Topic Highlight Open Access
Copyright ©2014 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Stem Cells. Jan 26, 2014; 6(1): 24-32
Published online Jan 26, 2014. doi: 10.4252/wjsc.v6.i1.24
Role of mesenchymal stem cells in cell life and their signaling
Shihori Tanabe, National Institute of Health Sciences, Tokyo 158-8501, Japan
Author contributions: Tanabe S solely contributed to this paper.
Correspondence to: Shihori Tanabe, PhD, National Institute of Health Sciences, 1-18-1, Kami-yoga, Setagaya-ku, Tokyo 158-8501, Japan. stanabe@nihs.go.jp
Telephone: +81-3-37001141 Fax: +81-3-37076950
Received: October 10, 2013
Revised: November 18, 2013
Accepted: December 12, 2013
Published online: January 26, 2014

Abstract

Mesenchymal stem cells (MSCs) have various roles in the body and cellular environment, and the cellular phenotypes of MSCs changes in different conditions. MSCs support the maintenance of other cells, and the capacity of MSCs to differentiate into several cell types makes the cells unique and full of possibilities. The involvement of MSCs in the epithelial-mesenchymal transition is an important property of these cells. In this review, the role of MSCs in cell life, including their application in therapy, is first described, and the signaling mechanism of MSCs is investigated for a further understanding of these cells.

Key Words: Mesenchymal stem cell, Differentiation, Stem cell, Application, Self-renewal

Core tip: Mesenchymal stem cells (MSCs) are important cells that have a differentiation and self-renewal capacity and an immune-modulation function. MSCs differentiate into osteogenic-, adipogenic-, chondrogenic- and other cells. The application of MSCs in many situations, such as disease treatment, is full of possibilities for future development. The gene and protein expression and cellular phenotypes of MSCs are described.



INTRODUCTION

Recent advances in stem cell research have brought about the possibility of stem cell therapies[1], and new approaches using human genetics have been developed to validate therapeutic targets[2]. Furthermore, some gene variations may be useful to detect drug effectiveness on these cells[2]. These novel technologies, when combined, demonstrate their possible application for stem-cell therapeutics. For example, human livers can now be generated from induced pluripotent stem cells (iPSCs) by the transplantation of three-dimensional liver buds that are self-organized in vitro[3]. Furthermore, hematopoietic stem cells can be generated from iPSCs by teratoma formation methods involving co-culturing with OP9 stromal cells, which may be useful for the treatment of hematologic and immunologic diseases[4]. Even diseased cells, such as sarcoma cells, are now being reprogrammed into stem cells with defined factors, such as Oct4, Sox2, c-Myc and Klf4, to change their cellular phenotype to lack tumorigenicity. Furthermore, sarcoma cells can dedifferentiate into mesenchymal stem cells (MSCs) and hematopoietic stem-like cells, and they can differentiate into connective tissue and erythroid cells[5,6].

The regulation of differentiation in stem cells involves the expression of several genes; for example, myogenic differentiation 1 induction in immature human iPSCs leads to the differentiation of these cells into mature myocytes[7]. In hepatic-lineage cell dedifferentiation, cell type-specific transcriptional profiles may correlate to the cell-type transition[8].

Recently, controlling stem-cell fate using chemical approaches has been implicated and could lead to new therapies for disease through the understanding of stem cells and regenerative biology[9]. Chemical therapeutics may induce the self-renewal of cells and target an in vitro niche that allows the cells to progress towards cellular differentiation, proliferation, reprogramming and homing[9]. However, optimization of the chemical structures of these therapeutics is an important factor for developing appropriate cell therapies. Several compounds that regulate cell fate have been selected to bind to nuclear receptors or regulate cellular signaling[9]. The chemical approach to regulating cells and their niches may open a new door for therapeutic strategies in regenerative medicine, such as targeting self-renewal of stem and progenitor cells as well as differentiation and reprogramming[9].

Reprogramming using small molecule compounds such as Forskolin, VPA, CHIR99021, 616452, Tranylcypromine and 3-deazaneplanocin A instead of gene transduction has been successful[10]. In Oct4 promoter-driven green fluorescent protein (GFP)-expressing mouse embryonic fibroblasts, these compounds induce GFP-positive clusters expressing Cdh1 (cadherin 1) (E-cadherin)[10], which is a marker for the mesenchymal-to-epithelial transition, which has recently been revealed to be an important mechanism for the nuclear reprogramming of mouse fibroblasts[11]. Chemically iPSCs have gene expression profiles that are similar to those of embryonic stem cells (ESCs)[10]; therefore, monitoring the gene expression profiles of a variety of iPSCs may be useful for checking the quality of the cells in clinical applications. Additionally, some chemicals important for the self-renewal of ESCs have also been found through screening and may provide insights into the mechanism of stem cell maintenance[12]. For example, a newly identified small molecule maintains the self-renewal ability of mouse ESCs and functions as a dual inhibitor of a protein kinase, ERK1, and a small GTPase-activating protein, Ras GAP[12]. Surprisingly, human iPSCs have been developed into a three-dimensional miniature brain, referred to as an in vitro cerebral organoid, and this method forecasts the future of organ regeneration[13,14]. Of the variety of stem cells, MSCs have the potential to differentiate into multi-lineage cells and have other properties such as immunoregulatory functions, which will be discussed in the following sections[15-19].

ORIGIN OF MSCS

MSCs, which are a type of stem cell, have the ability to differentiate into a variety of cell lineages, such as bone, cartilage, tendon, ligament and adipose tissue[15,16]. MSCs were originally found as nonhematopoietic cells in bone marrow by the observation of Cohnheim nearly 150 years ago, even before being recognized as MSCs[20,21]. Osteogenesis, one of the main characteristics of MSCs, was also observed in bone marrow transplantation before the discovery of MSCs[22,23]. MSCs began to be experimentally recognized and were studied using culture dishes in the 1970’s[24], and during that time, fibroblast colonies were found in bone marrow cultures[24]. The efficacy of MSCs in vivo has been investigated, and MSCs were found to suppress graft vs host disease[19]. Self-renewing MSCs in bone marrow are rare and are candidates for tissue engineering because of their multilineage-differentiation capacity into various cells[25]. The phenotype of MSCs is usually described as positive for many molecules, such as CD105 and CD73, and negative for other molecules, such as CD34, CD45 and CD14; however, the MSC phenotype is altered during cultivation[25-27]. MSCs are promising cells for tissue repair and immunomodulation because they have site-directed and systemic delivery functions[25]. Furthermore, MSCs have now been considered for the potential use in diabetes mellitus treatment, making the clinical application of MSCs more diverse[28]. The roles of MSCs are not restricted to disease treatment and include wound repair, as observed in their original discovery. The sources of MSCs now include bone marrow, amniotic fluid, placenta, umbilical cord blood, periosteum and adipose tissue[29-35].

