Abstract
Mesenchymal stem cells (MSCs) are multipotent, self-renewing cells harboring multi-lineage differentiation potential and immunosuppressive properties that make them an attractive candidate for biological cell-based regenerative medicine. In addition to its undoubted clinical interest, controlling the fate and behaviors of MSCs is a crucial prerequisite for their therapeutic applications in regenerative medicine. Stem cell differentiation and modulation of functional activities are generally controlled by “cocktails” of growth factors, signaling molecules, and/or genetic manipulations. However, these approaches have several limiting factors, such as undefined conditions leading to heterogeneous populations of cells and unexpected risks of virus-mediated genetic modifications. Small molecules targeting specific signaling pathways have been shown to be key modulators in controlling stem cells’ fate and function. Small molecules are also important tools for understanding mechanistic and developmental processes. Furthermore, the precise mode of action of small molecules for controlling MSCs fate is still under study. However, Wnt, GSK, and other protein kinases signaling pathways are likely to be involved. These target-based manipulations of stem cells fate by small molecules provide new insights into stem cell biology, and facilitate the development of regenerative medicine using stem cells. Here, we review the recent progress in controlling MSCs fate and functional activities by small molecules.
Keywords: Mesenchymal stem cells, cell fate, differentiation, small molecules
Introduction
Stem and progenitor cells are less specialized cells that have both the ability for self-renewal and the potential to differentiate into specialized cells in response to specific signals [1]. Mesenchymal stem cells (MSCs) are derived from a population of stromal cells present in bone marrow and various tissues, which also can differentiate into different tissue lineages [2-4]. In the last decade, it has been well established that MSCs have the ability to differentiate various cell types encompassing osteoblasts, chondrocytes, myocytes, marrow stromal cells, tendon-ligament fibroblasts, adipocytes, and other mesenchymal phenotypes [5], suggesting their use as a source of cells for various application of regenerative medicine. In addition to their potentials, understanding the fundamental mechanisms to control cell fate and function is a critical step to translational application of MSCs. In many studies, stem cell fate and function have been generally controlled by “cocktails” of growth factors, signaling molecules, and/or genetic manipulations [6,7]. However, the composition and/or conditions of most of these cocktails are not exactly defined and they do not specifically regulate stem cell fate, resulting in heterogeneous population of cells. Because cell-based therapies may require large quantities of stem cells for clinical uses, it would be advantageous to isolate specific small molecules that either maintain self-renewal or drive tissue specific fates of stem cells [8]. In addition, virus-mediated genetic manipulation introduces unacceptable risks of permanent transgene integration to the genome. The resulting genomic alteration and possible reactivation of viral transgenes pose serious clinical concerns [9].
Small molecules have shown to be useful tools for modulating cell fate and function by targeting specific signals and mechanisms [10-12]. In addition, they provide some distinct advantages over other techniques, such as gene manipulation, preconditioning, and pretreatment with effectors including growth factors and cytokines that are fast, reversible, and precise temporal regulators of protein function [10]. Indeed, various cell permeable small molecules have been proven to be clearly useful for inducing the differentiation of various stem cells including MSCs. Moreover, the immense potential of orally delivered small molecules as regenerative therapies are now widely recognized [13]. In this review, we focus on recent advances in the use of small molecules to control MSCs fates such as proliferation, differentiation, and functional activity.
The control of cell fate by small molecules
Small molecules
Small molecules have been synthesized and tested in phenotypic analysis to screen biologically useful molecules for over 100 years [14]. Although one compound was tested at a time in the earliest era, modern libraries contain thousands and even millions of small molecules that are synthesized through combinatorial chemistry and tested by high-throughput screening techniques [14]. The complementary approach to classical genetics, where small molecules are exploited to probe biological functions, is termed “chemical genetics” [15,16]. While classical genetics establishes genotype-phenotype relationships through means of genetic manipulation, chemical genetics uses small molecules that intervene in signaling pathways through direct interaction with proteins to unravel relationships between proteins and phenotypes. By analogy to classical genetics, small compounds in chemical genetics are equivalent to mutations in classical genetics [17]. In fact, given that chemicals can be removed from cells by simple washing, they are analogues to conditional mutations. Although small molecules allow reversible temporal and dose-dependent control of protein function, one of their major limitations is that a single compound often affects multiple proteins and subsequently multiple pathways [18,19]. Thus, treatment with a single small molecule can be analogous to simultaneous modulations in several genes.
