|
Yang L, Shen XM, Wang ZF, Li K, Wang W. The Notch signalling pathway and miRNA regulation play important roles in the differentiation of Schwann cells from adipose-derived stem cells. J Transl Med 2022; 102:320-328. [PMID: 34795395 DOI: 10.1038/s41374-021-00687-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/08/2022] Open
Abstract
An exploration of the underlying mechanisms is necessary to improve nerve myelin-forming cell Schwann cell (SC) differentiation from adipose-derived stem cells (ADSCs). Primary rat ADSCs were isolated and characterised for cell surface markers using flow cytometry analysis. After treatment with a mixture of glial growth factors, ADSCs were induced to differentiate and subsequently identified by immunofluorescence staining and western blotting. A miRNA microarray analysis was performed to explore the genes and signalling pathways regulating ADSC differentiation into SCs. ELISAs were conducted to measure the expression of neurotrophic factors and changes in the level of nerve cell adhesion factor. Dual luciferase reporter assays and RIP assays were performed to explore the potential mechanism of miR-21-5p in ADSC differentiation. The isolated ADSCs were positive for CD29 and CD44 but negative for CD49. After induction with specific cytokines, the differentiated ADSCs presented a spindle-like morphology similar to SCs and expressed S100. RNA-sequencing analyses revealed that 9821 mRNAs of protein-coding genes and 175 miRNAs were differentially expressed in differentiated SC-like cells compared to primary cultures of ADSCs. KEGG and Gene Ontology analyses revealed that the involvement of the Notch signalling pathway and miRNA negative regulation may be associated with the differentiation of ADSCs into SCs. Treatment with a Notch inhibitor promoted the differentiation of ADSCs. Furthermore, mechanistic studies showed that Jag1 bound to miR-21-5p and upregulated its target gene Jag1, thus affecting ADSC differentiation. These results revealed the mechanism underlying the important roles of miRNAs and the Notch signalling pathway in the differentiation of SCs from ADSCs, enabling potential therapeutic applications of ADSCs in peripheral nerve regeneration in the future.
Collapse
Affiliation(s)
- Liang Yang
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, Changsha, 410078, P.R. China
| | - Xiang-Min Shen
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, 410011, P.R. China
| | - Zhi-Fei Wang
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, Changsha, 410078, P.R. China
| | - Ke Li
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, 410011, P.R. China
| | - Wei Wang
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, 410011, P.R. China.
| |
Collapse
|
2
|
Apoptotic Bodies of Cardiomyocytes and Fibroblasts - Regulators of Directed Differentiation of Heart Stem Cells. Bull Exp Biol Med 2020; 170:112-117. [PMID: 33237531 DOI: 10.1007/s10517-020-05015-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Indexed: 12/22/2022]
Abstract
We studied the effects of apoptotic bodies of cardiomyocytes (ApBc) and fibroblasts (ApBf) on myocardial regeneration and contractility in rats and the dynamics of RNA concentrations in cardiomyocytes and fibroblasts at different stages of apoptosis. ApBc increase the contractility of rat myocardium, while ApBf reduce it. ApBc stimulate the development of clones of cardiomyocyte precursors in the myocardium, while ApBf stimulate the formation of endothelial precursor clones. In doxorubicin cardiomyopathy, ApBc, similar to the reference drug (ACE inhibitor) improve animal survival, while ApBf produce no such effect. RNA concentrations in cardiomyocytes and fibroblasts before apoptosis and at the beginning of cell death significantly differed, while in apoptotic bodies of these cells, it was practically the same. It has been hypothesized that RNA complex present in ApBc and ApBf represents an "epigenetic code" of directed differentiation of cardiac stem cells.
Collapse
|
3
|
Abstract
Adipose-derived stem/stromal cells (ASCs), together with adipocytes, vascular endothelial cells, and vascular smooth muscle cells, are contained in fat tissue. ASCs, like the human bone marrow stromal/stem cells (BMSCs), can differentiate into several lineages (adipose cells, fibroblast, chondrocytes, osteoblasts, neuronal cells, endothelial cells, myocytes, and cardiomyocytes). They have also been shown to be immunoprivileged, and genetically stable in long-term cultures. Nevertheless, unlike the BMSCs, ASCs can be easily harvested in large amounts with minimal invasive procedures. The combination of these properties suggests that these cells may be a useful tool in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Simone Ciuffi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Roberto Zonefrati
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Lao S, Xu J, Liu Y, Cai S, Lin L, Zhang J, Cai D, Yin S. A comparative study of the influence of two types of PHEMA stents on the differentiation of ASCs to myocardial cells. Mol Med Rep 2017; 16:507-514. [PMID: 28586071 PMCID: PMC5482065 DOI: 10.3892/mmr.2017.6680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 02/07/2017] [Indexed: 01/16/2023] Open
Abstract
In the present study, subcutaneous fat was obtained from adult women that had undergone conventional liposuction surgery. A comparative study was performed to investigate the effect of transparent and white poly-β-hydroxyethyl methacrylate (PHEMA) stents, which have different surface and cross-sectional morphological characteristics, on the differentiation of adipose-derived stem cells (ASCs) into myocardial cells. The cell counting kit-8 assay revealed that cell growth increased at varying rates among the different treatment groups. The absorbance of the experimental transparent PHEMA treated group increased in a time-dependent manner with the duration of incubation. The highest levels of proliferation were observed in the transparent PHEMA group. In addition, the transparent PHEMA treated group exhibited the strongest cell adhesion ability, which was significantly different to that of the white PHEMA group (P<0.01 and P<0.05 for Matrigel and fibronectin assay, respectively). Comparisons between the two stent materials with the inducer control group revealed statistically significant differences in the rate of ASC differentiation (P<0.05). The level of differentiation was the greatest in the transparent PHEMA group, and was significantly different to the white PHEMA group (P<0.05) and the blank control group (P<0.01). The results suggest that the inducers 5-aza-2-deoxycytidin and laminin, and material microstructure stents effectively promote the proliferation, growth and adhesion of ASCs. However, the transparent material microstructure may be a more suitable candidate for ASC-associated injections. The present study provides further evidence that a PHEMA stent structure, comprised of a high number of matrixes and a low water content, induces a high level of ASC differentiation to myocardial cells.
Collapse
Affiliation(s)
- Shen Lao
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Jing Xu
- Department of Ultrasonography, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Yunqi Liu
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Songwang Cai
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Lin Lin
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Junhang Zhang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510630, P.R. China
| | - Dongmei Cai
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Shengli Yin
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
5
|
Lee TM, Harn HJ, Chiou TW, Chuang MH, Chen CH, Lin PC, Lin SZ. Targeting the pathway of GSK-3β/nerve growth factor to attenuate post-infarction arrhythmias by preconditioned adipose-derived stem cells. J Mol Cell Cardiol 2017; 104:17-30. [PMID: 28130118 DOI: 10.1016/j.yjmcc.2017.01.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/01/2017] [Accepted: 01/23/2017] [Indexed: 10/20/2022]
Abstract
Adipose-derived stem cell (ADSC) transplantation is a promising new therapy to improve cardiac function after myocardial infarction. However, its low efficacy of transdifferentiation hampers its usefulness. Glycogen synthase kinase-3β (GSK-3β) signal has been shown to play a role in preconditioning-induced cardioprotection. We assessed whether n-butylidenephthalide (BP) primed ADSCs can attenuate arrhythmias by a GSK-3β-dependent pathway after myocardial infarction. Male Wistar rats after coronary ligation was randomly allocated to receive intramyocardial injection of vehicle, ADSCs, BP-preconditioned ADSCs, (BP+lithium)-preconditioned ADSCs, (BP+SB216763)-preconditioned ADSCs, and (BP+LY294002)-preconditioned ADSCs. ADSCs were primed for 16h before implantation. After 4weeks of implantation, ADSCs were retained in myocardium, reduced fibrosis and improved cardiac function. Sympathetic hyperinnervation was blunted after administering ADSCs, assessed by immunofluorescent analysis, and Western blotting and real-time quantitative RT-PCR of nerve growth factor. Arrhythmic scores during programmed stimulation in the ADSC-treated infarcted rats were significantly lower than vehicle. BP-preconditioned ADSCs had superior cardioprotection, greater ADSC engraftment and transdifferentiation, and antiarrhythmic effects compared with ADSCs alone. Simultaneously, BP increased the levels of phospho-Akt and down-regulated GSK-3β activity. The effects of BP against sympathetic hyperinnervation were blocked by LY294002, a PI3K inhibitor. Addition of either lithium or SB216763 did not have additional effects compared with BP alone. Compared with ADSC alone, BP-primed ADSC implantation improved stem cell engraftment and attenuated sympathetic hyperinnervation and arrhythmias through a PI3K/Akt/GSK-3β-dependent pathway, suggesting that a synergic action was achieved between BP pretreatment and ADSCs.