The criteria for defining MSCs by the International Society for Cellular Therapy position statement are: (1) plastic adherence under standard culture conditions; (2) expression of CD105, CD73 and CD90 and no expression of CD45, CD34, CD14 or CD11b, CD79A or CD19 and HLA-DR; and (3) capacity to differentiate into osteoblasts, chondroblasts and adipocytes in vitro, which is termed the trilineage differentiation potential[36,37]. MSCs have been reported to be transformed into sarcomas, especially in mice, or to promote tumor growth; however, human MSCs are considered relatively safe for clinical applications[36,38]. Recently, MSCs have been used for various purposes that utilize their multi-potential abilities to treat disease, and a new cellular model using disease-derived MSCs has been developed[39]. For the application of MSCs in clinical use, the monitoring of MSC features is essential[40].

MSC APPLICATION

MSCs have been applied for various purposes, such as the clinical replacement of tissues, and as sources of cells in immune-regulation[41,42]. Recently, MSCs have been applied as vaccines[43]. To apply MSCs as a novel vaccine platform, MSCs are expanded ex vivo and genetically modified[43]. However, the quality control of MSCs ex vivo is important for the safe application of these cells; therefore, a bank of stored MSCs has been established[44]. Human iPSCs can be used as sources of MSCs[45]. The induction of human iPSCs using a small-molecule inhibitor of transforming growth factor (TGF)-β into MSCs has been successful[45]. In this section, applications of MSCs and differentiated-MSCs in therapeutics are described.

Osteogenic differentiation of MSCs

MSCs are recognized as sources of bone-related regenerative medicine because they can undergo osteogenesis. One of the mechanisms of osteoporosis has been suggested to be an inability of MSCs to differentiate into osteoblasts[46]. Therefore, a precise investigation for revealing the gene expression profile and molecular signaling of osteogenic differentiation is needed.

Osteogenic differentiation is often induced with dexamethasone, sodium L-ascorbate, and β-glycerol phosphate[47], but the roles of microRNAs (miRNAs) in the osteogenesis of MSCs have also been investigated[48]. Gene expression studies of MSCs using genome-wide association analyses revealed that the EphrinA-EphR pathway for femoral neck bone geometry is coordinated with osteogenesis[49]. Investigation of bovine MSCs has revealed that the osteogenic differentiation of MSCs was highly induced by ascorbic acid and fetal bovine serum[50], and upon the osteogenic differentiation of MSCs, mechanical stress could induce the capacity of MSCs[51]. Hypoxia-mediated signaling in osteogenic differentiation has shown to be mediated via regulation of RUNX2 by TWIST[52]. Moreover, epigenetic regulation is involved in MSC differentiation, and transcription regulation by RUNX2 is important for the osteogenic differentiation capacity of MSCs[47].

Chondrogenic differentiation of MSCs

One application of MSCs is their use in cartilage repair[53]. MSCs from adipose tissue have been chondrogenic-differentiated in 3D culture with hydrogel[53]. Chondrogenic differentiation can be induced by insulin, transferrin, sodium selenite, sodium L-ascorbate, dexamethasone and TGF-β1[54]. In some cases, the modulation of MSCs with chemically oversulfated polysaccharide of marine origin up-regulates the TGF-β-dependent chondrogenesis of MSCs[54]. During chondrogenic differentiation, epigenetic changes have been observed using genome-wide analysis[55], and the expression of several chondrogenic signature genes were found to be up-regulated. For example, it is known that the trimethylation of lysine 4 of histone 3 (H3K4me3) is up-regulated during chondrogenesis[55].

Adipogenic differentiation of MSCs

The adipogenesis of MSCs is usually induced by 3-isobutyl-1-methylxanthine, dexamethasone, indomethacin and insulin, but a mechanically induced signal transduction using the depolymerizing drug cytochalasin D has been shown in adipogenesis[56]. Cytoskeletal mechanisms and signaling molecules such as ERK and AKT are involved in this process[56]. MSCs have been clinically applied in several situations[41]. Adipogenesis-related factors may be used for the treatment of obesity and other related disorders[41]. Importantly, upon adipogenesis of MSCs, the adipogenic differentiation capacity was decreased during in vitro, long-term culture[57]. It is also known that miRNA expression changes upon in vitro senescence of MSCs, which suggests that differential miRNA expression might be useful for distinguishing between MSC phenotypes[57].

Trans-differentiation of MSCs

MSCs also trans-differentiate into non-mesoderm lineages, such as Schwann-like cells which play roles for development, myelination and regeneration in the peripheral nervous system[58-63]. Adipose-derived stem cells can be differentiated into cells with glial phenotype expressing GFAP[58]. Another study has also reported that adipose-derived stem cells can be induced into cells showing neural and glial cell phenotype[59]. MSCs derived from bone marrow and adipose are shown to express mRNAs and proteins of myelin that is formed by Schwann cells[64]. MSCs are also suggested to differentiate into myoblast[65]. On the other hand, MSCs are also used for various diseases including kidney injury, diabetes and brain tumors, although it needs to be elucidated whether the anti-disease effect of MSCs arise from trans-differentiation or other paracrine effects of MSCs[66-70]. The effect of MSCs for treatment of brain tumors is suggested to be caused by paracrine effect of MSCs towards cancer cells[70].

MSC effects in the immune response

MSCs are known to be involved in the immune response during circumstances such as the allogenic transplantation of bone marrow, which mainly causes an immunosuppressive effect. A MSC-induced immunosuppressive effect might be caused by the down-regulation of T-cell differentiation into T helper 17 (Th17) cells and of the function of mature Th17 cells[71]. This inhibition of mature Th17 cells could occur via the cell-to-cell contact mechanism of MSCs and may be mediated by the programmed death-1 pathway[71]. Allogenic MSCs have also been demonstrated to regulate the function of Th17 cells derived from rheumatoid arthritis patients[72]. Co-cultures of MSCs with peripheral blood mononuclear cells (PBMCs) cause a decrease in orphan nuclear receptor gamma (ROR-γ), which is involved in Th17 differentiation and is expressed in PBMCs[72]. MSCs produce TGF-β and interleukin-6 (IL-6) and regulate the differentiation of T cells into regulatory T cells or Th17 cells[73]. Therefore, the immune response may be regulated by MSCs via Th17 signaling.

MSCs have also been applied as novel vaccine platforms[43]. MSC vaccination strategies include the modified application of MSCs in anti-microbial or cancer immunization[43]. Genetically modified MSCs may act as antigen presenters or mediators as well as suppliers of immune-related cytokines[43]. MSCs from the placenta are known to suppress allogeneic umbilical cord blood lymphocyte proliferation[74], and it has been suggested that placenta-derived MSCs may be applied in allograft transplantations[74]. The immunomodulatory properties of equine adult-derived MSCs derived from bone marrow, adipose tissue, umbilical cord blood and umbilical cord tissue have been compared, and it was revealed that lymphocyte proliferation is suppressed by MSCs and secretion of prostaglandin E2 and IL-6 is increased upon allogenic PBMC or phytohemagglutinin stimulation[75]. MSCs also decrease the production of tumor necrosis factor-α and interferon-γ[75]. The immunomodulatory effect of MSCs on B and T cells have also been studied[76]. One of the main roles of MSCs on B cells is the inhibition of B cell proliferation, but their effects on B cells are still controversial[76]. The MSC-induced regulation of the proliferative response of lymphocytes has been reported to be independent of the major histocompatibility complex, although MSCs mainly induce inhibition and sometimes cause enhancement of the mixed-lymphocyte reaction[77].

