共查询到20条相似文献,搜索用时 0 毫秒
1.
Xiaorong Feng Wen Yin Jialing Wang Li Feng Y James Kang 《Experimental biology and medicine (Maywood, N.J.)》2021,246(1):97
Previous studies demonstrated that mitochondrial fission arguments the stemness of bone marrow-derived mesenchymal stem cells (BMSCs). Because mitophagy is critical in removing damaged or surplus mitochondrial fragments and maintaining mitochondrial integrity, the present study was undertaken to test the hypothesis that mitophagy is involved in mitochondrial fission-enhanced stemness of BMSCs. Primary cultures of rat BMSCs were treated with tyrphostin A9 (TA9, a potent inducer of mitochondrial fission) to increase mitochondrial fission, which was accompanied by enhanced mitophagy as defined by increased co-staining of MitoTracker Green for mitochondria and LysoTracker Deep Red for lysosomes, as well as the increased co-localization of autophagy markers (LC3B, P62) and mitochondrial marker (Tom20). A mitochondrial uncoupler, carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP) was used to promote mitophagy, which was confirmed by an increased co-localization of mitochondrial and lysosome biomarkers. The argumentation of mitophagy was associated with enhanced stemness of BMSCs as defined by increased expression of stemness markers Oct4 and Sox2, and enhanced induction of BMSCs to adipocytes or osteocytes. Conversely, transfection of BMSCs with siRNA targeting mitophagy-essential genes Pink1/Prkn led to diminished stemness of the stem cells, as defined by depressed stemness markers. Importantly, concomitant promotion of mitochondrial fission and inhibition of mitophagy suppressed the stemness of BMSCs. These results thus demonstrate that mitophagy is critically involved in mitochondrial fission promotion of the stemness of BMSCs. 相似文献
2.
Shipeng Dang Huanbai Xu Congfeng Xu Wei Cai Qian Li Yiji Cheng Min Jin Ru-Xing Wang Yongde Peng Yi Zhang Changping Wu Xiaozhou He Bing Wan Yanyun Zhang 《Autophagy》2014,10(7):1301-1315
Mesenchymal stem cell (MSC)-based therapy is a promising approach to treat various inflammatory disorders including multiple sclerosis. However, the fate of MSCs in the inflammatory microenvironment is largely unknown. Experimental autoimmune encephalomyelitis (EAE) is a well-studied animal model of multiple sclerosis. We demonstrated that autophagy occurred in MSCs during their application for EAE treatment. Inflammatory cytokines, e.g., interferon gamma and tumor necrosis factor, induced autophagy in MSCs synergistically by inducing expression of BECN1/Beclin 1. Inhibition of autophagy by knockdown of Becn1 significantly improved the therapeutic effects of MSCs on EAE, which was mainly attributable to enhanced suppression upon activation and expansion of CD4+ T cells. Mechanistically, inhibition of autophagy increased reactive oxygen species generation and mitogen-activated protein kinase 1/3 activation in MSCs, which were essential for PTGS2 (prostaglandin-endoperoxide synthase 2 [prostaglandin G/H synthase and cyclooxygenase]) and downstream prostaglandin E2 expression to exert immunoregulatory function. Furthermore, pharmacological treatment of MSCs to inhibit autophagy increased their immunosuppressive effects on T cell-mediated EAE. Our findings indicate that inflammatory microenvironment-induced autophagy downregulates the immunosuppressive function of MSCs. Therefore, modulation of autophagy in MSCs would provide a novel strategy to improve MSC-based immunotherapy. 相似文献
3.