Collapse
Affiliation(s)
- Tsung-Ming Lee
- Department of Medicine, Cardiology Section, China Medical University-An Nan Hospital, Tainan, Taiwan; Department of Medicine, China Medical University, Taichung, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Tzu Chi Foundation; Department of Pathology, Buddhist Tzu Chi General Hospital, Tzu Chi University
| | - Tzyy-Wen Chiou
- Department of Life Science, Graduate Institute of Biotechnology, National Dong Hwa University, Hualien, Taiwan
| | - Ming-Hsi Chuang
- Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan; Department of Bioinformatics, Chung Hua University, Hsinchu, Taiwan
| | | | - Po-Cheng Lin
- Gwo Xi Stem Cell Applied Technology, Hsinchu, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Tzu Chi Foundation; Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Tzu Chi University.
| |
Collapse
|
6
|
Zhao B, Liu JQ, Zheng Z, Zhang J, Wang SY, Han SC, Zhou Q, Guan H, Li C, Su LL, Hu DH. Human amniotic epithelial stem cells promote wound healing by facilitating migration and proliferation of keratinocytes via ERK, JNK and AKT signaling pathways. Cell Tissue Res 2016; 365:85-99. [PMID: 26888423 DOI: 10.1007/s00441-016-2366-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 01/19/2016] [Indexed: 11/29/2022]
Abstract
Wound healing is a highly orchestrated physiological process consisting in a complex interaction of cellular and biochemical events. Human amniotic epithelial stem cells (HAESCs) have been shown to be an attractive resource for wound healing because they are primitive stem cells. However, the exact effects of amnion-derived stem cells on the migration or proliferation of keratinocytes and their potential mechanism are not fully understood. We have found that HAESCs accelerate the migration of keratinocytes and induce a remarkable increase in the activity of phospho-ERK, phospho-JNK, and phospho-AKT, the blockade of which by their specific inhibitors significantly inhibits migration induced by HAESC-conditioned medium (CM). Furthermore, the co-culture of keratinocytes with HAESCs up-regulates the expression levels of cell proliferation proteins Cyclin D1, Cyclin D3 and Mdm2. In vivo animal experiments have shown that HAESC-CM improves wound healing, whereas blockade with ERK, JNK and AKT inhibitors significantly impairs wound healing. Taken together, these results reveal, for the first time, that HAESCs promote wound healing by facilitating the migration and proliferation of keratinocytes via ERK, JNK and AKT signaling pathways and might be a potential therapy in skin wound healing.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jia-Qi Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhao Zheng
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jun Zhang
- Department of Plastic Surgery, Shenyang Northern Hospital, Shenyang, Liaoning, China
| | - Shu-Yue Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shi-Chao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qin Zhou
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hao Guan
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chao Li
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lin-Lin Su
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Da-Hai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Gao S, Zheng Y, Cai Q, Wu X, Yao W, Wang J. Different methods for inducing adipose-derived stem cells to differentiate into Schwann-like cells. Arch Med Sci 2015; 11:886-92. [PMID: 26322102 PMCID: PMC4548042 DOI: 10.5114/aoms.2015.53310] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/12/2013] [Accepted: 08/26/2013] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION The aim of the study was to explore an effective method to induce adipose-derived stem cells (ADSCs) to differentiate into Schwann-like cells in vitro. MATERIAL AND METHODS Reagents were applied in two different ways (Dezawa inducing method and modified inducing method) in which inducers including β-mercaptoethanol (β-ME), all-trans-retinoic acid (ATRA), type I collagenase, forskolin, heregulin, basic fibroblast growth factor (BFGF) and brain-derived neurotrophic factor (BDNF) were used in different ways to induce ADSCs of rats to differentiate into Schwann-like cells. After induction, the cell morphologic characteristics and the cellular immunohistochemical staining positive rate of anti-S100 and anti-GFAP (glial fibrillary acidic protein) antibodies and the gray value of immunocytochemical dye with anti-S100 and anti-GFAP antibodies and cell activity measured by the MTT method were compared with each other to evaluate the induction effects. RESULTS Both methods can induce differentiation of ADSCs of rats into Schwann-like cells, but the cellular morphology of the modified method was more similar to Schwann cells than that of the Dezawa inducing method, there was a higher cellular immunohistochemical staining positive rate and staining grey value in immunocytochemical dye with anti-S100 and anti-GFAP antibodies, and less damage in the cell activity of the modified inducing method than that of the Dezawa inducing method. CONCLUSIONS The effect of the modified method to induce ADSCs to differentiate into Schwann-like cells in vitro is superior to that of the Dezawa inducing method.
Collapse
Affiliation(s)
- Songtao Gao
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Zheng
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiqing Cai
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejian Wu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weitao Yao
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiaqiang Wang
- Department of Orthopedics, The Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
8
|
Kikuchi C, Bienengraeber M, Canfield S, Koopmeiner A, Schäfer R, Bosnjak ZJ, Bai X. Comparison of Cardiomyocyte Differentiation Potential Between Type 1 Diabetic Donor- and Nondiabetic Donor-Derived Induced Pluripotent Stem Cells. Cell Transplant 2015; 24:2491-504. [PMID: 25562386 DOI: 10.3727/096368914x685762] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is the most common type of diabetes in children and adolescents. Diabetic subjects are more likely to experience a myocardial infarction compared to nondiabetic subjects. In recent years, induced pluripotent stem cells (iPSCs) have received increasing attention from basic scientists and clinicians and hold promise for myocardial regeneration due to their unlimited proliferation potential and differentiation capacity. However, cardiomyogenesis of type 1 diabetic donor-derived iPSCs (T1DM-iPSCs) has not been investigated yet. The aim of the study was to comparatively analyze cardiomyocyte (CM) differentiation capacity of nondiabetic donor-derived iPSCs (N-iPSCs) and T1DM-iPSCs. The differentiated CMs were confirmed by both expression of cardiac-specific markers and presence of cardiac action potential. Since mitochondrial bioenergetics is vital to every aspect of CM function, extracellular acidification rates and oxygen consumption rates were measured using Seahorse extracellular flux analyzer. The results showed that N-iPSCs and T1DM-iPSCs demonstrated similar capacity of differentiation into spontaneously contracting CMs exhibiting nodal-, atrial-, or ventricular-like action potentials. Differentiation efficiency was up to 90%. In addition, the CMs differentiated from N-iPSCs and T1DM-iPSCs (N-iPSC-CMs and T1DM-iPSC-CMs, respectively) showed 1) well-regulated glucose utilization at the level of glycolysis and mitochondrial oxidative phosphorylation and 2) the ability to switch metabolic pathways independent of extracellular glucose concentration. Collectively, we demonstrate for the first time that T1DM-iPSCs can differentiate into functional CMs with well-regulated glucose utilization as shown in N-iPSCs, suggesting that T1DM-iPSC-CMs might be a promising autologous cell source for myocardial regeneration in type 1 diabetes patients.
Collapse
Affiliation(s)
- Chika Kikuchi
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Immunogenicity of allogeneic mesenchymal stem cells transplanted via different routes in diabetic rats. Cell Mol Immunol 2014; 12:444-55. [PMID: 25242276 DOI: 10.1038/cmi.2014.70] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Revised: 07/02/2014] [Accepted: 07/03/2014] [Indexed: 02/06/2023] Open
Abstract
Due to their hypoimmunogenicity and unique immunosuppressive properties, mesenchymal stem cells (MSCs) are considered one of the most promising adult stem cell types for cell therapy. Although many studies have shown that MSCs exert therapeutic effects on several acute and subacute conditions, their long-term effects are not confirmed in chronic diseases. Immunogenicity is a major limitation for cell replacement therapy, and it is not well understood in vivo. We evaluated the immunogenicity of allogeneic MSCs in vivo by transplanting MSCs into normal and diabetic rats via the tail vein or pancreas and found that MSCs exhibited low immunogenicity in normal recipients and even exerted some immunosuppressive effects in diabetic rats during the initial phase. However, during the later stage in the pancreas group, MSCs expressed insulin and MHC II, eliciting a strong immune response in the pancreas. Simultaneously, the peripheral blood mononuclear cells in the recipients in the pancreas group were activated, and alloantibodies developed in vivo. Conversely, in the tail vein group, MSCs remained immunoprivileged and displayed immunosuppressive effects in vivo. These data indicate that different transplanting routes and microenvironments can lead to divergent immunogenicity of MSCs.