MSC GENE EXPRESSION

MSCs have a variety of gene expression profiles during their developmental stages. Gene expression of MSCs differentiating into adipocytes has been investigated and adenomatosis polyposis coli down-regulated-1 (APCDD1), chitinase 3-like 1 (cartilage glycoprotein-39) (CHI3L1), retinoic acid receptor responder (tazarotene induced 1) (RARRES1) and sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3G (SEMA3G) have been identified as potential adipogenic-specific genes[78]. The expression of these genes is regulated in a time-dependent manner during adipogenesis[78]. During in vitro adipogenic and osteogenic differentiation of MSCs, various genes alter their mRNA expression[79]. Genes related to cell proliferation and cytoskeleton organization are activated during the osteogenic differentiation of MSCs, whereas genes in PPAR signaling are regulated during the adipogenic differentiation of MSCs[79].

The gene expression signature is used for the prediction of disease progression or cancer phenotype. The gene expression of patients with breast cancer has been analyzed, and the disease outcomes of young patients have been profiled using predictors[80]. In anticancer drug discovery, gene expression profiling has been performed on colon cancer cell lines[81]. MSCs with migration capacity have been used in anti-tumor therapy and must be examined carefully for safety and efficacy[82]. The gene expression of pluripotency-related genes have been examined in MSCs derived from bone marrow, adipocytes, amniotic membrane and epithelial endometrium-derived stem cells and stroma endometrium-derived stem cells, and these studies suggest that pluripotency-related gene expression varies in different tissues[82]. Xenograft imaging of mice differentiates between the gene expression patterns of human MSCs and human iPSCs, and the tumor sizes of tumor xenografts of iPSCs are larger than those of MSCs, indicating differences in the migration capacity of MSCs and iPSCs[82]. Comparison of phenotypic markers and the neural differentiation capacity of MSCs and multipotent adult progenitor cells has been analyzed, and MSCs expressing CD44, CD73 and CD105 have a higher differentiation capacity into neuro-ectodermal lineages than multipotent adult progenitor cells[83].

The miRNA expression in adipose-derived MSCs has been analyzed, and miR-27b has been identified to be involved in the tolerogenic response[84]; moreover, it was stated that miR-27b is associated with cell differentiation function[84]. Another study has revealed that miR-574-3p, which is regulated with Sox9, inhibits the chondrogenic differentiation of MSCs[85]. The expression of miRNAs, such as the miR-30 family, let-7 family, miR-21, miR-16, miR-155, miR-322 and Snord85, is regulated during the osteoblastic and osteocytic differentiation of MSCs[86]. These miRNAs are thought to target osteogenic differentiation-, stemness-, epigenetics- and cell cycle-related mRNAs[86]. The effects of the mechanical stimulation of MSCs that are seeded on calcium phosphate cement have been analyzed, and it was found that a small number of immediate-early response genes that were associated with transcription were activated[87].

MSC PROTEIN EXPRESSION

Protein expression in MSCs has been investigated in various experimental systems. Rat oligodendroglial cell maturation is promoted by MSC-derived soluble factors and induces an increase in myelin expression and a decrease in glial fibrillary acidic protein expression[88]. In thyroid hormone-induced hypertrophy in MSC chondrogenesis, bone morphogenetic protein-4 (BMP4) is up-regulated; therefore, BMP4 signaling is suggested to be involved[89]. These investigations may provide useful insights into the application of chondrogenic-differentiated MSCs[90]. MSCs exhibit biocompatibility and favorable responses towards the fibronectin-gold nanocomposite film coating that is used in cardiovascular devices[90]. MSCs on fibronectin-gold nanocomposites increase the protein expression levels of matrix metalloproteinase-9 and endothelial nitric oxide synthase[90]. Fibronectin expression has also been linked to MSC lung adherence[91]. In breast and prostate tumors, MSCs promote the growth and angiogenesis of tumors via the expression of pro-angiogenic factors associated with neovascularization, such as macrophage inflammatory protein-2, vascular endothelial growth factor, TGF-β and IL-6[92]. In the tissue engineering of articular cartilage, chondrocytes from healthy-donor-derived MSCs exhibit similar properties to those of osteoarthritis joints[93]. Specifically, chondrocytes from MSCs and osteoarthritis joints contain hyaline cartilage-specific type II and fibrocartilage-specific type I collagen[93]. Differentiated MSCs have increased chitinase family glycoprotein YKL-40 protein levels, and considering that the mRNA of YKL-40 is expressed in undifferentiated MSCs, the regulation between the mRNA and protein levels would be interesting to investigate[94]. The Fas ligand (FasL) plays an important role in regulating the determination of MSC fates into proliferation or adipogenic differentiation[95]. Low levels of FasL induce proliferation, whereas high levels inhibit adipogenic differentiation[95]. Adhesion and osteogenic differentiation of animal, serum-free, expanded MSCs are promoted by laminin-5 and type I collagen[96]; therefore, these proteins may be considered for the application of the in vitro proliferation of MSCs in animal serum-free conditions[96]. MSCs are committed to adipogenic differentiation under protein malnutrition conditions when PPAR-γ protein and mRNA levels increase[97]. Furthermore, MSCs decrease the levels of TGF-β1 in microglia/macrophages after stroke, and this is followed by a decrease in the levels of plasminogen activator inhibitor 1 in astrocytes[98].

Upon the isolation of MSCs from bone marrow, surface antigens, such as CD10, CD73, CD140b, CD146, GD2 and CD271, can be used as MSC markers[99]. In addition to those antigens, pluripotency-related proteins, such as Oct4, Nanog and SSEA-4, have been identified to distinguish cellular populations in the human trabecular bone and bone marrow[99]. Another report has shown that the transcription factor Ebf2 in adult bone marrow is useful for distinguishing between MSC or mesenchymal progenitor-like cell phenotypes[100].

CANCER SIGNALING IN MSCS

Cancer is maintained by cancer stem cells (CSCs), which emphasizes the importance of the identification, targeting and elimination of these types of cells[101]. The initiation of cancer is thought to occur by the activation of self-renewal mechanisms that are usually restricted to stem cells[101]. Cancer cells showing CSC-like phenotypes may remain in the stem-cell state and tend to avoid cancer differentiation. Cancers exhibiting epithelial CSC-like phenotypes have an increased probability of migration and death, which indicates the possibility of epithelial CSC-like phenotypes as diagnosis predictors for cancer. This phenotype factor is shown to be independent from usual cancer diagnosis factors, such as patient age, cancer diameter, cancer progression, estrogen receptor status, lymphoid node status and blood vessel infiltration. A portion of these cells in cancer may be involved in initiation and infinite cancer proliferation[101].