The method of using small molecules to modulate and control the stem cell fate has a long history. Indeed, all-trans-retinoic acid has been used for over 30 years to induce differentiation of both mouse and human embryonal carcinoma cells [20,21]. Hexamethylene-bisacetamide and DMSO also have been used to promote differentiation of embryonal carcinoma cells and erythroleukemia cells, respectively [22,23]. In addition, stem cells share many properties with tumor cells and these similarities can provide insights to control and direct cell behavior. Indeed, small molecules are already standard chemotherapeutics in the treatment for cancer [8]. A number of small molecules have been examined for anti-cancer effects (especially induction of apoptosis), and recently, for stem cell self-renewal and specific differentiation in potential approaches to regenerative medicine. Indeed, both natural and synthetic small molecules have been shown to be useful chemical tools for manipulating the fates of cells [8].
Modulation of stem cell fate by small molecules
As mentioned above, small molecules have been used to modulate and/or control cell fate, and behavior, especially stem cells, for a long time. In fact, most of the studies that are currently being conducted and/or are ongoing in this field focus on the revealing the mechanisms of self-renewal and differentiation of embryonic stem cells (ESCs) and the generation of induced pluripotent stem cells (iPSCs) [8,24]. Indeed, various small molecules have been identified to regulate the differentiation and self-renewal of ESCs. For example, Pluripotin, CHIR99021, and A83-01 can support self-renewal of ESCs through targeting the RasGAP, GSK-3, and ALK5 signaling pathways, respectively [25-27]. (-) Indolactam V can enhance the pancreatic differentiation of ESCs through PKC pathway, and stauprimide and SB431542 induces differentiation of ESCs through NME2 and ALK5 pathways [28,29]. In addition, several small molecules were revealed to affects somatic cell reprogramming. BIX-01294, RG108, and parnate can promote MEFs reprogramming through regulating G9a HMTase, DNA MTase, and lysine-specific demethylase, respectively [10,30,31]. Interestingly, a small molecule, Reversine, can induce dedifferentiation of muscle and fibroblast cells into a more primitive multipotent state [32-34].
Regulation of differentiation in MSCs by small molecules
Mesenchymal stem cells: A brief overview
MSCs are a heterogeneous subset of stromal cells that are classically derived from bone marrow [2]. But they have been isolated from most connective tissues including adipose tissue [35], periosteum [36], and synovial membrane [11]. MSCs have apparent advantages, such as easy availability, few ethical concerns, and low immunogenicity, compared to embryonic stem cells and other tissue-specific stem cells. Indeed, because MSCs can be easily isolated and expanded in culture, they can be administered immediately instead of waiting weeks and/or months for adequate numbers of cells to be achieved by cell culture. MSCs retain their growth and multilineage potential over several passages, although they are moral [37,38]. In addition to their multilineage transdifferentiation potential, one particularly useful characteristic of MSCs is their apparent immunoprivilege. They display local immunosuppressive properties that permit successful transplanting in an allogenic setting. Indeed, experiments in non-human primates have shown that allogenic MSCs were not rejected but showed similar effects on the autologous MSCs and were detected nine months after transplantation in the recipient [39-41].Overall, these studies have suggested MSCs as an attractive candidate cell type for tissue engineering, regenerative medicine, and autoimmune disease treatment.