Katie L. Pricola Nastaran Z. Kuhn Hana Haleem‐Smith Yingjie Song Rocky S. Tuan 《Journal of cellular biochemistry》2009,108(3):577-588
Adult human mesenchymal stem cells (MSCs) hold promise for an increasing list of therapeutic uses due to their ease of isolation, expansion, and multi‐lineage differentiation potential. To maximize the clinical potential of MSCs, the underlying mechanisms by which MSC functionality is controlled must be understood. We have taken a deconstructive approach to understand the individual components in vitro, namely the role of candidate “stemness” genes. Our recent microarray gene expression profiling data suggest that interleukin‐6 (IL‐6) may contribute to the maintenance of MSCs in their undifferentiated state. In this study, we showed that IL‐6 gene expression is significantly higher in undifferentiated MSCs as compared to their chondrogenic, osteogenic, and adipogenic derivatives. Moreover, we found that MSCs secrete copious amounts of IL‐6 protein, which decreases dramatically during osteogenic differentiation. We further evaluated the role of IL‐6 for maintenance of MSC “stemness,” using a series of functional assays. The data showed that IL‐6 is both necessary and sufficient for enhanced MSC proliferation, protects MSCs from apoptosis, inhibits adipogenic and chondrogenic differentiation of MSCs, and increases the rate of in vitro wound healing of MSCs. We further identified ERK1/2 activation as the key pathway through which IL‐6 regulates both MSC proliferation and inhibition of differentiation. Taken together, these findings show for the first time that IL‐6 maintains the proliferative and undifferentiated state of bone marrow‐derived MSCs, an important parameter for the optimization of both in vitro and in vivo manipulation of MSCs. J. Cell. Biochem. 108: 577–588, 2009. Published 2009 Wiley‐Liss, Inc. 相似文献
4.
Zheng Y. Yu Dan Ma Zheng C. He Ping Liu Jun Huang Qin Fang Jiang Y. Zhao Ji S. Wang 《Experimental cell research》2018,362(1):28-42
Iron overload (IO) caused by frequent blood transfusion in hematological diseases has become a major concern. In this study, up-regulation of heme oxygenase-1 (HO-1), a protector against oxidative stress, was observed in bone marrow mesenchymal stem cells (BMMSCs) at the early stage of IO and had favorable prognosis in an IO mouse model. Given that the protective role of HO-1 in IO damage of BMMSCs was still unknown, the mechanism was explored in vitro and in vivo. BMMSCs were transfected with HO-1/siHO-1 in vitro, and the mouse model was established to further evaluate the effect of HO-1 on IO in vivo. As a result, HO-1 decreased the apoptotic rate of BMMSCs with IO through reducing intracellular reactive oxygen species (ROS) but increasing IL-10 secretion. In addition, IL-10 was mediated by HO-1 via the ERK pathway. Intracellular iron was down-regulated by hepcidin depending on IL-10. In conclusion, HO-1 protects BMMSCs from ROS by secreting IL-10 upon iron overload. 相似文献
5.
Jun-Lin Guan Anna Katharina Simon Mark Prescott Javier A. Menendez Fei Liu Fen Wang Chenran Wang Ernst Wolvetang Alejandro Vazquez-Martin Jue Zhang 《Autophagy》2013,9(6):830-849
Autophagy is a highly conserved cellular process by which cytoplasmic components are sequestered in autophagosomes and delivered to lysosomes for degradation. As a major intracellular degradation and recycling pathway, autophagy is crucial for maintaining cellular homeostasis as well as remodeling during normal development, and dysfunctions in autophagy have been associated with a variety of pathologies including cancer, inflammatory bowel disease and neurodegenerative disease. Stem cells are unique in their ability to self-renew and differentiate into various cells in the body, which are important in development, tissue renewal and a range of disease processes. Therefore, it is predicted that autophagy would be crucial for the quality control mechanisms and maintenance of cellular homeostasis in various stem cells given their relatively long life in the organisms. In contrast to the extensive body of knowledge available for somatic cells, the role of autophagy in the maintenance and function of stem cells is only beginning to be revealed as a result of recent studies. Here we provide a comprehensive review of the current understanding of the mechanisms and regulation of autophagy in embryonic stem cells, several tissue stem cells (particularly hematopoietic stem cells), as well as a number of cancer stem cells. We discuss how recent studies of different knockout mice models have defined the roles of various autophagy genes and related pathways in the regulation of the maintenance, expansion and differentiation of various stem cells. We also highlight the many unanswered questions that will help to drive further research at the intersection of autophagy and stem cell biology in the near future. 相似文献
6.