CD44, which is proposed to be a tumor-initiating marker for glioma sphere cultures, has been shown to be a mesenchymal signature because mesenchymal-differentiated glioma sphere cultures have radiation resistance and include a CD44 subpopulation[102]. Recent studies demonstrate that MSCs play an important role in the formation of CSCs[103].

It is known that subunits of the polycomb repressor complex (PRC), such as BMI1 polycomb ring finger oncogene, are associated with the epithelial-mesenchymal transition, cancer progression and stem-like expression profile[104-106]. PRC2 is involved in the regulation of gene repression through chromatin modifications and is thought to be important in stem cells[107-109]. A recent study in Drosophila has shown that a mutation in lysine 27 of histone H3 causes the same phenotype as flies, with a loss of PRC2, which indicates that the methylation of lysine 27 of histone H3 is important for PRC function[110]. The role of chromatin regulation in CSCs will be investigated in the future.

Great efforts to understand CSCs have revealed the feature of CSCs as specific cell populations in brain, skin and intestinal tumors and the possibility of effective treatments by targeting these cells[111-114]. The markers for CSCs have been identified and include aldehyde dehydrogenase 1 (ALDH1), CD24, CD44, CD90, CD133, Hedgehog-Gli activity and α6-integrin in breast cancer as well as ABCB5, ALDH1, β-catenin activity, CD24, CD26, CD29, CD44, CD133, CD166 and leucine-rich repeat containing G protein-coupled receptor 5 in colon cancer[115].

CONCLUSION

In summary, MSCs play important roles in cell life. MSCs differentiate into various cell types, and their many applications, such as for disease treatment, are being studied. Further investigation of the MSC phenotypes is needed for the development of the safe and effective application of MSCs.

Footnotes

P- Reviewers: Bonetti B, Shawcross SG, Zaminy A S- Editor: Cui XM L- Editor: A E- Editor: Liu SQ