Collapse
|
10
|
Combination of acellular nerve graft and schwann cells-like cells for rat sciatic nerve regeneration. Neural Plast 2014; 2014:139085. [PMID: 25114806 PMCID: PMC4120921 DOI: 10.1155/2014/139085] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 05/28/2014] [Accepted: 06/16/2014] [Indexed: 01/30/2023] Open
Abstract
Objective. To investigate the effect of tissue engineering nerve on repair of rat sciatic nerve defect. Methods. Forty-five rats with defective sciatic nerve were randomly divided into three groups. Rats in group A were repaired by acellular nerve grafts only. Rats in group B were repaired by tissue engineering nerve. In group C, rats were repaired by autogenous nerve grafts. After six and twelve weeks, sciatic nerve functional index (SFI), neural electrophysiology (NEP), histological and transmission electron microscope observation, recovery ratio of wet weight of gastrocnemius muscle, regenerated myelinated nerve fibers number, nerve fiber diameter, and thickness of the myelin sheath were measured to assess the effect. Results. After six and twelve weeks, the recovery ratio of SFI and wet weight of gastrocnemius muscle, NEP, and the result of regenerated myelinated nerve fibers in groups B and C were superior to that of group A (P < 0.05), and the difference between groups B and C was not statistically significant (P > 0.05). Conclusion. The tissue engineering nerve composed of acellular allogenic nerve scaffold and Schwann cells-like cells can effectively repair the nerve defect in rats and its effect was similar to that of the autogenous nerve grafts.
Collapse
|
11
|
Blazquez-Martinez A, Chiesa M, Arnalich F, Fernandez-Delgado J, Nistal M, De Miguel MP. c-Kit identifies a subpopulation of mesenchymal stem cells in adipose tissue with higher telomerase expression and differentiation potential. Differentiation 2014; 87:147-60. [PMID: 24713343 DOI: 10.1016/j.diff.2014.02.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 01/24/2014] [Accepted: 02/24/2014] [Indexed: 12/14/2022]
Abstract
The stromal vascular fraction (SVF) of adipose tissue is an easy to obtain source of adipose tissue-derived stem cells (ADSCs). We and others have achieved significant but suboptimal therapeutic effects with ADSCs in various settings, mainly due to low rates of differentiation into specific cell types and with the downside of undesired side effects as a consequence of the undifferentiated ADSCs. These data prompted us to find new stem cell-specific markers for ADSCs and/or subpopulations with higher differentiation potential to specific lineages. We found a subpopulation of human ADSCs, marked by c-Kit positiveness, resides in a perivascular location, and shows higher proliferative activity and self-renewal capacity, higher telomerase activity and expression, higher in vitro adipogenic efficiency, a higher capacity for the maintenance of cardiac progenitors, and higher pancreatogenic and hepatogenic efficiency independently of CD105 expression. Our data suggests that the isolation of ADSC subpopulations with anti-c-Kit antibodies allows for the selection of a more homogeneous subpopulation with increased cardioprotective properties and increased adipogenic and endodermal differentiation potential, providing a useful tool for specific therapies in regenerative medicine applications.
Collapse
Affiliation(s)
- A Blazquez-Martinez
- Cell Engineering Laboratory, La Paz University Hospital Research Institute, Madrid, Spain
| | - M Chiesa
- Cell Engineering Laboratory, La Paz University Hospital Research Institute, Madrid, Spain
| | - F Arnalich
- Department of Internal Medicine, La Paz University Hospital, Madrid, Spain
| | - J Fernandez-Delgado
- Department of Plastic and Reconstructive Surgery, Santa Cristina Hospital, and Centrocim, Madrid, Spain
| | - M Nistal
- Department of Pathology, La Paz University Hospital, Madrid, Spain
| | - M P De Miguel
- Cell Engineering Laboratory, La Paz University Hospital Research Institute, Madrid, Spain.
| |
Collapse
|
12
|
Ilmer M, Vykoukal J, Boiles AR, Coleman M, Alt E. Two sides of the same coin: stem cells in cancer and regenerative medicine. FASEB J 2014; 28:2748-61. [DOI: 10.1096/fj.13-244640] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Matthias Ilmer
- Department of Translational Molecular PathologyThe University of Texas M. D. Anderson Cancer CenterHoustonTexasUSA
| | - Jody Vykoukal
- Department of Translational Molecular PathologyThe University of Texas M. D. Anderson Cancer CenterHoustonTexasUSA
| | - Alejandro Recio Boiles
- Department of Translational Molecular PathologyThe University of Texas M. D. Anderson Cancer CenterHoustonTexasUSA
| | | | - Eckhard Alt
- Center for Stem Cell and Developmental BiologyThe University of Texas M. D. Anderson Cancer CenterHoustonTexasUSA
- Applied Stem Cell Laboratory, Heart and Vascular InstituteDepartment of MedicineTulane University Health Science CenterNew OrleansLouisianaUSA
| |
Collapse
|
13
|
Bekhite MM, Finkensieper A, Rebhan J, Huse S, Schultze-Mosgau S, Figulla HR, Sauer H, Wartenberg M. Hypoxia, Leptin, and Vascular Endothelial Growth Factor Stimulate Vascular Endothelial Cell Differentiation of Human Adipose Tissue-Derived Stem Cells. Stem Cells Dev 2014; 23:333-51. [DOI: 10.1089/scd.2013.0268] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Mohamed M. Bekhite
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Andreas Finkensieper
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Jennifer Rebhan
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Stephanie Huse
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Stefan Schultze-Mosgau
- Department of Cranio-Maxillofacial Surgery and Plastic Surgery, Jena University Hospital, Jena, Germany
| | - Hans-Reiner Figulla
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| | - Heinrich Sauer
- Department of Physiology, Faculty of Medicine, Justus Liebig University, Giessen, Germany
| | - Maria Wartenberg
- Clinic of Internal Medicine I, Department of Cardiology, University Heart Center, Jena University Hospital, Jena, Germany
| |
Collapse
|
14
|
Sousa BR, Parreira RC, Fonseca EA, Amaya MJ, Tonelli FMP, Lacerda SMSN, Lalwani P, Santos AK, Gomes KN, Ulrich H, Kihara AH, Resende RR. Human adult stem cells from diverse origins: An overview from multiparametric immunophenotyping to clinical applications. Cytometry A 2013; 85:43-77. [DOI: 10.1002/cyto.a.22402] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Bruna R. Sousa
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Ricardo C. Parreira
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Emerson A Fonseca
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Maria J. Amaya
- Department of Internal Medicine, Section of Digestive Diseases; Yale University School of Medicine; New Haven Connecticut
| | - Fernanda M. P. Tonelli
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Samyra M. S. N. Lacerda
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Pritesh Lalwani
- Faculdade de Ciências Farmacêuticas; Universidade Federal do Amazonas; Manaus AM Brazil
| | - Anderson K. Santos
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Katia N. Gomes
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Henning Ulrich
- Departamento de Bioquímica; Instituto de Química, Universidade de São Paulo; São Paulo SP Brazil
| | - Alexandre H. Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição; Universidade Federal do ABC; Santo André SP Brazil
| | - Rodrigo R. Resende
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| |
Collapse
|
15
|
Abstract
The perception of adipose tissue has changed considerably with the dramatic increase in the incidence of obesity and obesity-related comorbidities over the past 3 decades. Excess fat is no longer associated with wealth, but is instead recognized as a risk factor for many diseases. Adipose tissue is increasingly being identified as a vital, complex endocrine organ, and not simply as a fat store. Not all fat is created equal--regional, developmental, structural, and functional variations exist. Epicardial adipose tissue is a metabolically active organ producing a number of factors that modulate cardiac structure and function. The global epidemic of obesity and metabolic syndrome imposes a major disease burden, particularly of cardiovascular disease. In this Review, we describe the various types of adipose tissue--their developmental biology, differentiation, cell heterogeneity, and functional characteristics. We discuss the link between adipose tissue and inflammation, the signaling factors released by adipose tissue, as well as cardiac adiposity and its relevance to cardiovascular diseases. Finally, we review the myocardial regenerative potential of adipose-tissue-derived stem cells. We believe that a thorough understanding of adipose tissue is of great clinical value.