Specific differentiation of MSCs by small molecules
For practical use of stem cells for regeneration therapy, there are at least three prerequisites: (i) the directed differentiation of stem cells to specific cell types, (ii) achievement of high survival rate of the cells after transplantation, and (iii) prevention of undifferentiated stem cells that are prone to form teratomas and/or cancers [42]. Amongst them, the ex vivo directed differentiation of stem cells to specific cell types for treating target diseases may provide better clinical results. Classically, osteogenic differentiation of human MSCs requires incubation in fetal bovine serum (FBS)-containing medium supplemented with ascorbic acid, β-glycerophosphate, and dexamethasone, resulting in an increase in alkaline phosphatase activity and calcium deposition [43,44]. The chondrogenic differentiation is performed with a high cell-density pellet or a micromass culture treated with transforming growth factor (TGF)-β in a serum-free medium; this results in the production of cartilage-specific, highly sulfated proteoglycans and type II collagen [45]. For adipogenic differentiation, MSCs are treated with FBS containing medium supplemented with dexamethasone, insulin, isobutyl methyl xanthine, and indomethacin. The differentiation is detected bythe oil red O staining for lipid vacuoles [45]. In addition to these reports, many studies have shown that a number of small molecules can be used to control the fate of MSCs for a variety of applications. For example, 5-azacytidine (5-aza-C) can induce murine and human bone marrow stromal cells into cardiomyocytes [46-48], and also induce mouse mesenchymal progenitor cell line, C3H10T1/2 cells, into myoblast, osteoblasts, adipocytes, and chondrocytes (Table 1) [43]. It has been revealed that 5-aza-C does not directly induce or activate the specific differentiation pathways of MSCs, but rather convert the cells into a competent spontaneous differentiation state [49]. However, recent reports showed that 5-aza-C-treated human mesenchymal stem/progenitor cells derived from umbilical cord, cord blood and bone marrow do not generate cardiomyocytes in vitro at high frequencies [50,51]. This suggests that the induction of specific differentiation by small molecules might be dependent on cell types. All-trans retinoic acid is one of the prominent molecules that has long been in control of cell fate. Recently, it was reported that retinoic acid may induce chondrogenesis, osteoblastogenesis, and neuronal differentiation of MSCs [52-54]. Peroxisome proliferator activated receptor γ (PPARγ) agonist and antagonists are used as adipogenesis modulators [55,56]. Dexamethasone, ascorbic acid, and β-glycerophosphate can also be used to induce osteogenesis or adipogenesis of MSCs under defined conditions (Table 1) [43,57].
Table 1.
Small molecules in controlling the fate of mesenchymal stem cells
|
To identify small molecules that selectively differentiate MSCs into defined-lineages, we screened a small library (41 compounds) of characterized and commercially available inhibitors of six major subfamilies of protein kinases that are known to inhibit various cellular processes: TK (tyrosine kinase families), TKL (tyrosine kinase-like families), CMGC (CDK, MAPK, GSK3, CLK families), CAMK (Ca/calmodulin dependent protein kinase), AGC (PKA, PKG, and PKC families), and CKI (Casein kinase family) [58]. Because such important cellular processes are likely to be controlled by a complicated orchestration of many signaling pathways including many undiscovered ones, common signal modulators, such as protein kinases, are likely to play an important role in balancing multiple signals affecting several aspects of the above prerequisites [42]. Protein kinases belong to one of the largest protein families in the human genome, and they play critical roles in signaling pathways implicated in the development, differentiation, proliferation, and death of cells. Several signaling pathways have been known to induce or suppress differentiation of stem cells, such as the pathways involving mitogen-activated kinases, glycogen synthase kinase-3, PI3-kinase, and others [24,44,59-61]. A glycogen synthase kinase 3β(GSK-3β) inhibitor synthesized by combinatorial chemistry has been shown to induce neurogenesis [24]. In this study, among several candidates that drive rat MSCs into specific cell types, H-89, an inhibitor of protein kinase A, was found to be potentially implicated in chondrogenesis of the MSCs, which is a derivative of isoquinolinesulfonamide, N-[2-((p-bromocinnamyl) amino)ethyl]-5-isoquinolinesulfonamide, 2HCl [42]. We also found that H-1152, a Rho kinase inhibitor, was the best inducer of differentiation of ESCs into tyrosine hydroxylase (TH), a catecholaminergic neuronal marker, -positive neurons. H-1152 is another cell-permeable isoquinoline sulfonamide derivative that acts as a highly specific, potent, and ATP-competitive inhibitor of G protein ROCK (Ki=1.6 nM) [42]. Based on the above results, the enhanced differentiation of rat MSCs to specific lineages support the notion that, although cell differentiation may be the result of a complex orchestration of many signals from multiple signaling pathways, even a single chemical reagent is enough to alter the relative balance of many signals to enhance differentiation of stem cells to particular cell types. They also suggest that the process may be optimized by a “mixture” of various multiple kinase inhibitors as well as other kinds of small molecules. Because these compounds may interact with “off-target” kinases as well as other unknown proteins, our observation should be considered as a practical approach to finding chemical reagents for inducing stem cell differentiation to a specific cell type even when most of the signaling pathways are unknown [42]. Thus, the approach described or some variation of it may be applied to other stem cells, including human stem cells, to find chemical molecules that can trigger initiation, inhibition, or even reversion of the differentiation process of stem cells or progenitor cells.