References
1.  Sandoe J, Eggan K. Opportunities and challenges of pluripotent stem cell neurodegenerative disease models. Nat Neurosci. 2013;16:780-789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 159]  [Article Influence: 14.5]  [Reference Citation Analysis (0)]
2.  Plenge RM, Scolnick EM, Altshuler D. Validating therapeutic targets through human genetics. Nat Rev Drug Discov. 2013;12:581-594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 420]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
3.  Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T, Zhang RR, Ueno Y, Zheng YW, Koike N. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature. 2013;499:481-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1350]  [Cited by in F6Publishing: 1352]  [Article Influence: 122.9]  [Reference Citation Analysis (0)]
4.  Suzuki N, Yamazaki S, Yamaguchi T, Okabe M, Masaki H, Takaki S, Otsu M, Nakauchi H. Generation of engraftable hematopoietic stem cells from induced pluripotent stem cells by way of teratoma formation. Mol Ther. 2013;21:1424-1431.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 162]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
5.  Lang JY, Shi Y, Chin YE. Reprogramming cancer cells: back to the future. Oncogene. 2013;32:2247-2248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
6.  Zhang X, Cruz FD, Terry M, Remotti F, Matushansky I. Terminal differentiation and loss of tumorigenicity of human cancers via pluripotency-based reprogramming. Oncogene. 2013;32:2249-2260, 2260.e1-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 97]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
7.  Tanaka A, Woltjen K, Miyake K, Hotta A, Ikeya M, Yamamoto T, Nishino T, Shoji E, Sehara-Fujisawa A, Manabe Y. Efficient and reproducible myogenic differentiation from human iPS cells: prospects for modeling Miyoshi Myopathy in vitro. PLoS One. 2013;8:e61540.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 153]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
8.  Hikichi T, Matoba R, Ikeda T, Watanabe A, Yamamoto T, Yoshitake S, Tamura-Nakano M, Kimura T, Kamon M, Shimura M. Transcription factors interfering with dedifferentiation induce cell type-specific transcriptional profiles. Proc Natl Acad Sci USA. 2013;110:6412-6417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 37]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
9.  Xu Y, Shi Y, Ding S. A chemical approach to stem-cell biology and regenerative medicine. Nature. 2008;453:338-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 267]  [Cited by in F6Publishing: 248]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
10.  Hou P, Li Y, Zhang X, Liu C, Guan J, Li H, Zhao T, Ye J, Yang W, Liu K. Pluripotent stem cells induced from mouse somatic cells by small-molecule compounds. Science. 2013;341:651-654.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 938]  [Cited by in F6Publishing: 938]  [Article Influence: 85.3]  [Reference Citation Analysis (0)]
11.  Li R, Liang J, Ni S, Zhou T, Qing X, Li H, He W, Chen J, Li F, Zhuang Q. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell Stem Cell. 2010;7:51-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 895]  [Cited by in F6Publishing: 890]  [Article Influence: 63.6]  [Reference Citation Analysis (0)]
12.  Chen S, Do JT, Zhang Q, Yao S, Yan F, Peters EC, Schöler HR, Schultz PG, Ding S. Self-renewal of embryonic stem cells by a small molecule. Proc Natl Acad Sci USA. 2006;103:17266-17271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 250]  [Article Influence: 13.9]  [Reference Citation Analysis (0)]
13.  Brüstle O. Developmental neuroscience: Miniature human brains. Nature. 2013;501:319-320.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
14.  Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM, Jackson AP, Knoblich JA. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501:373-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3083]  [Cited by in F6Publishing: 3038]  [Article Influence: 276.2]  [Reference Citation Analysis (0)]
15.  Caplan AI. Mesenchymal stem cells. J Orthop Res. 1991;9:641-650.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3539]  [Cited by in F6Publishing: 3167]  [Article Influence: 96.0]  [Reference Citation Analysis (0)]
16.  Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR. Multilineage potential of adult human mesenchymal stem cells. Science. 1999;284:143-147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15372]  [Cited by in F6Publishing: 14786]  [Article Influence: 591.4]  [Reference Citation Analysis (0)]
17.  Siegel G, Kluba T, Hermanutz-Klein U, Bieback K, Northoff H, Schäfer R. Phenotype, donor age and gender affect function of human bone marrow-derived mesenchymal stromal cells. BMC Med. 2013;11:146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 308]  [Cited by in F6Publishing: 318]  [Article Influence: 28.9]  [Reference Citation Analysis (0)]
18.  Wang Z, Tang X, Xu W, Cao Z, Sun L, Li W, Li Q, Zou P, Zhao Z. The different immunoregulatory functions on dendritic cells between mesenchymal stem cells derived from bone marrow of patients with low-risk or high-risk myelodysplastic syndromes. PLoS One. 2013;8:e57470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
19.  Bonfield TL, Nolan Koloze MT, Lennon DP, Caplan AI. Defining human mesenchymal stem cell efficacy in vivo. J Inflamm (Lond). 2010;7:51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 61]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
20.  Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science. 1997;276:71-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3486]  [Cited by in F6Publishing: 3310]  [Article Influence: 122.6]  [Reference Citation Analysis (0)]
21.  Cohnheim J. Ueber Entzündung und Eiterung. Arch Path Anat Physiol Klin Med. 1867;40:1-79.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Friedenstein AJ, Piatetzky-Shapiro II, Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol. 1966;16:381-390.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation. 1968;6:230-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1396]  [Cited by in F6Publishing: 1208]  [Article Influence: 21.6]  [Reference Citation Analysis (0)]
24.  Friedenstein AJ, Gorskaja JF, Kulagina NN. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol. 1976;4:267-274.  [PubMed]  [DOI]  [Cited in This Article: ]
25.  Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25:2739-2749.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1697]  [Cited by in F6Publishing: 1616]  [Article Influence: 95.1]  [Reference Citation Analysis (0)]
26.  Qian H, Le Blanc K, Sigvardsson M. Primary mesenchymal stem and progenitor cells from bone marrow lack expression of CD44 protein. J Biol Chem. 2012;287:25795-25807.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 109]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
27.  Tanabe S, Sato Y, Suzuki T, Suzuki K, Nagao T, Yamaguchi T. Gene expression profiling of human mesenchymal stem cells for identification of novel markers in early- and late-stage cell culture. J Biochem. 2008;144:399-408.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
28.  Domínguez-Bendala J, Lanzoni G, Inverardi L, Ricordi C. Concise review: mesenchymal stem cells for diabetes. Stem Cells Transl Med. 2012;1:59-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 59]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
29.  Maxson S, Lopez EA, Yoo D, Danilkovitch-Miagkova A, Leroux MA. Concise review: role of mesenchymal stem cells in wound repair. Stem Cells Transl Med. 2012;1:142-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 572]  [Cited by in F6Publishing: 509]  [Article Influence: 42.4]  [Reference Citation Analysis (0)]
30.  O’Donoghue K, Choolani M, Chan J, de la Fuente J, Kumar S, Campagnoli C, Bennett PR, Roberts IA, Fisk NM. Identification of fetal mesenchymal stem cells in maternal blood: implications for non-invasive prenatal diagnosis. Mol Hum Reprod. 2003;9:497-502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 108]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
31.  Int Anker PS, Scherjon SA, Kleijburg-van der Keur C, Noort WA, Claas FH, Willemze R, Fibbe WE, Kanhai HH. Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood. 2003;102:1548-1549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 580]  [Cited by in F6Publishing: 507]  [Article Influence: 24.1]  [Reference Citation Analysis (0)]
32.  Int Anker PS, Scherjon SA, Kleijburg-van der Keur C, de Groot-Swings GM, Claas FH, Fibbe WE, Kanhai HH. Isolation of mesenchymal stem cells of fetal or maternal origin from human placenta. Stem Cells. 2004;22:1338-1345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 824]  [Cited by in F6Publishing: 790]  [Article Influence: 41.6]  [Reference Citation Analysis (0)]