Karina D. Asensi Rodrigo S. Fortunato Danúbia S. dos Santos Thaísa S. Pacheco Danielle F. de Rezende Deivid C. Rodrigues Fernanda C. P. Mesquita Tais H. Kasai‐Brunswick Antonio C. Campos de Carvalho Denise P. Carvalho Adriana B. Carvalho Regina C. dos S. Goldenberg 《Journal of cellular and molecular medicine》2014,18(5):824-831
Properties of induced pluripotent stem cells (iPSC) have been extensively studied since their first derivation in 2006. However, the modification in reactive oxygen species (ROS) production and detoxification caused by reprogramming still needs to be further elucidated. The objective of this study was to compare the response of iPSC generated from menstrual blood–derived mesenchymal stem cells (mb‐iPSC), embryonic stem cells (H9) and adult menstrual blood–derived mesenchymal stem cells (mbMSC) to ROS exposure and investigate the effects of reprogramming on cellular oxidative stress (OS). mbMSC were extremely resistant to ROS exposure, however, mb‐iPSC were 10‐fold less resistant to H2O2, which was very similar to embryonic stem cell sensitivity. Extracellular production of ROS was also similar in mb‐iPSC and H9 and almost threefold lower than in mbMSC. Furthermore, intracellular amounts of ROS were higher in mb‐iPSC and H9 when compared with mbMSC. As the ability to metabolize ROS is related to antioxidant enzymes, we analysed enzyme activities in these cell types. Catalase and superoxide dismutase activities were reduced in mb‐iPSC and H9 when compared with mbMSC. Finally, cell adhesion under OS conditions was impaired in mb‐iPSC when compared with mbMSC, albeit similar to H9. Thus, reprogramming leads to profound modifications in extracellular ROS production accompanied by loss of the ability to handle OS. 相似文献
7.
The metabolic function of hepatocytes differentiated from human mesenchymal stem cells is inversely related to cellular glutathione levels 下载免费PDF全文
Abdolamir Allameh Hamidreza Ahmadi‐Ashtiani Mohammad Sajad Emami Aleagha Hossein Rastegar 《Cell biochemistry and function》2014,32(2):194-200
Differentiation of mesenchymal stem cells (MSCs) to hepatocytes‐like cells is associated with alteration in the level of reactive oxygen species (ROS) and antioxidant defense system. Here, we report the role of glutathione in the functions of hepatocytes derived from MSCs. The stem cells undergoing differentiation were treated with glutathione modifiers [buthionine sulfoxide (BSO) or N‐acetyl cysteine (NAC)], and hepatocytes were collected on day 14 of differentiation and analysed for their biological and metabolic functions. Differentiation process has been performed in presence of glutathione modifiers viz. BSO and NAC. Depending on the level of cellular glutathione, the proliferation rate of MSCs was affected. Glutathione depletion by BSO resulted in increased levels of albumin and ROS in hepatocytes. Whereas, albumin and ROS were inhibited in cells treated with glutathione precursor (NAC). The metabolic function of hepatocytes was elevated in BSO‐treated cells as judged by increased urea, transferrin, albumin, alanine transaminase and aspartate transaminase secretions in the media. However, the metabolic activity of the hepatocytes was inhibited when glutathione was increased by NAC. We conclude that the efficiency of metabolic function of hepatocytes is inversely related to the levels of cellular glutathione. These data may suggest a novel role of glutathione in regulation of metabolic function of hepatocytes. Copyright © 2013 John Wiley & Sons, Ltd. 相似文献
8.
Mammalian aging of many tissues is associated with a decline in the replicative and functional capacity of somatic stem cells. Understanding the basis of this decline is a major goal of aging research. Human bone marrow-derived multipotent stromal cells (MSCs) have been applied in the treatment of fracture nonunion. Clinical application of MSCs requires abundant cells that can be overcome by ex vivo expansion of cells, but often at the expense of stemness and differentiation potentiality. We first demonstrated that late-passage MSCs exhibited decreased proliferation capacity, reduced expression of stemness markers such as Oct-4 and Nanog, and deterioration of osteogenic potential. Further, late-passage MSCs showed increased expression of p21(Cip1/Waf1) (p21), an inhibitor of the cyclin-dependent kinase. Knockdown of p21 by lentivirus-mediated shRNAs against p21 in late-passage MSCs increased the proliferation capacity, the expression of Oct-4 and Nanog, and osteogenic potential compared with cells transduced with control shRNA. More importantly, reduction in p21 expression in MSCs enhanced the bone repair capacity of MSCs in a rodent calvarial defect model. Knockdown of p21 in MSCs also increased the telomerase activity and telomere length, and did not show chromosomal abnormalities or acquire transformation ability. Therefore, these data successfully demonstrate the involvement of senescence gene in the expression of stemness markers and osteogenic potential of MSCs. 相似文献
9.