In recent studies, we found that phorbol myristate acetate (PMA), a PKC activator, and can upregulate cardiogenic properties from MSCs and subsequently chemically activated cardiogenic MSCs preventing sudden deaths after engraftment onto infarcted rats by electromechanically synchronizing with the host myocardium [62]. These suggest that small molecules may regulate the stem cell functions as well as the fate. In addition, we also found that a kind of glycogen synthase kinase-3 (GSK-3) inhibitor may induce the endothelial differentiation of MSCs in recent studies, which might be an important implication for treating vascular diseases (unpublished).
Wu et al. also identified a small molecule that selectively differentiates MSCs into osteogenic lineages through screening a combinatorial heterocyclic compound library in the mouse mesenchymal progenitor cells [63]. Purmorphamine, 2,6,9-trisubstituted purine compound, was identified as a potent osteoblast differentiation inducing agent that can activate Cbfa1/Runx2 (a key regulator of bone development) as well as upregulate other bone specific markers, such as osteopontin and collagen I. In addition, it was reported that decalpenic acid, a novel small molecule from Penicillium-verruculosum CR37010, induces early osteoblastic markers in mouse C3H10T1/2 cells [64]. Pevsner-Fischer et al. reported that toll-like receptors are associated with MSCs function and their inhibitor, Pam3Cys, increases MSC proliferation and inhibits MSC differentiation into chondrocytes, osteocytes, and adipocytes (Table 1) [65].
Concluding remarks
Although substantial growth arose in recent years, stem cell research and development are still in an early stage. The control of stem cell fate and function is an important step for clinical applications of stem cells in regenerative medicine. In addition to the focus on pluripotent stem cells including ESCs and iPSCs, understanding and controlling the fate and function of adult stem cells such as, MSCs, in vitro and in vivo might be a significant challenge for developing better therapeutic approaches to regenerative medicine. Among various approaches in controlling stem cell fate, small molecules are considered as the best tool in the next era, because small molecules can target signaling transduction pathways (for example, tyrosine kinase receptors) and affect various cellular events, such as, DNA replication, differentiation, proliferation, and apoptosis. Obviously the field is interdisciplinary and involves chemists, a range of biologists, and bioinformaticians. Indeed, chemists have to build small molecule libraries based on the structural motifs of known and/or potential regulators in specific pathways, and stem cell biologists may have to study to better understand the molecular interactions between small molecules and specific behaviors of stem cells. Small molecules are also important tools for understanding mechanistic and developmental processes because they can be added or removed at any point during development while mutations tend to persist throughout the organism’s life [8]. Such temporal control is critical in understanding the specific timing of developmental processes [66]. Because of the current challenges in stem cell biology to control stem cell fate and function, small molecules might emerge as a powerful strategy to identify drugs that may regulate stem cell activity and may ultimately be useful to in vivo stem cell biology and therapy.
Acknowledgements
This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (2010-0023615) and a Korea Science and Engineering Foundation Grant funded by theKorea government (MEST) (2011-0019243, 2011-0019254). It was also supported by a grant (SC-2150) from the Stem Cell Research Center of the 21st Century Frontier Research Program funded by MOEST, and a grant from the Korea Health 21 R & D Project, Ministry of Health & Welfare, Republic of Korea (A085136).