33.  Jin HJ, Bae YK, Kim M, Kwon SJ, Jeon HB, Choi SJ, Kim SW, Yang YS, Oh W, Chang JW. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci. 2013;14:17986-18001.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in F6Publishing: 420]  [Article Influence: 38.2]  [Reference Citation Analysis (0)]
34.  Nakahara H, Dennis JE, Bruder SP, Haynesworth SE, Lennon DP, Caplan AI. In vitro differentiation of bone and hypertrophic cartilage from periosteal-derived cells. Exp Cell Res. 1991;195:492-503.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 166]  [Cited by in F6Publishing: 173]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
35.  Zuk PA, Zhu M, Ashjian P, De Ugarte DA, Huang JI, Mizuno H, Alfonso ZC, Fraser JK, Benhaim P, Hedrick MH. Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 2002;13:4279-4295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4817]  [Cited by in F6Publishing: 4876]  [Article Influence: 221.6]  [Reference Citation Analysis (0)]
36.  Xiao W, Mohseny AB, Hogendoorn PC, Cleton-Jansen AM. Mesenchymal stem cell transformation and sarcoma genesis. Clin Sarcoma Res. 2013;3:10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 71]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
37.  Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, Deans R, Keating A, Prockop Dj, Horwitz E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315-317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11055]  [Cited by in F6Publishing: 11806]  [Article Influence: 694.5]  [Reference Citation Analysis (1)]
38.  Huang WH, Chang MC, Tsai KS, Hung MC, Chen HL, Hung SC. Mesenchymal stem cells promote growth and angiogenesis of tumors in mice. Oncogene. 2013;32:4343-4354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 160]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
39.  Boone N, Loriod B, Bergon A, Sbai O, Formisano-Tréziny C, Gabert J, Khrestchatisky M, Nguyen C, Féron F, Axelrod FB. Olfactory stem cells, a new cellular model for studying molecular mechanisms underlying familial dysautonomia. PLoS One. 2010;5:e15590.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
40.  Wagner W, Bork S, Lepperdinger G, Joussen S, Ma N, Strunk D, Koch C. How to track cellular aging of mesenchymal stromal cells. Aging (Albany NY). 2010;2:224-230.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Rastegar F, Shenaq D, Huang J, Zhang W, Zhang BQ, He BC, Chen L, Zuo GW, Luo Q, Shi Q. Mesenchymal stem cells: Molecular characteristics and clinical applications. World J Stem Cells. 2010;2:67-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 148]  [Article Influence: 10.6]  [Reference Citation Analysis (2)]
42.  Panetta NJ, Gupta DM, Quarto N, Longaker MT. Mesenchymal cells for skeletal tissue engineering. Panminerva Med. 2009;51:25-41.  [PubMed]  [DOI]  [Cited in This Article: ]
43.  Tomchuck SL, Norton EB, Garry RF, Bunnell BA, Morris CA, Freytag LC, Clements JD. Mesenchymal stem cells as a novel vaccine platform. Front Cell Infect Microbiol. 2012;2:140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
44.  Sabatino M, Ren J, David-Ocampo V, England L, McGann M, Tran M, Kuznetsov SA, Khuu H, Balakumaran A, Klein HG. The establishment of a bank of stored clinical bone marrow stromal cell products. J Transl Med. 2012;10:23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 38]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
45.  Chen YS, Pelekanos RA, Ellis RL, Horne R, Wolvetang EJ, Fisk NM. Small molecule mesengenic induction of human induced pluripotent stem cells to generate mesenchymal stem/stromal cells. Stem Cells Transl Med. 2012;1:83-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 144]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
46.  Xu XH, Dong SS, Guo Y, Yang TL, Lei SF, Papasian CJ, Zhao M, Deng HW. Molecular genetic studies of gene identification for osteoporosis: the 2009 update. Endocr Rev. 2010;31:447-505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
47.  Eslaminejad MB, Fani N, Shahhoseini M. Epigenetic regulation of osteogenic and chondrogenic differentiation of mesenchymal stem cells in culture. Cell J. 2013;15:1-10.  [PubMed]  [DOI]  [Cited in This Article: ]
48.  Schoolmeesters A, Eklund T, Leake D, Vermeulen A, Smith Q, Force Aldred S, Fedorov Y. Functional profiling reveals critical role for miRNA in differentiation of human mesenchymal stem cells. PLoS One. 2009;4:e5605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 145]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
49.  Chen Y, Xiong DH, Guo YF, Pan F, Zhou Q, Zhang F, Deng HW. Pathway-based genome-wide association analysis identified the importance of EphrinA-EphR pathway for femoral neck bone geometry. Bone. 2010;46:129-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
50.  Cortes Y, Ojeda M, Araya D, Dueñas F, Fernández MS, Peralta OA. Isolation and multilineage differentiation of bone marrow mesenchymal stem cells from abattoir-derived bovine fetuses. BMC Vet Res. 2013;9:133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
51.  Delaine-Smith RM, Reilly GC. Mesenchymal stem cell responses to mechanical stimuli. Muscles Ligaments Tendons J. 2012;2:169-180.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Yang DC, Yang MH, Tsai CC, Huang TF, Chen YH, Hung SC. Hypoxia inhibits osteogenesis in human mesenchymal stem cells through direct regulation of RUNX2 by TWIST. PLoS One. 2011;6:e23965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 128]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
53.  Portron S, Merceron C, Gauthier O, Lesoeur J, Sourice S, Masson M, Fellah BH, Geffroy O, Lallemand E, Weiss P. Effects of in vitro low oxygen tension preconditioning of adipose stromal cells on their in vivo chondrogenic potential: application in cartilage tissue repair. PLoS One. 2013;8:e62368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 60]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
54.  Merceron C, Portron S, Vignes-Colombeix C, Rederstorff E, Masson M, Lesoeur J, Sourice S, Sinquin C, Colliec-Jouault S, Weiss P. Pharmacological modulation of human mesenchymal stem cell chondrogenesis by a chemically oversulfated polysaccharide of marine origin: potential application to cartilage regenerative medicine. Stem Cells. 2012;30:471-480.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 54]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
55.  Herlofsen SR, Bryne JC, Høiby T, Wang L, Issner R, Zhang X, Coyne MJ, Boyle P, Gu H, Meza-Zepeda LA. Genome-wide map of quantified epigenetic changes during in vitro chondrogenic differentiation of primary human mesenchymal stem cells. BMC Genomics. 2013;14:105.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 63]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
56.  Müller P, Langenbach A, Kaminski A, Rychly J. Modulating the actin cytoskeleton affects mechanically induced signal transduction and differentiation in mesenchymal stem cells. PLoS One. 2013;8:e71283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 85]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
57.  Wagner W, Horn P, Castoldi M, Diehlmann A, Bork S, Saffrich R, Benes V, Blake J, Pfister S, Eckstein V. Replicative senescence of mesenchymal stem cells: a continuous and organized process. PLoS One. 2008;3:e2213.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 758]  [Cited by in F6Publishing: 808]  [Article Influence: 50.5]  [Reference Citation Analysis (0)]
58.  Faroni A, Rothwell SW, Grolla AA, Terenghi G, Magnaghi V, Verkhratsky A. Differentiation of adipose-derived stem cells into Schwann cell phenotype induces expression of P2X receptors that control cell death. Cell Death Dis. 2013;4:e743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
59.  di Summa PG, Kalbermatten DF, Raffoul W, Terenghi G, Kingham PJ. Extracellular matrix molecules enhance the neurotrophic effect of Schwann cell-like differentiated adipose-derived stem cells and increase cell survival under stress conditions. Tissue Eng Part A. 2013;19:368-379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 60]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
60.  Armstrong SJ, Wiberg M, Terenghi G, Kingham PJ. ECM molecules mediate both Schwann cell proliferation and activation to enhance neurite outgrowth. Tissue Eng. 2007;13:2863-2870.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 116]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