Collapse
Affiliation(s)
- Mohamed Hassan
- Aswan Heart Center, Kasr El Hajjar Street, P. O. Box 81512, Aswan, Egypt
| | | | | |
Collapse
|
16
|
KASAMATSU ATSUSHI, IYODA MANABU, USUKURA KATSUYA, SAKAMOTO YOSUKE, OGAWARA KATSUNORI, SHIIBA MASASHI, TANZAWA HIDEKI, UZAWA KATSUHIRO. Gibberellic acid induces α-amylase expression in adipose-derived stem cells. Int J Mol Med 2012; 30:243-7. [DOI: 10.3892/ijmm.2012.1007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/19/2012] [Indexed: 11/06/2022] Open
|
17
|
Shu W, Shu YT, Dai CY, Zhen QZ. Comparing the biological characteristics of adipose tissue-derived stem cells of different persons. J Cell Biochem 2012; 113:2020-6. [DOI: 10.1002/jcb.24070] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
18
|
Lentiviral vectors and cardiovascular diseases: a genetic tool for manipulating cardiomyocyte differentiation and function. Gene Ther 2012; 19:642-8. [DOI: 10.1038/gt.2012.19] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
19
|
Mesenchymal stem cells and cardiovascular disease: a bench to bedside roadmap. Stem Cells Int 2012; 2012:175979. [PMID: 22315617 PMCID: PMC3270473 DOI: 10.1155/2012/175979] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/13/2011] [Indexed: 02/08/2023] Open
Abstract
In recent years, the incredible boost in stem cell research has kindled the expectations of both patients and physicians. Mesenchymal progenitors, owing to their availability, ease of manipulation, and therapeutic potential, have become one of the most attractive options for the treatment of a wide range of diseases, from cartilage defects to cardiac disorders. Moreover, their immunomodulatory capacity has opened up their allogenic use, consequently broadening the possibilities for their application. In this review, we will focus on their use in the therapy of myocardial infarction, looking at their characteristics, in vitro and in vivo mechanisms of action, as well as clinical trials.
Collapse
|
20
|
Tracking long-term survival of intramyocardially delivered human adipose tissue-derived stem cells using bioluminescence imaging. Mol Imaging Biol 2011; 13:633-45. [PMID: 20730500 DOI: 10.1007/s11307-010-0392-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE Transplantation of a regenerative cell population derived from human subcutaneous adipose tissue (hASCs) for cardiac regeneration represents a promising therapy due to the capacity of these cells for proliferation and differentiation. Understanding the fate of injected hASCs would help to understand how hASCs work in vivo. The aim of this study was to track the long-term fate, including survival, differentiation, proliferation, apoptosis, migration, and growth factor secretion of intramyocardially injected hASCs following experimental acute myocardial infarction in an immunodeficient mouse model. METHODS Myocardial infarction was experimentally induced in severe combined immunodeficient mice by permanent ligation of the left anterior descending coronary artery. Lentivirally labeled hASCs (5 × 10(5); expressing green fluorescence protein [GFP] and luciferase) were injected into the peri-infarct region. Colony formation, growth kinetics, and differentiation of transduced hASCs were analyzed in vitro and compared to those of untransduced hASCs. The survival and migration of injected hASCs were tracked by luciferase-based bioluminescence imaging for 10 weeks. Immunofluorescence and terminal deoxynucleotidyl transferase dUTP nick end labeling staining were used to assess differentiation, proliferation, growth factor expression, or apoptosis of grafted hASCs in infarcted hearts and potential distribution to other tissues. RESULTS Lentivirus transduction and GFP and luciferase expression did not influence proliferation or differentiation of hASCs. Bioluminescence imaging demonstrated that injected hASCs survived in infarcted hearts during the follow-up of 10 weeks. Immunofluorescence confirmed that hASCs engrafted in ischemic hearts expressed bFGF and IGF-1, and did not migrate into other organs. Of all engrafted hASCs, 3.5% differentiated into cardiomyocytes or endothelial cells. Other cells maintained their proliferative potential or underwent apoptosis. CONCLUSION Luciferase-based bioluminescence imaging allows long-term tracking of intramyocardially injected hASCs in living mice. The hASCs might enhance function of injured hearts through long-term engraftment, growth factor secretion, and transdifferentiation to cardiomyocytes and endothelial cells.
Collapse
|
21
|
Lindroos B, Suuronen R, Miettinen S. The potential of adipose stem cells in regenerative medicine. Stem Cell Rev Rep 2011; 7:269-91. [PMID: 20853072 DOI: 10.1007/s12015-010-9193-7] [Citation(s) in RCA: 321] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Adipose stem cells (ASCs) are an attractive and abundant stem cell source with therapeutic applicability in diverse fields for the repair and regeneration of acute and chronically damaged tissues. Importantly, unlike the human bone marrow stromal/stem stem cells (BMSCs) that are present at low frequency in the bone marrow, ASCs can be retrieved in high number from either liposuction aspirates or subcutaneous adipose tissue fragments and can easily be expanded in vitro. ASCs display properties similar to that observed in BMSCs and, upon induction, undergo at least osteogenic, chondrogenic, adipogenic and neurogenic, differentiation in vitro. Furthermore, ASCs have been shown to be immunoprivileged, prevent severe graft-versus-host disease in vitro and in vivo and to be genetically stable in long-term culture. They have also proven applicability in other functions, such as providing hematopoietic support and gene transfer. Due to these characteristics, ASCs have rapidly advanced into clinical trials for treatment of a broad range of conditions. As cell therapies are becoming more frequent, clinical laboratories following good manufacturing practices are needed. At the same time as laboratory processes become more extensive, the need for control in the processing laboratory grows consequently involving a greater risk of complications and possibly adverse events for the recipient. Therefore, the safety, reproducibility and quality of the stem cells must thoroughly be examined prior to extensive use in clinical applications. In this review, some of the aspects of examination on ASCs in vitro and the utilization of ASCs in clinical studies are discussed.
Collapse
Affiliation(s)
- Bettina Lindroos
- Regea-Institute for Regenerative Medicine, University of Tampere and Tampere University Hospital, Tampere, Finland.
| | | | | |
Collapse
|
22
|
|
23
|
Gimble JM, Grayson W, Guilak F, Lopez MJ, Vunjak-Novakovic G. Adipose tissue as a stem cell source for musculoskeletal regeneration. Front Biosci (Schol Ed) 2011; 3:69-81. [PMID: 21196358 DOI: 10.2741/s133] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Adipose tissue is an abundant, easily accessible, and reproducible cell source for musculo-skeletal regenerative medicine applications. Initial derivation steps yield a heterogeneous population of cells of stromal vascular fraction (SVF) cells. Subsequent adherent selection of the SVF results in a relatively homogeneous population of adipose-derived stromal/stem cells (ASCs) capable of adipogenic, chondrogenic, myogenic, and osteogenic differentiation in vitro on scaffolds in bioreactors and in vivo in pre-clinical animal models. Unlike hematopoietic cells, ASCs do not elicit a robust lymphocyte reaction and instead release immunosuppressive factors, such as prostaglandin E2. These immunomodulatory features suggest that allogeneic and autologous ASCs will engraft successfully for tissue regeneration purposes. The differentiation and expansion potential of ASCs can be modified by growth factors, bio-inductive scaffolds, and bioreactors providing environmental control and biophysical stimulation. Gene therapy approaches using lentiviral transduction can be used to direct differentiation of ASCs to particular lineages. We discuss the utility of ASCs for musculo-skeletal tissue repair and some of the technologies that can be implemented to unlock the full regenerative potential of these highly valuable cells.
Collapse
Affiliation(s)
- Jeffrey M Gimble
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | | | | | | | | |
Collapse
|
24
|
Mazo M, Gavira JJ, Pelacho B, Prosper F. Adipose-derived Stem Cells for Myocardial Infarction. J Cardiovasc Transl Res 2010; 4:145-53. [DOI: 10.1007/s12265-010-9246-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 11/15/2010] [Indexed: 02/06/2023]
|
25
|
Bai X, Alt E. Myocardial regeneration potential of adipose tissue-derived stem cells. Biochem Biophys Res Commun 2010; 401:321-6. [PMID: 20833143 DOI: 10.1016/j.bbrc.2010.09.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 09/05/2010] [Indexed: 12/17/2022]
Abstract
Various tissue resident stem cells are receiving attention from basic scientists and clinicians as they hold promise for myocardial regeneration. For practical reasons, adipose tissue-derived stem cells (ASCs) are attractive cells for clinical application in repairing damaged myocardium based on the following advantages: abundant adipose tissue in most patients and easy accessibility with minimally invasive lipoaspiration procedure. Several recent studies have demonstrated that both cultured and freshly isolated ASCs could improve cardiac function in animal model of myocardial infarction. The mechanisms underlying the beneficial effect of ASCs on myocardial regeneration are not fully understood. Growing evidence indicates that transplantation of ASCs improve cardiac function via the differentiation into cardiomyocytes and vascular cells, and through paracrine pathways. Paracrine factors secreted by injected ASCs enhance angiogenesis, reduce cell apoptosis rates, and promote neuron sprouts in damaged myocardium. In addition, Injection of ASCs increases electrical stability of the injured heart. Furthermore, there are no reported cases of arrhythmia or tumorigenesis in any studies regarding myocardial regeneration with ASCs. This review summarizes the characteristics of both cultured and freshly isolated stem cells obtained from adipose tissue, their myocardial regeneration potential, and the underlying mechanisms for beneficial effect on cardiac function, and safety issues.