Necrotic and apoptotic cells serve as nuclei for calcification on osteoblastic differentiation of human mesenchymal stem cells in vitro 下载免费PDF全文
Hirofumi Fujita Masanao Yamamoto Tetsuya Ogino Hirotsugu Kobuchi Naoko Ohmoto Eriko Aoyama Takashi Oka Tohru Nakanishi Keiji Inoue Junzo Sasaki 《Cell biochemistry and function》2014,32(1):77-86
A close relationship between cell death and pathological calcification has recently been reported, such as vascular calcification in atherosclerosis. However, the roles of cell death in calcification by osteoblast lineage have not been elucidated in detail. In this study, we investigated whether cell death is involved in the calcification on osteoblastic differentiation of human bone marrow mesenchymal stem cells (hMSC) under osteogenic culture in vitro. Apoptosis and necrosis occurred in an osteogenic culture of hMSC, and cell death preceded calcification. The generation of intracellular reactive oxygen species, chromatin condensation and fragmentation, and caspase‐3 activation increased in this culture. A pan‐caspase inhibitor (Z‐VAD‐FMK) and anti‐oxidants (Tiron and n ‐acetylcysteine) inhibited osteogenic culture‐induced cell death and calcification. Furthermore, calcification was significantly promoted by the addition of necrotic dead cells or its membrane fraction. Spontaneously dead cells by osteogenic culture and exogenously added necrotic cells were surrounded by calcium deposits. Induction of localized cell death by photodynamic treatment in the osteogenic culture resulted in co‐localized calcification. These findings show that necrotic and apoptotic cell deaths were induced in an osteogenic culture of hMSC and indicated that both necrotic and apoptotic cells of osteoblast lineage served as nuclei for calcification on osteoblastic differentiation of hMSC in vitro. Copyright © 2013 John Wiley & Sons, Ltd. 相似文献
10.
Hua Wei Zongwei Li Shengshou Hu Xi Chen Xiangfeng Cong 《Journal of cellular biochemistry》2010,111(4):967-978
Poor survival of mesenchymal stem cells (MSCs) compromised the efficacy of stem cell therapy for myocardial infarction. The increase of exogenous reactive oxygen species (ROS) in infracted heart is one of the important factors that challenged the survival of donor MSCs. In the study we aimed to evaluate the effect of oxidative stress on the cell death of MSCs and investigate its mechanisms in order to help with the identification of new biological compounds to reduce donor cells damage. Apoptosis of MSCs were evaluated with Hoechst 33342 staining and flow cytometry analysis. The mitochondrial membrane potential of MSCs was analyzed with JC‐1 staining. Signaling pathways involved in H2O2 induced apoptosis were analyzed with Western blot. H2O2 induced apoptosis of MSCs in a dose‐ and time‐dependent manner. H2O2 induced apoptosis of MSCs via both endoplasmic reticulum (ER) and mitochondrial pathways rather than extrinsic apoptosis pathway. H2O2 caused transient rather than sustained activation of p38 and JNK with no effect on ERK1/2 pathway. P38 was involved in the regulation of early apoptosis of MSCs while JNK was involved in the late apoptosis. P38 directed both ER stress and mitochondria death pathway in the early apoptosis. In conclusion, exogenous ROS was a major factor to induce apoptosis of MSCs. Both ER stress and mitochondria death pathway were involved in the apoptosis of MSCs. H2O2 activated p38 that directed the above two pathways in the regulation of early apoptosis of MSCs while JNK was involved in the late apoptosis of MSCs. J. Cell. Biochem. 111: 967–978, 2010. © 2010 Wiley‐Liss, Inc. 相似文献
11.