61.  Grigoryan T, Stein S, Qi J, Wende H, Garratt AN, Nave KA, Birchmeier C, Birchmeier W. Wnt/Rspondin/β-catenin signals control axonal sorting and lineage progression in Schwann cell development. Proc Natl Acad Sci USA. 2013;110:18174-18179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
62.  Jonsson S, Wiberg R, McGrath AM, Novikov LN, Wiberg M, Novikova LN, Kingham PJ. Effect of delayed peripheral nerve repair on nerve regeneration, Schwann cell function and target muscle recovery. PLoS One. 2013;8:e56484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 130]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
63.  Keilhoff G, Goihl A, Stang F, Wolf G, Fansa H. Peripheral nerve tissue engineering: autologous Schwann cells vs. transdifferentiated mesenchymal stem cells. Tissue Eng. 2006;12:1451-1465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 109]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
64.  Mantovani C, Mahay D, Kingham M, Terenghi G, Shawcross SG, Wiberg M. Bone marrow- and adipose-derived stem cells show expression of myelin mRNAs and proteins. Regen Med. 2010;5:403-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 53]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
65.  Caplan AI, Correa D. The MSC: an injury drugstore. Cell Stem Cell. 2011;9:11-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1127]  [Cited by in F6Publishing: 1144]  [Article Influence: 95.3]  [Reference Citation Analysis (0)]
66.  Yu X, Lu C, Liu H, Rao S, Cai J, Liu S, Kriegel AJ, Greene AS, Liang M, Ding X. Hypoxic preconditioning with cobalt of bone marrow mesenchymal stem cells improves cell migration and enhances therapy for treatment of ischemic acute kidney injury. PLoS One. 2013;8:e62703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
67.  Ezquer F, Ezquer M, Contador D, Ricca M, Simon V, Conget P. The antidiabetic effect of mesenchymal stem cells is unrelated to their transdifferentiation potential but to their capability to restore Th1/Th2 balance and to modify the pancreatic microenvironment. Stem Cells. 2012;30:1664-1674.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 118]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
68.  Hughey CC, Ma L, James FD, Bracy DP, Wang Z, Wasserman DH, Rottman JN, Hittel DS, Shearer J. Mesenchymal stem cell transplantation for the infarcted heart: therapeutic potential for insulin resistance beyond the heart. Cardiovasc Diabetol. 2013;12:128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
69.  Frattini F, Lopes FR, Almeida FM, Rodrigues RF, Boldrini LC, Tomaz MA, Baptista AF, Melo PA, Martinez AM. Mesenchymal stem cells in a polycaprolactone conduit promote sciatic nerve regeneration and sensory neuron survival after nerve injury. Tissue Eng Part A. 2012;18:2030-2039.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 59]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
70.  Chan JK, Lam PY. Human mesenchymal stem cells and their paracrine factors for the treatment of brain tumors. Cancer Gene Ther. 2013;20:539-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
71.  Luz-Crawford P, Noël D, Fernandez X, Khoury M, Figueroa F, Carrión F, Jorgensen C, Djouad F. Mesenchymal stem cells repress Th17 molecular program through the PD-1 pathway. PLoS One. 2012;7:e45272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 146]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
72.  Wang Q, Li X, Luo J, Zhang L, Ma L, Lv Z, Xue L. The allogeneic umbilical cord mesenchymal stem cells regulate the function of T helper 17 cells from patients with rheumatoid arthritis in an in vitro co-culture system. BMC Musculoskelet Disord. 2012;13:249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
73.  Svobodova E, Krulova M, Zajicova A, Pokorna K, Prochazkova J, Trosan P, Holan V. The role of mouse mesenchymal stem cells in differentiation of naive T-cells into anti-inflammatory regulatory T-cell or proinflammatory helper T-cell 17 population. Stem Cells Dev. 2012;21:901-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 90]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
74.  Li CD, Zhang WY, Li HL, Jiang XX, Zhang Y, Tang PH, Mao N. Mesenchymal stem cells derived from human placenta suppress allogeneic umbilical cord blood lymphocyte proliferation. Cell Res. 2005;15:539-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 102]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
75.  Carrade DD, Lame MW, Kent MS, Clark KC, Walker NJ, Borjesson DL. Comparative Analysis of the Immunomodulatory Properties of Equine Adult-Derived Mesenchymal Stem Cells(). Cell Med. 2012;4:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 140]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
76.  Franquesa M, Hoogduijn MJ, Bestard O, Grinyó JM. Immunomodulatory effect of mesenchymal stem cells on B cells. Front Immunol. 2012;3:212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 134]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
77.  Le Blanc K, Tammik L, Sundberg B, Haynesworth SE, Ringdén O. Mesenchymal stem cells inhibit and stimulate mixed lymphocyte cultures and mitogenic responses independently of the major histocompatibility complex. Scand J Immunol. 2003;57:11-20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1048]  [Cited by in F6Publishing: 972]  [Article Influence: 46.3]  [Reference Citation Analysis (0)]
78.  Ullah M, Stich S, Häupl T, Eucker J, Sittinger M, Ringe J. Reverse differentiation as a gene filtering tool in genome expression profiling of adipogenesis for fat marker gene selection and their analysis. PLoS One. 2013;8:e69754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
79.  Monaco E, Bionaz M, Rodriguez-Zas S, Hurley WL, Wheeler MB. Transcriptomics comparison between porcine adipose and bone marrow mesenchymal stem cells during in vitro osteogenic and adipogenic differentiation. PLoS One. 2012;7:e32481.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 57]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
80.  van de Vijver MJ, He YD, van’t Veer LJ, Dai H, Hart AA, Voskuil DW, Schreiber GJ, Peterse JL, Roberts C, Marton MJ. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002;347:1999-2009.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4800]  [Cited by in F6Publishing: 4354]  [Article Influence: 197.9]  [Reference Citation Analysis (0)]
81.  Sampson ER, McMurray HR, Hassane DC, Newman L, Salzman P, Jordan CT, Land H. Gene signature critical to cancer phenotype as a paradigm for anticancer drug discovery. Oncogene. 2013;32:3809-3818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
82.  Belmar-Lopez C, Mendoza G, Oberg D, Burnet J, Simon C, Cervello I, Iglesias M, Ramirez JC, Lopez-Larrubia P, Quintanilla M. Tissue-derived mesenchymal stromal cells used as vehicles for anti-tumor therapy exert different in vivo effects on migration capacity and tumor growth. BMC Med. 2013;11:139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 55]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
83.  Singh SP, Tripathy NK, Nityanand S. Comparison of phenotypic markers and neural differentiation potential of multipotent adult progenitor cells and mesenchymal stem cells. World J Stem Cells. 2013;5:53-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
84.  Chen KD, Goto S, Hsu LW, Lin TY, Nakano T, Lai CY, Chang YC, Weng WT, Kuo YR, Wang CC. Identification of miR-27b as a novel signature from the mRNA profiles of adipose-derived mesenchymal stem cells involved in the tolerogenic response. PLoS One. 2013;8:e60492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
85.  Guérit D, Philipot D, Chuchana P, Toupet K, Brondello JM, Mathieu M, Jorgensen C, Noël D. Sox9-regulated miRNA-574-3p inhibits chondrogenic differentiation of mesenchymal stem cells. PLoS One. 2013;8:e62582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 80]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
86.  Eguchi T, Watanabe K, Hara ES, Ono M, Kuboki T, Calderwood SK. OstemiR: a novel panel of microRNA biomarkers in osteoblastic and osteocytic differentiation from mesencymal stem cells. PLoS One. 2013;8:e58796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 134]  [Article Influence: 12.2]  [Reference Citation Analysis (0)]
87.  Gharibi B, Cama G, Capurro M, Thompson I, Deb S, Di Silvio L, Hughes FJ. Gene expression responses to mechanical stimulation of mesenchymal stem cells seeded on calcium phosphate cement. Tissue Eng Part A. 2013;19:2426-2438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
88.  Jadasz JJ, Kremer D, Göttle P, Tzekova N, Domke J, Rivera FJ, Adjaye J, Hartung HP, Aigner L, Küry P. Mesenchymal stem cell conditioning promotes rat oligodendroglial cell maturation. PLoS One. 2013;8:e71814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