Collapse
Affiliation(s)
- Xiaowen Bai
- Department of Molecular Pathology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe, Houston, TX 77030, USA
| | | |
Collapse
|
26
|
Isolation, identification and multipotential differentiation of mouse adipose tissue-derived stem cells. Tissue Cell 2010; 42:211-6. [DOI: 10.1016/j.tice.2010.04.003] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 04/03/2010] [Accepted: 04/07/2010] [Indexed: 11/19/2022]
|
27
|
Perán M, Marchal JA, López E, Jiménez-Navarro M, Boulaiz H, Rodríguez-Serrano F, Carrillo E, Sánchez-Espin G, de Teresa E, Tosh D, Aranega A. Human cardiac tissue induces transdifferentiation of adult stem cells towards cardiomyocytes. Cytotherapy 2010; 12:332-7. [PMID: 20230311 DOI: 10.3109/14653240903548202] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS The goal was to induce the transdifferentiation (or conversion) of human adipose-derived stem cells to cardiomyocytes using an intracellular extract obtained from adult human heart tissue. METHODS Human adult stem cells from lipoaspirates were transiently permeabilized, exposed to human atrial extracts and allowed to recover in culture. RESULTS After 21 days, the cells acquired a cardiomyocyte phenotype, as demonstrated by morphologic changes (appearance of binucleate, striated cells and branching fibers), immunofluorescence detection of cardiac-specific markers (connexin-43, sarcomeric alpha-actinin, cardiac troponin I and T, and desmin) and the presence of cardiomyocyte-related genes analyzed by reverse transcription-polymerase chain reaction (cardiac myosin light chain 1, alpha-cardiac actin, cardiac troponin T and cardiac beta-myosin). CONCLUSIONS We have demonstrated for the first time that adult cardiomyocytes obtained from human donors retain the capacity to induce cardiomyocyte differentiation of mesenchymal stromal cells. The use of autologous extracts for reprogramming adult stem cells may have potential therapeutic implications for treating heart disease.
Collapse
Affiliation(s)
- Macarena Perán
- Department of Health Sciences, University of Jaén, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Icariin-mediated differentiation of mouse adipose-derived stem cells into cardiomyocytes. Mol Cell Biochem 2010; 344:1-9. [PMID: 20563742 DOI: 10.1007/s11010-010-0523-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 06/05/2010] [Indexed: 12/27/2022]
Abstract
In this study, we investigated the ability of mouse adipose-derived stem cells (ADSCs) to differentiate into a cardiac phenotype in vitro. Icariin (ICA) has previously been shown to induce cardiomyocyte (CM) differentiation of murine embryonic stem cells in vitro, but its effect on ADSCs remains unclear. We isolated ADSCs from white adipose tissue and analyzed selected surface antigens using flow cytometry. ADSCs and CMs were co-cultured in transwell plates, with or without the addition of either ICA or ICA plus the extracellular signal-regulated kinase (ERK) inhibitor PD98059. Cardiac-specific gene expression was examined by reverse transcription-polymerase chain reaction and western blotting. ICA facilitated differentiation of ADSCs into CMs that expressed cardiac-specific genes, including the transcription factors NKX-2.5, GATA-4, MLC-2v, α-actinin, and cardiac troponin-T. Expression of α-actinin, the Z band-constituting protein, was promoted by ICA in a dose- and time-dependent manner. ICA can induce ERK activation and cardiac-specific gene expression was partially inhibited by PD98059 after treatment with ICA. These results suggest that ICA-stimulated CM differentiation of ADSCs, and that it acted partially by activating ERK-dependent signaling pathways in vitro.
Collapse
|
29
|
Bursac N, Kirkton RD, McSpadden LC, Liau B. Characterizing functional stem cell-cardiomyocyte interactions. Regen Med 2010; 5:87-105. [PMID: 20017697 DOI: 10.2217/rme.09.69] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Despite the progress in traditional pharmacological and organ transplantation therapies, heart failure still afflicts 5.3 million Americans. Since June 2000, stem cell-based approaches for the prevention and treatment of heart failure have been pursued in clinics with great excitement; however, the exact mechanisms of how transplanted cells improve heart function remain elusive. One of the main difficulties in answering these questions is the limited ability to directly access and study interactions between implanted cells and host cardiomyocytes in situ. With the growing number of candidate cell types for potential clinical use, it is becoming increasingly more important to establish standardized, well-controlled in vitro and in situ assays to compare the efficacy and safety of different stem cells in cardiac repair. This article describes recent innovative methodologies to characterize direct functional interactions between stem cells and cardiomyocytes, aimed to facilitate the rational design of future cell-based therapies for heart disease.
Collapse
Affiliation(s)
- Nenad Bursac
- Department of Biomedical Engineering, Duke University, Room 136 Hudson Hall, Durham, NC 27708, USA.
| | | | | | | |
Collapse
|
30
|
Tandon N, Goh B, Marsano A, Chao PHG, Montouri-Sorrentino C, Gimble J, Vunjak-Novakovic G. Alignment and elongation of human adipose-derived stem cells in response to direct-current electrical stimulation. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2010; 2009:6517-21. [PMID: 19964171 DOI: 10.1109/iembs.2009.5333142] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In vivo, direct current electric fields are present during embryonic development and wound healing. In vitro, direct current (DC) electric fields induce directional cell migration and elongation. For the first time, we demonstrate that cultured human adipose tissue-derived stem cells (hASCs) respond to the presence of direct-current electric fields. Cells were stimulated for 2-4 hours with DC electric fields of 6 V/cm that were similar to those encountered in vivo post-injury. Upon stimulation, hASCs were observed to elongate and align perpendicularly to the applied electric field, disassemble gap junctions, and upregulate the expression of genes for connexin-43, thrombomodulin, vascular endothelial growth factor, and fibroblast growth factor. In separate related studies, human epicardial fat-derived stem cells (heASCs) were also observed to align and elongate. It is interesting that the morphological and phenotypic characteristics of mesenchymal stem cells derived both from liposuction aspirates and from cardiac fat can be modulated by direct current electric fields. In further studies, we will quantify the effects of the electrical fields in the context of wound healing.
Collapse
Affiliation(s)
- Nina Tandon
- Columbia University, Department of Biomedical Engineering, 622 west 168th Street, Vanderbilt Clinic 12-234, New York, NY 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Tandon N, Marsano A, Maidhof R, Numata K, Montouri-Sorrentino C, Cannizzaro C, Voldman J, Vunjak-Novakovic G. Surface-patterned electrode bioreactor for electrical stimulation. LAB ON A CHIP 2010; 10:692-700. [PMID: 20221556 PMCID: PMC3618471 DOI: 10.1039/b917743d] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
We present a microscale cell culture system with an interdigitated microarray of excimer-laser-ablated indium tin oxide electrodes for electrical stimulation of cultured cells. The system has been characterized in a range of geometeries and stimulation regimes via electrochemical impedance spectroscopy and used to culture primary cardiomyocytes and human adipose derived stem cells. Over 6 days of culture with electrical stimulation (2 ms duration, 1 Hz, 180 microm wide electrodes with 200 microm spacing), both cell types exhibited enhanced proliferation, elongation and alignment, and adipose derived stem cells exhibited higher numbers of Connexin-43-composed gap junctions.