Jeong-Eun Park Young-Kwon Seo Hee-Hoon Yoon Chan-Wha Kim Jung-Keug Park Songhee Jeon 《Neurochemistry international》2013
Even though the inducing effect of electromagnetic fields (EMF) on the neural differentiation of human bone marrow mesenchymal stem cells (hBM-MSCs) is a distinctive, the underlying mechanism of differentiation remains unclear. To find out the signaling pathways involved in the neural differentiation of BM-MSCs by EMF, we examined the CREB phosphorylation and Akt or ERK activation as an upstream of CREB. In hBM-MSCs treated with ELF-EMF (50 Hz, 1 mT), the expression of neural markers such as NF-L, MAP2, and NeuroD1 increased at 6 days and phosphorylation of Akt and CREB but not ERK increased at 90 min in BM-MSCs. Moreover, EMF increased phosphorylation of epidermal growth factor receptor (EGFR) as an upstream receptor tyrosine kinase of PI3K/Akt at 90 min. It has been well documented that ELF-MF exposure may alter cellular processes by increasing intracellular reactive oxygen species (ROS) concentrations. Thus, we examined EMF-induced ROS production in BM-MSCs. Moreover, pretreatment with a ROS scavenger, N-acetylcystein, and an EGFR inhibitor, AG-1478, prevented the phosphorylation of EGFR and downstream molecules. These results suggest that EMF induce neural differentiation through activation of EGFR signaling and mild generation of ROS. 相似文献
12.
ROS与造血干细胞损伤研究进展 总被引:1,自引:0,他引:1
辐射可以通过引起造血干细胞(hematopoietic stem cell,HSC)内活性氧(reactive oxygen species,ROS)系统水平升高导致HSC损伤。HSC损伤患者出现难治性血液系统疾病,严重影响患者生存质量,甚至威胁患者生命。ROS可以通过多种机制引起组织、器官和细胞损伤。ROS的来源包括:线粒体、NOX(NADPH oxidases)、细胞色素P450酶、黄嘌呤氧化酶、非偶联NO合酶。已证实HSC内ROS来源于NOX。ROS升高后影响HSC在成骨细胞微环境定位,导致HSC与微环境相互作用减弱,从而影响HSC功能。此外,ROS升高后通过激活P38MAPK-P16Ink4途径,损伤HSC自我更新能力,并且使HSC定向分化产生更多的髓系克隆而不是红细胞系克隆;P13K—Akt-mTOR途径可能也是ROS诱导HSC损伤途径。ROS对细胞周期影响为:促使HSC离开G0期进入细胞周期,导致干细胞池的耗尽。基于NOX在氧化还原信号传递过程中的重要作用,证实辐射通过NOX产生的ROS以及鉴定产生ROS的NOX亚型,这一工作会为临床靶向治疗辐射诱发的血液系统疾病提供重要的价值。 相似文献
13.
Mesenchymal stem cells(MSCs)have the potential for use in cell-based regenerative therapies.Currently,hundreds of clinical trials are using MSCs for the treatment of various diseases.However,MSCs are low in number in adult tissues;they show heterogeneity depending upon the cell source and exhibit limited proliferative potential and early senescence in in vitro cultures.These factors negatively impact the regenerative potential of MSCs and therefore restrict their use for clinical applications.As a result,novel methods to generate induced MSCs(iMSCs)from induced pluripotent stem cells have been explored.The development and optimization of protocols for generation of iMSCs from induced pluripotent stem cells is necessary to evaluate their regenerative potential in vivo and in vitro.In addition,it is important to compare iMSCs with primary MSCs(isolated from adult tissues)in terms of their safety and efficacy.Careful investigation of the properties of iMSCs in vitro and their long term behavior in animals is important for their translation from bench to bedside. 相似文献
14.
Human mesenchymal stem or stromal cell (hMSC) therapies have promise across a wide range of diseases. However, inefficient cell delivery and low cell survival at injury sites reduce efficacy and are the major barriers in hMSC‐based therapy. Formation of three‐dimensional (3D) hMSC aggregates has been found to activate hMSC functions from enhancing secretion of therapeutic factors for improving cell migration and survival. As the stromal cells in bone marrow, hMSCs are significant sources of extracellular matrix (ECM) proteins and growth factors, which form an interactive microenvironment to influence hMSC fate via paracrine and autocrine actions. To date, however, the impact of the extracellular microenvironment on hMSC properties in the aggregates remains unknown. In the present study, we investigated the role of endogenous ECM matrices on hMSC aggregate formation and survival under ischemic stress. The results demonstrated that the preservation of endogenous ECM in the aggregates formed by thermal lifting (termed TLAs) as opposed to the aggregates formed by enzymatically detached hMSCs (termed EDAs) enhanced cell proliferation, multilineage potential, and survival under ischemic stress. The improved cell proliferation and viability in the TLAs is attributed to the incorporation of endogenous ECM proteins in the TLAs and their promitotic and antioxidant properties. The results demonstrate a novel method for the formation of hMSC aggregates via thermal responsive surface and highlight the significant contribution of the ECM in preserving hMSC properties in the 3D aggregates. © 2013 American Institute of Chemical Engineers Biotechnol. Prog., 29: 441–451, 2013 相似文献
15.