89.  Karl A, Olbrich N, Pfeifer C, Berner A, Zellner J, Kujat R, Angele P, Nerlich M, Mueller MB. Thyroid hormone-induced hypertrophy in mesenchymal stem cell chondrogenesis is mediated by bone morphogenetic protein-4. Tissue Eng Part A. 2014;20:178-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
90.  Hung HS, Tang CM, Lin CH, Lin SZ, Chu MY, Sun WS, Kao WC, Hsien-Hsu H, Huang CY, Hsu SH. Biocompatibility and favorable response of mesenchymal stem cells on fibronectin-gold nanocomposites. PLoS One. 2013;8:e65738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
91.  Nystedt J, Anderson H, Tikkanen J, Pietilä M, Hirvonen T, Takalo R, Heiskanen A, Satomaa T, Natunen S, Lehtonen S. Cell surface structures influence lung clearance rate of systemically infused mesenchymal stromal cells. Stem Cells. 2013;31:317-326.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 91]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
92.  Zhang T, Lee YW, Rui YF, Cheng TY, Jiang XH, Li G. Bone marrow-derived mesenchymal stem cells promote growth and angiogenesis of breast and prostate tumors. Stem Cell Res Ther. 2013;4:70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 161]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
93.  Fernandes AM, Herlofsen SR, Karlsen TA, Küchler AM, Fløisand Y, Brinchmann JE. Similar properties of chondrocytes from osteoarthritis joints and mesenchymal stem cells from healthy donors for tissue engineering of articular cartilage. PLoS One. 2013;8:e62994.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 48]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
94.  Hoover DJ, Zhu V, Chen R, Briley K, Rameshwar P, Cohen S, Coffman FD. Expression of the chitinase family glycoprotein YKL-40 in undifferentiated, differentiated and trans-differentiated mesenchymal stem cells. PLoS One. 2013;8:e62491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
95.  Rippo MR, Babini L, Prattichizzo F, Graciotti L, Fulgenzi G, Tomassoni Ardori F, Olivieri F, Borghetti G, Cinti S, Poloni A. Low FasL levels promote proliferation of human bone marrow-derived mesenchymal stem cells, higher levels inhibit their differentiation into adipocytes. Cell Death Dis. 2013;4:e594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
96.  Mittag F, Falkenberg EM, Janczyk A, Götze M, Felka T, Aicher WK, Kluba T. Laminin-5 and type I collagen promote adhesion and osteogenic differentiation of animal serum-free expanded human mesenchymal stromal cells. Orthop Rev (Pavia). 2012;4:e36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
97.  Cunha MC, Lima Fda S, Vinolo MA, Hastreiter A, Curi R, Borelli P, Fock RA. Protein malnutrition induces bone marrow mesenchymal stem cells commitment to adipogenic differentiation leading to hematopoietic failure. PLoS One. 2013;8:e58872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 39]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
98.  Xin H, Chopp M, Shen LH, Zhang RL, Zhang L, Zhang ZG, Li Y. Multipotent mesenchymal stromal cells decrease transforming growth factor β1 expression in microglia/macrophages and down-regulate plasminogen activator inhibitor 1 expression in astrocytes after stroke. Neurosci Lett. 2013;542:81-86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
99.  Rasini V, Dominici M, Kluba T, Siegel G, Lusenti G, Northoff H, Horwitz EM, Schäfer R. Mesenchymal stromal/stem cells markers in the human bone marrow. Cytotherapy. 2013;15:292-306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 82]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
100.  Qian H, Badaloni A, Chiara F, Stjernberg J, Polisetti N, Nihlberg K, Consalez GG, Sigvardsson M. Molecular characterization of prospectively isolated multipotent mesenchymal progenitors provides new insight into the cellular identity of mesenchymal stem cells in mouse bone marrow. Mol Cell Biol. 2013;33:661-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 26]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
101.  Wong DJ, Liu H, Ridky TW, Cassarino D, Segal E, Chang HY. Module map of stem cell genes guides creation of epithelial cancer stem cells. Cell Stem Cell. 2008;2:333-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 532]  [Cited by in F6Publishing: 547]  [Article Influence: 34.2]  [Reference Citation Analysis (0)]
102.  Bhat KP, Balasubramaniyan V, Vaillant B, Ezhilarasan R, Hummelink K, Hollingsworth F, Wani K, Heathcock L, James JD, Goodman LD. Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell. 2013;24:331-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 700]  [Cited by in F6Publishing: 754]  [Article Influence: 68.5]  [Reference Citation Analysis (0)]
103.  Bao B, Ahmad A, Li Y, Azmi AS, Ali S, Banerjee S, Kong D, Sarkar FH. Targeting CSCs within the tumor microenvironment for cancer therapy: a potential role of mesenchymal stem cells. Expert Opin Ther Targets. 2012;16:1041-1054.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 34]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
104.  Yang MH, Hsu DS, Wang HW, Wang HJ, Lan HY, Yang WH, Huang CH, Kao SY, Tzeng CH, Tai SK. Bmi1 is essential in Twist1-induced epithelial-mesenchymal transition. Nat Cell Biol. 2010;12:982-992.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 489]  [Cited by in F6Publishing: 512]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
105.  Chapman EJ, Kelly G, Knowles MA. Genes involved in differentiation, stem cell renewal, and tumorigenesis are modulated in telomerase-immortalized human urothelial cells. Mol Cancer Res. 2008;6:1154-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 38]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
106.  Grimm C, Chavez L, Vilardell M, Farrall AL, Tierling S, Böhm JW, Grote P, Lienhard M, Dietrich J, Timmermann B. DNA-methylome analysis of mouse intestinal adenoma identifies a tumour-specific signature that is partly conserved in human colon cancer. PLoS Genet. 2013;9:e1003250.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
107.  Kong D, Li Y, Wang Z, Sarkar FH. Cancer Stem Cells and Epithelial-to-Mesenchymal Transition (EMT)-Phenotypic Cells: Are They Cousins or Twins. Cancers (Basel). 2011;3:716-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 253]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
108.  Boyer LA, Plath K, Zeitlinger J, Brambrink T, Medeiros LA, Lee TI, Levine SS, Wernig M, Tajonar A, Ray MK. Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature. 2006;441:349-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1937]  [Cited by in F6Publishing: 1912]  [Article Influence: 106.2]  [Reference Citation Analysis (0)]
109.  Lee TI, Jenner RG, Boyer LA, Guenther MG, Levine SS, Kumar RM, Chevalier B, Johnstone SE, Cole MF, Isono K. Control of developmental regulators by Polycomb in human embryonic stem cells. Cell. 2006;125:301-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1748]  [Cited by in F6Publishing: 1737]  [Article Influence: 96.5]  [Reference Citation Analysis (0)]
110.  Pengelly AR, Copur Ö, Jäckle H, Herzig A, Müller J. A histone mutant reproduces the phenotype caused by loss of histone-modifying factor Polycomb. Science. 2013;339:698-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 225]  [Cited by in F6Publishing: 216]  [Article Influence: 19.6]  [Reference Citation Analysis (0)]
111.  Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, Parada LF. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012;488:522-526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1518]  [Cited by in F6Publishing: 1612]  [Article Influence: 134.3]  [Reference Citation Analysis (0)]
112.  Driessens G, Beck B, Caauwe A, Simons BD, Blanpain C. Defining the mode of tumour growth by clonal analysis. Nature. 2012;488:527-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 543]  [Cited by in F6Publishing: 554]  [Article Influence: 46.2]  [Reference Citation Analysis (0)]
113.  Schepers AG, Snippert HJ, Stange DE, van den Born M, van Es JH, van de Wetering M, Clevers H. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science. 2012;337:730-735.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 831]  [Cited by in F6Publishing: 825]  [Article Influence: 68.8]  [Reference Citation Analysis (0)]
114.  Gilbertson RJ, Graham TA. Cancer: Resolving the stem-cell debate. Nature. 2012;488:462-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
115.  Medema JP. Cancer stem cells: the challenges ahead. Nat Cell Biol. 2013;15:338-344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 516]  [Cited by in F6Publishing: 525]  [Article Influence: 47.7]  [Reference Citation Analysis (0)]