Collapse
Affiliation(s)
- Nina Tandon
- Department of Biomedical Engineering, Columbia University, 622 west 168 Street, New York NY 10032, Fax: 1-212-305-4692; Tel: 1-212-305-2304
- Department of Electrical Engineering, The Cooper Union, 55 Astor Place, New York, NY 10003
| | - Anna Marsano
- Department of Biomedical Engineering, Columbia University, 622 west 168 Street, New York NY 10032, Fax: 1-212-305-4692; Tel: 1-212-305-2304
| | - Robert Maidhof
- Department of Biomedical Engineering, Columbia University, 622 west 168 Street, New York NY 10032, Fax: 1-212-305-4692; Tel: 1-212-305-2304
| | - Keiji Numata
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford MA 02155
| | | | - Christopher Cannizzaro
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford MA 02155
| | - Joel Voldman
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge MA 02139
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, 622 west 168 Street, New York NY 10032, Fax: 1-212-305-4692; Tel: 1-212-305-2304
| |
Collapse
|
32
|
Mazo M, Pelacho B, Prósper F. Stem cell therapy for chronic myocardial infarction. J Cardiovasc Transl Res 2010; 3:79-88. [PMID: 20560022 DOI: 10.1007/s12265-009-9159-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 11/23/2009] [Indexed: 11/29/2022]
Abstract
Although recent advances for the treatment of myocardial infarction have dramatically increased the rate of survival after the ischemic event, this has also led to a rise in the number of chronic patients, making the finding of a suitable therapy a compulsory subject for modern medicine. Over the last decade, stem cells have been a promise for the cure of several diseases not only due to their plasticity but also to their capacity to act in a paracrine manner and influence the affected tissue, prompting the launching of several clinical trials. In spite of the knowledge already acquired, stem cell application to chronically infarcted hearts has been much less approached than its acute counterpart. Through this review, we will focus in stem cell therapy in animal models of chronic myocardial infarction: cell types employed, functional results, mechanisms analyzed, and questions raised.
Collapse
Affiliation(s)
- Manuel Mazo
- Hematology and Cell Therapy, Clinica Universidad de Navarra, and Division of Cancer, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | | | | |
Collapse
|
33
|
Adipose Tissue-derived Stem Cells Differentiated into Cardiomyocytes in Cardiac Microenvironment*. PROG BIOCHEM BIOPHYS 2009. [DOI: 10.3724/sp.j.1206.2008.00599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
Bai X, Yan Y, Song YH, Seidensticker M, Rabinovich B, Metzele R, Bankson JA, Vykoukal D, Alt E. Both cultured and freshly isolated adipose tissue-derived stem cells enhance cardiac function after acute myocardial infarction. Eur Heart J 2009; 31:489-501. [PMID: 20037143 DOI: 10.1093/eurheartj/ehp568] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AIMS We assessed whether freshly isolated human adipose tissue-derived cells (fhADCs) or cultured human adipose tissue-derived stem cells (hASCs) have beneficial effects on cardiac function after myocardial infarction (MI), whether the injected cells can survive long term, and whether their effects result from direct differentiation or paracrine mechanisms. METHODS AND RESULTS Myocardial infarction was experimentally induced in severe combined immunodeficient mice, and either fhADCs, cultured hASCs, or phosphate-buffered saline was injected into the peri-infarct region. Myocardial function improved significantly in mice treated with hASCs or fhADCs 4 weeks after MI. Immunofluorescence revealed that grafted hASCs and fhADCs underwent cardiomyogenic differentiation pathway, as indicated by expression of connexin 43 and troponin I in a fusion-independent manner. Some of the injected cells integrated with host cardiomyocytes through connexin 43, and others were incorporated into newly formed vessels. Human adipose tissue-derived stem cells survived in injured hearts up to 4 months, as detected by luciferase-based bioluminescence imaging. Vascular density was significantly increased, and fewer apoptotic cells were present in the peri-infarct region of cell-injected mice. CONCLUSION This is the first study to systematically compare the effects of fhADCs and hASCs on myocardial regeneration. Both cell types engraft into infarcted myocardium, survive, and improve myocardial function, suggesting that fhADCs, like hASCs, are a promising alternative cell source for myocardial repair after MI.
Collapse
Affiliation(s)
- Xiaowen Bai
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, SCRB2, Unit 951, 7435 Fannin Street, Houston, TX 77054, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Grajek S, Popiel M, Gil L, Breborowicz P, Lesiak M, Czepczynski R, Sawinski K, Straburzynska-Migaj E, Araszkiewicz A, Czyz A, Kozlowska-Skrzypczak M, Komarnicki M. Influence of bone marrow stem cells on left ventricle perfusion and ejection fraction in patients with acute myocardial infarction of anterior wall: randomized clinical trial: Impact of bone marrow stem cell intracoronary infusion on improvement of microcirculation. Eur Heart J 2009; 31:691-702. [DOI: 10.1093/eurheartj/ehp536] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
36
|
Sliwa A, Balwierz A, Kiec-Wilk B, Polus A, Knapp A, Dembinska-Kiec A. Differentiation of human adipose tissue SVF cells into cardiomyocytes. GENES & NUTRITION 2009; 4:195-8. [PMID: 19533197 PMCID: PMC2745743 DOI: 10.1007/s12263-009-0127-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 05/27/2009] [Indexed: 11/27/2022]
Abstract
Progenitor cells have been extensively studied and therapeutically applied in tissue reconstructive therapy. Stromal vascular fraction (SVF) cells, which are derived from adipose tissue, may represent a potential source of the cells which undergo phenotypical differentiation into many lineages both in vitro as well as in vivo. The goal of this study was to check whether human SVF cells may differentiate into cardiomyocyte-like entities. Human SVF cells were induced to differentiate by their incubation in Methocult medium in the presence of SCF, IL-3 and IL-6. Morphological transformation of the cells was monitored using optical light microscope, whereas changes in expression of the genes typical for cardiac phenotype were measured by qRT-PCR. Incubation of the human SVF cells in the medium that promotes cardiomyocyte differentiation in vitro resulted in formation of myotubule-like structures accompanied by up-regulation of the myocardium-characteristic genes, such as GATA, MEF2C, MYOD1, but not ANP. Human SVF cells differentiate into cardiomyocyte-like cells in the presence of the certain set of myogenesis promoting cytokines.
Collapse
Affiliation(s)
- Agnieszka Sliwa
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, 15a Kopernika Str., 31-501 Krakow, Poland
| | - A. Balwierz
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, 15a Kopernika Str., 31-501 Krakow, Poland
- Postgraduate School of Molecular Medicine, 61 Żwirki i Wigury Str., 02-091 Warsaw, Poland
| | - B. Kiec-Wilk
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, 15a Kopernika Str., 31-501 Krakow, Poland
| | - A. Polus
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, 15a Kopernika Str., 31-501 Krakow, Poland
| | - A. Knapp
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, 15a Kopernika Str., 31-501 Krakow, Poland
| | - A. Dembinska-Kiec
- Department of Clinical Biochemistry, Collegium Medicum, Jagiellonian University, 15a Kopernika Str., 31-501 Krakow, Poland
| |
Collapse
|
37
|
de Villiers JA, Houreld N, Abrahamse H. Adipose derived stem cells and smooth muscle cells: implications for regenerative medicine. Stem Cell Rev Rep 2009; 5:256-65. [PMID: 19669954 DOI: 10.1007/s12015-009-9084-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 07/22/2009] [Indexed: 01/18/2023]
Abstract
The treatment of chronic wounds and other damaged tissues and organs remains a difficult task, in spite of greater adherence to recognised standards of care and a better understanding of pathophysiologic principles. Adipose derived stem cells (ADSCs), with their proliferative and impressive differentiation potential, may be used in the future in autologous cell therapy or grafting to replace damaged tissues. At this point in time, transplanted tissues are often rejected by the body. Autologous grafting would eliminate this problem. ADSCs are able to differentiate into a number of cells in vitro, for example smooth muscle cells (SMCs), when treated with lineage specific factors. SMCs play a key role in physiology and pathology as they form the principle layer of all SMC tissues. Smooth muscle biopsies are often impractical and morbid, and often lead to a low cell harvest. It has also been shown that SMCs derived from a diseased organ can lead to abnormal cells. Therefore, there is a great need for alternative sources of healthy SMCs. The use of ADSCs for cell-based tissue engineering (TE) represents a promising alternative for smooth muscle repair. This review discusses the potential uses of ADSCs and SMCs in regenerative medicine, and the potential of ADSCs to be differentiated into functional SMCs for TE and regenerative cellular therapies to repair diseased organs.