16.
17.
间充质干细胞(mesenchymal stem cells,MSCs)是骨髓中除造血干细胞以外的另一种成体干细胞,广泛分布于动物体内骨髓、肝脏、脂肪等多种组织中。MSCS具有强大的自我更新能力和多向分化潜能,是移植领域应用前景广阔的再生来源细胞;同时,MSCs是一种重要的免疫调节细胞,MSCs在炎症细胞因子刺激后对免疫系统表现出很强的抑制作用,所以MSCs有望应用于减少免疫排斥,延长移植物存活时间,治疗相关免疫失调症,如自身免疫疾病等方面。本文主要对间充质干细胞与免疫系统相互作用的研究做相关介绍。 相似文献
18.
Mesenchymal stem cells (MSCs) are a heterogeneous population of stem/progenitor cells with pluripotent capacity to differentiate into mesodermal and non‐mesodermal cell lineages, including osteocytes, adipocytes, chondrocytes, myocytes, cardiomyocytes, fibroblasts, myofibroblasts, epithelial cells, and neurons. MSCs reside primarily in the bone marrow, but also exist in other sites such as adipose tissue, peripheral blood, cord blood, liver, and fetal tissues. When stimulated by specific signals, these cells can be released from their niche in the bone marrow into circulation and recruited to the target tissues where they undergo in situ differentiation and contribute to tissue regeneration and homeostasis. Several characteristics of MSCs, such as the potential to differentiate into multiple lineages and the ability to be expanded ex vivo while retaining their original lineage differentiation commitment, make these cells very interesting targets for potential therapeutic use in regenerative medicine and tissue engineering. The feasibility for transplantation of primary or engineered MSCs as cell‐based therapy has been demonstrated. In this review, we summarize the current knowledge on the signals that control trafficking and differentiation of MSCs. J. Cell. Biochem. 106: 984–991, 2009. © 2009 Wiley‐Liss, Inc. 相似文献
19.
Xinghui Song Yanwei Li Xiao Chen Guoli Yin Qiong Huang Yingying Chen Guowei Xu Linlin Wang 《Genetics and molecular biology》2014,37(1):127-134
In this work we describe the establishment of mesenchymal stem cells (MSCs) derived from embryonic stem cells (ESCs) and the role of bFGF in adipocyte differentiation. The totipotency of ESCs and MSCs was assessed by immunofluorescence staining and RT-PCR of totipotency factors. MSCs were successfully used to induce osteoblasts, chondrocytes and adipocytes. MSCs that differentiated into adipocytes were stimulated with and without bFGF. The OD/DNA (optical density/content of total DNA) and expression levels of the specific adipocyte genes PPARγ2 (peroxisome proliferator activated receptor γ2) and C/EBPs were higher in bFGF cells. Embryonic bodies had a higher adipocyte level compared with cells cultured in plates. These findings indicate that bFGF promotes adipocyte differentiation. MSCs may be useful cells for seeding in tissue engineering and have enormous therapeutic potential for adipose tissue engineering. 相似文献
20.
Treatment of osteoarthritis with mesenchymal stem cells 总被引:1,自引:0,他引:1
Osteoarthritis(OA)is one of the most prevalent joint diseases with prominent symptoms affecting the daily life of millions of middle aged and elderly people.Despite this,there are no successful medical interventions that can prevent the progressive destruction of OA joints.The onset of pathological changes in OA is associated with deviant activity of mesenchymal stem cells(MSCs),the multipotent precursors of connective tissue cells that reside in joints.Current therapies for OA have resulted in poor clinical outcomes without repairing the damaged cartilage.Intra-articular delivery of culture-expanded MSCs has opened new avenues of OA treatment.Pre-clinical and clinical trials demonstrated the feasibility,safety,and efficacy of MSC therapy.The Wnt/β-catenin,bone morphogenetic protein 2,Indian hedgehog,and Mitogen-activated protein kinase signaling pathways have been demonstrated to be involved in OA and the mechanism of action of MSC therapies. 相似文献