Collapse
Affiliation(s)
- Jennifer Anne de Villiers
- Laser Research Group, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein, 2028, South Africa
| | | | | |
Collapse
|
38
|
Schimrosczyk K, Song Y, Vykoukal J, Vykoukal D, Bai X, Krohn A, Freyberg S, Alt EU. Liposome‐mediated transfection with extract from neonatal rat cardiomyocytes induces transdifferentiation of human adipose‐derived stem cells into cardiomyocytes. Scandinavian Journal of Clinical and Laboratory Investigation 2009; 68:464-72. [DOI: 10.1080/00365510701836907] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
39
|
Alt E, Pinkernell K, Scharlau M, Coleman M, Fotuhi P, Nabzdyk C, Matthias N, Gehmert S, Song YH. Effect of freshly isolated autologous tissue resident stromal cells on cardiac function and perfusion following acute myocardial infarction. Int J Cardiol 2009; 144:26-35. [PMID: 19443059 DOI: 10.1016/j.ijcard.2009.03.124] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2008] [Revised: 02/11/2009] [Accepted: 03/26/2009] [Indexed: 02/08/2023]
Abstract
BACKGROUND The aim of this study was to investigate the effect of intracoronary administration of freshly isolated, uncultured autologous tissue-derived stromal cells on cardiac function and perfusion after acute infarction in pigs. METHODS A transmural myocardial infarction in a porcine model was induced by occlusion of the mid LAD with an angioplasty balloon for 3 h. Upon reperfusion, freshly isolated, uncultured autologous stromal cells (1.5×10⁶ cells/kg) or control solution was injected into the infarct artery. Cardiac function and area at risk were determined by (99m)Tc-SPECT. RESULTS Eight weeks after infarction, cell treated pigs showed a 20% smaller myocardial perfusion defect compared to control animals (35±9% vs. 44±5% of LV, treated vs. control, respectively, p<0.05). The reduction of the perfusion defect was associated with a significantly higher myocardial salvage index in the cell group as well as a significant increase in ejection fraction compared to control (EF at 8 weeks 43±7% vs. 35±3%, treated vs. control, respectively, p<0.05). This functional improvement was reflected by an increased wall thickness of the infarct and border zone in the treated group (11.2±2.2 mm) compared to control (8.6±1.6 mm, p<0.05) as well as an increased capillary density in the border zone (treated vs. control; 41.6±17.9 vs. 32.9±12.6 capillaries per 0.1 mm², p<0.05). CONCLUSIONS This study demonstrates for the first time that recovery and intracoronary delivery of uncultured autologous tissue derived stromal cells at time of vessel reperfusion is feasible and improves ventricular function.
Collapse
Affiliation(s)
- Eckhard Alt
- Department of Medicine, Section of Cardiology, Tulane University Health Sciences Center, New Orleans, LA 70112, United States.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kuroda H, Kutner RH, Bazan NG, Reiser J. Simplified lentivirus vector production in protein-free media using polyethylenimine-mediated transfection. J Virol Methods 2009; 157:113-21. [DOI: 10.1016/j.jviromet.2008.11.021] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2008] [Revised: 11/25/2008] [Accepted: 11/27/2008] [Indexed: 02/07/2023]
|
41
|
Heterogeneity of Mitochondrial Potential as a Marker for Isolation of Pure Cardiomyoblast Population. Bull Exp Biol Med 2009; 146:506-11. [DOI: 10.1007/s10517-009-0327-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
42
|
ADSCs differentiated into cardiomyocytes in cardiac microenvironment. Mol Cell Biochem 2008; 324:117-29. [PMID: 19107327 DOI: 10.1007/s11010-008-9990-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2008] [Accepted: 12/11/2008] [Indexed: 10/21/2022]
Abstract
The microenvironment plays a critical role in directing the progression of stem cells into differentiated cells. So we investigated the role that cardiac microenvironment plays in directing this differentiation process. Adipose tissue-derived stem cells (ADSCs) were cultured with cardiomyocytes directly ("co-culture directly") or by cell culture insert ("co-culture indirectly"). For co-culture indirectly, differentiated ADSCs were collected and identified. For co-culture directly, ADSCs were labeled with carboxyfluorescein succinimidyl ester (CFSE), Fluorescence-activated cell sorting was used to extract and examine the differentiated ADSCs. The ultrastructure and the expression of cardiac specific proteins and genes were analyzed by SEM, TEM, western blotting, and RT-PCR, respectively. Differentiated ADSCs experienced the co-culture presented cardiac ultrastructure and expressed cardiac specific genes and proteins, and the fractions of ADSCs expressing these markers by co-culture directly were higher than those of co-culture indirectly. These data indicate that in addition to soluble signaling molecules, direct cell-to-cell contact is obligatory in relaying the external cues of the microenvironment controlling the differentiation of ADSCs to cardiomyocytes.
Collapse
|
43
|
Kuroda H, Kutner RH, Bazan NG, Reiser J. A comparative analysis of constitutive and cell-specific promoters in the adult mouse hippocampus using lentivirus vector-mediated gene transfer. J Gene Med 2008; 10:1163-75. [PMID: 18773500 DOI: 10.1002/jgm.1249] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Viral vectors provide powerful tools for transgene delivery to the mammalian brain to assess the effects of therapeutic proteins, antisense RNAs or small interfering RNAs. A key advantage of such approaches is that specific brain regions implicated in a particular disease can be independently targeted. METHODS To optimize transgene expression in sub-regions of the mouse hippocampus and with a view towards devising gene therapy strategies for Alzheimer's disease, we designed lentivirus-based reporter vectors bearing various promoters, including constitutive and cell-specific promoters. Furthermore, we devised methods allowing a side-by-side comparison of transgene expression levels in neural cells both in vitro and in vivo. RESULTS Following stereotaxic injection into the adult mouse hippocampus, titer-adjusted lentiviral vectors bearing constitutive promoters resulted in robust and sub-region-specific transgene expression. Our results show that the human CMV-IE promoter resulted in efficient transgene expression in the entire hippocampus whereas transgene expression mediated by the hybrid hEF1alpha/HTLV promoter was limited mainly in the dentate gyrus and the CA2/3 region. Finally, the neuron-specific human synapsin I promoter was particularly effective in the dentate gyrus. CONCLUSIONS These findings indicate that subregion-specific transgene expression in the hippocampus can be achieved following lentivirus vector-mediated gene transfer.
Collapse
Affiliation(s)
- Hitoshi Kuroda
- Gene Therapy Program, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | | | | | | |
Collapse
|
44
|
van Dijk A, Niessen HWM, Zandieh Doulabi B, Visser FC, van Milligen FJ. Differentiation of human adipose-derived stem cells towards cardiomyocytes is facilitated by laminin. Cell Tissue Res 2008; 334:457-67. [PMID: 18989703 DOI: 10.1007/s00441-008-0713-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 09/25/2008] [Indexed: 11/30/2022]
Abstract
Adipose-derived stem cells (ASCs) are promising candidates for therapy in myocardial infarction (MI). However, the frequency of human ASCs that differentiate towards cardiomyocytes is low. We hypothesized that adherence to extracellular matrix molecules that are upregulated after MI might increase human stem cell differentiation towards cardiomyocytes. We analysed putative ASC differentiation on fibronectin-coated, laminin-coated and uncoated culture plates. Expression of cardiac markers in cells was analysed 1, 3 and 5 weeks after stimulation with 5-aza-2-deoxycytidine. After 1 week, mRNA expression of myosin light chain-2alpha (MLC-2alpha), an early marker in cardiomyocyte development, was increased significantly in treated cells, independent of coating. At 5 weeks, however, mRNA expression of the late cardiomyocyte development marker SERCA2alpha was only significantly increased in 5-aza-2-deoxycytidine-treated cells cultured on laminin. Significantly higher numbers of cells were immunopositive for MLC-2alpha in cultures of treated cells grown on laminin-coated wells, when compared with cultures of treated cells grown on uncoated wells, both at 1 week and at 5 weeks. Furthermore, after 3 weeks, significantly more alpha-actinin- and desmin-positive cells were detected after treatment with 5-aza-2-deoxycytidine, but only in uncoated wells. After 5 weeks, however, the number of desmin-positive cells was only significantly increased after treatment of cells with 5-aza-2-deoxycytidine and culture on laminin (61% positive cells). Thus, we have found that a high percentage of human ASCs can be differentiated towards cardiomyocytes; this effect can be improved by laminin, especially during late differentiation.
Collapse
Affiliation(s)
- A van Dijk
- Department of Pathology, VU University Medical Centre, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
45
|
Phillips MI, Tang YL, Pinkernell K. Stem cell therapy for heart failure: the science and current progress. Future Cardiol 2008; 4:285-98. [DOI: 10.2217/14796678.4.3.285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cell therapy, particularly with stem cells, has created great interest as a solution to the fact that there are limited treatments for postischemic heart disease and none that can regenerate damaged heart cells to strengthen cardiac performance. From the first efforts with myoblasts to recent clinical trials with bone marrow-derived stem cells, early reports of cell therapy suggest improvement in cardiac performance as well as other clinical end points. Based on these exciting but tentative results, other stem cell types are being explored for their particular advantages as a source of adult stem cells. Autologous adipose-derived stem cells are multilinear and can be obtained relatively easily in large quantities from patients; cardiac-derived stem cells are highly appropriate for engraftment in their natural niche, the heart. Human umbilical cord blood cells are potentially forever young and allogenic adult mesenchymal stem cells appear not to evoke the graft versus host reaction. Human embryonic stem cells are effective and can be scaled up for supply purposes. The recent discovery of induced pluripotentcy in human adult stem cells, with only three transcription factor genes, opens a whole new approach to making autologous human pluripotent stem cells from skin or other available tissues. Despite the excitement, stem cells may have to be genetically modified with heme oxygenase, Akt or other genes to survive transplantation in a hypoxic environment. Homing factors and hormones secreted from transplanted stem cells may be more important than cells if they provide the necessary stimulus to trigger cardiac regrowth to replace scar tissue. As we await results from larger and more prolonged clinical trials, the science of stem cell therapy in cardiac disease keeps progressing.
Collapse
Affiliation(s)
- M Ian Phillips
- Keck Graduate Institute, Stem Cell Labs, 535 Watson Drive, Claremont, CA 91711, USA
| | - Yao Liang Tang
- Keck Graduate Institute, Stem Cell Labs, 535 Watson Drive, Claremont, CA 91711, USA
| | - Kai Pinkernell
- Cytori Therapeutics Inc., 3020 Callan Road, San Diego, CA 92121, USA
| |
Collapse
|
46
|
Kirkton RD, Bursac N. Genetic engineering and stem cells: combinatorial approaches for cardiac cell therapy. IEEE ENGINEERING IN MEDICINE AND BIOLOGY MAGAZINE : THE QUARTERLY MAGAZINE OF THE ENGINEERING IN MEDICINE & BIOLOGY SOCIETY 2008; 27:85-8. [PMID: 18519188 PMCID: PMC2722747 DOI: 10.1109/memb.2008.922356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
47
|
Klinger R, Bursac N. Cardiac cell therapy in vitro: reproducible assays for comparing the efficacy of different donor cells. IEEE ENGINEERING IN MEDICINE AND BIOLOGY MAGAZINE : THE QUARTERLY MAGAZINE OF THE ENGINEERING IN MEDICINE & BIOLOGY SOCIETY 2008; 27:72-80. [PMID: 18270054 PMCID: PMC2715011 DOI: 10.1109/memb.2007.913849] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
48
|
Sadat S, Gehmert S, Song YH, Yen Y, Bai X, Gaiser S, Klein H, Alt E. The cardioprotective effect of mesenchymal stem cells is mediated by IGF-I and VEGF. Biochem Biophys Res Commun 2007; 363:674-9. [PMID: 17904522 DOI: 10.1016/j.bbrc.2007.09.058] [Citation(s) in RCA: 230] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2007] [Accepted: 09/06/2007] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggests that adipose tissue-derived stem cells (ASCs) can be used for the treatment of ischemic heart diseases. However, the mechanisms underlying their therapeutic effects have not been clearly defined. In this study cytokines released by ASCs were detected by ELISA and pro-angiogenic effects were assessed by tube formation assay. To define the anti-apoptotic effect of ASCs, neonatal rat cardiomyocytes were subjected to hypoxia condition in a co-culture system. Our data show that ASCs secrete significant amounts of VEGF (810.65+/-56.92 pg/microg DNA) and IGF-I (328.33+/-22.7 pg/microg DNA). Cardiomyocytes apoptosis was significantly prevented by ASCs and 62.5% of the anti-apoptotic effect was mediated by IGF-I and 34.2% by VEGF. ASCs promoted endothelial cell tube formation by secreting VEGF. In conclusion we demonstrated that ASCs have a marked impact on anti-apoptosis and angiogenesis and helps to explain data of stem cells benefit without transdifferentiation.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Apoptosis/physiology
- Cell Hypoxia
- Cell Line
- Cell Survival/drug effects
- Cell Survival/physiology
- Cells, Cultured
- Coculture Techniques
- Endothelial Cells/cytology
- Endothelial Cells/drug effects
- Endothelial Cells/physiology
- Enzyme-Linked Immunosorbent Assay
- Humans
- Insulin-Like Growth Factor I/genetics
- Insulin-Like Growth Factor I/metabolism
- Insulin-Like Growth Factor I/pharmacology
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/metabolism
- Myocytes, Cardiac/cytology
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/physiology
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/physiology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/genetics
- Rats
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
- Transfection
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/pharmacology
Collapse
Affiliation(s)
- Sanga Sadat
- Department of Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Palpant NJ, Yasuda SI, MacDougald O, Metzger JM. Non-canonical Wnt signaling enhances differentiation of Sca1+/c-kit+ adipose-derived murine stromal vascular cells into spontaneously beating cardiac myocytes. J Mol Cell Cardiol 2007; 43:362-70. [PMID: 17706246 PMCID: PMC2048991 DOI: 10.1016/j.yjmcc.2007.06.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 06/08/2007] [Accepted: 06/25/2007] [Indexed: 02/04/2023]
Abstract
Recent reports have described a stem cell population termed stromal vascular cells (SVCs) derived from the stromal vascular fraction of adipose tissue, which are capable of intrinsic differentiation into spontaneously beating cardiomyocytes in vitro. The objective of this study was to further define the cardiac lineage differentiation potential of SVCs in vitro and to establish methods for enriching SVC-derived beating cardiac myocytes. SVCs were isolated from the stromal vascular fraction of murine adipose tissue. Cells were cultured in methylcellulose-based murine stem cell media. Analysis of SVC-derived beating myocytes included Western blot and calcium imaging. Enrichment of acutely isolated SVCs was carried out using antibody-tagged magnetic nanoparticles, and pharmacologic manipulation of Wnt and cytokine signaling. Under initial media conditions, spontaneously beating SVCs expressed both cardiac developmental and adult protein isoforms. Functionally, this specialized population can spontaneously contract and pace under field stimulation and shows the presence of coordinated calcium transients. Importantly, this study provides evidence for two independent mechanisms of enriching the cardiac differentiation of SVCs. First, this study shows that differentiation of SVCs into cardiac myocytes is augmented by non-canonical Wnt agonists, canonical Wnt antagonists, and cytokines. Second, SVCs capable of cardiac lineage differentiation can be enriched by selection for stem cell-specific membrane markers Sca1 and c-kit. Adipose-derived SVCs are a unique population of stem cells that show evidence of cardiac lineage development making them a potential source for stem cell-based cardiac regeneration studies.
Collapse
Affiliation(s)
- Nathan J Palpant
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109-0622, USA
| | | | | | | |
Collapse
|
50
|
Bai X, Ma J, Pan Z, Song YH, Freyberg S, Yan Y, Vykoukal D, Alt E. Electrophysiological properties of human adipose tissue-derived stem cells. Am J Physiol Cell Physiol 2007; 293:C1539-50. [PMID: 17687001 DOI: 10.1152/ajpcell.00089.2007] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human adipose tissue-derived stem cells (hASCs) represent a potentially valuable cell source for clinical therapeutic applications. The present study was designed to investigate properties of ionic channel currents present in undifferentiated hASCs and their impact on hASCs proliferation. The functional ion channels in hASCs were analyzed by whole-cell patch-clamp recording and their mRNA expression levels detected by RT-PCR. Four types of ion channels were found to be present in hASCs: most of the hASCs (73%) showed a delayed rectifier-like K(+) current (I(KDR)); Ca(2+)-activated K(+) current (I(KCa)) was detected in examined cells; a transient outward K(+) current (I(to)) was recorded in 19% of the cells; a small percentage of cells (8%) displayed a TTX-sensitive transient inward sodium current (I(Na.TTX)). RT-PCR results confirmed the presence of ion channels at the mRNA level: Kv1.1, Kv2.1, Kv1.5, Kv7.3, Kv11.1, and hEAG1, possibly encoding I(KDR); MaxiK, KCNN3, and KCNN4 for I(KCa); Kv1.4, Kv4.1, Kv4.2, and Kv4.3 for I(to) and hNE-Na for I(Na.TTX). The I(KDR) was inhibited by tetraethyl ammonium (TEA) and 4-aminopyridine (4-AP), which significantly reduced the proliferation of hASCs in a dose-dependent manner (P < 0.05), as suggested by bromodeoxyurindine (BrdU) incorporation. Other selective potassium channel blockers, including linopiridine, iberiotoxin, clotrimazole, and apamin also significantly inhibited I(KDR). TTX completely abolished I(Na.TTX). This study demonstrates for the first time that multiple functional ion channel currents such as I(KDR), I(KCa), I(to), and I(Na.TTX) are present in undifferentiated hASCs and their potential physiological function in these cells as a basic understanding for future in vitro experiments and in vivo clinical investigations.
Collapse
Affiliation(s)
- Xiaowen Bai
- Department of Molecular Pathology, University of Texas, MD Anderson Cancer Center, Houston, Texas 77054, USA
| | | | | | | | | | | | | | | |
Collapse
|