References
- 1.Segers VF, Lee RT. Stem-cell therapy for cardiac disease. Nature. 2008;451:937–942. doi: 10.1038/nature06800. [DOI] [PubMed] [Google Scholar]
- 2.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. doi: 10.1126/science.284.5411.143. [DOI] [PubMed] [Google Scholar]
- 3.Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, Du J, Aldrich S, Lisberg A, Low WC, Largaespada DA, Verfaillie CM. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002;418:41–49. doi: 10.1038/nature00870. [DOI] [PubMed] [Google Scholar]
- 4.Lee OK, Kuo TK, Chen WM, Lee KD, Hsieh SL, Chen TH. Isolation of multipotent mesenchymal stem cells from umbilical cord blood. Blood. 2004;103:1669–1675. doi: 10.1182/blood-2003-05-1670. [DOI] [PubMed] [Google Scholar]
- 5.Bartholomew A, Sturgeon C, Siatskas M, Ferrer K, McIntosh K, Patil S, Hardy W, Devine S, Ucker D, Deans R, Moseley A, Hoffman R. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Experimental hematology. 2002;30:42–48. doi: 10.1016/s0301-472x(01)00769-x. [DOI] [PubMed] [Google Scholar]
- 6.Sasaki K, Ichikawa H, Takei S, No HS, Tomotsune D, Kano Y, Yokoyama T, Sirasawa S, Mogi A, Yoshie S, Sasaki S, Yamada S, Matsumoto K, Mizuguchi M, Yue F, Tanaka Y. Hepatocyte differentiation from human ES cells using the simple embryoid body formation method and the staged-additional cocktail. ScientificWorldJournal. 2009;9:884–890. doi: 10.1100/tsw.2009.97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Otsuru S, Rasini V, Bussolari R, Hofmann TJ, Dominici M, Horwitz EM. Cytokine-induced osteopoietic differentiation of transplanted marrow cells. Blood. 2011;118:2358–61. doi: 10.1182/blood-2011-01-331397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Schugar RC, Robbins PD, Deasy BM. Small molecules in stem cell self-renewal and differentiation. Gene therapy. 2008;15:126–135. doi: 10.1038/sj.gt.3303062. [DOI] [PubMed] [Google Scholar]
- 9.Feng B, Ng JH, Heng JC, Ng HH. Molecules that promote or enhance reprogramming of somatic cells to induced pluripotent stem cells. Cell stem cell. 2009;4:301–312. doi: 10.1016/j.stem.2009.03.005. [DOI] [PubMed] [Google Scholar]
- 10.Li W, Ding S. Small molecules that modulate embryonic stem cell fate and somatic cell reprogramming. Trends in pharmacological sciences. 2010;31:36–45. doi: 10.1016/j.tips.2009.10.002. [DOI] [PubMed] [Google Scholar]
- 11.De Bari C, Dell'Accio F, Tylzanowski P, Luyten FP. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis and rheumatism. 2001;44:1928–1942. doi: 10.1002/1529-0131(200108)44:8<1928::AID-ART331>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
- 12.Barbaric I, Gokhale PJ, Jones M, Glen A, Baker D, Andrews PW. Novel regulators of stem cell fates identified by a multivariate phenotype screen of small compounds on human embryonic stem cell colonies. Stem cell research. 2010;5:104–119. doi: 10.1016/j.scr.2010.04.006. [DOI] [PubMed] [Google Scholar]
- 13.Jagasia R, Song H, Gage FH, Lie DC. New regulators in adult neurogenesis and their potential role for repair. Trends in molecular medicine. 2006;12:400–405. doi: 10.1016/j.molmed.2006.07.006. [DOI] [PubMed] [Google Scholar]
- 14.Emre N, Coleman R, Ding S. A chemical approach to stem cell biology. Current opinion in chemical biology. 2007;11:252–258. doi: 10.1016/j.cbpa.2007.04.024. [DOI] [PubMed] [Google Scholar]
- 15.Mitchison TJ. Towards a pharmacological genetics. Chemistry & biology. 1994;1:3–6. doi: 10.1016/1074-5521(94)90034-5. [DOI] [PubMed] [Google Scholar]
- 16.Schreiber SL. Chemical genetics resulting from a passion for synthetic organic chemistry. Bioorganic & medicinal chemistry. 1998;6:1127–1152. doi: 10.1016/s0968-0896(98)00126-6. [DOI] [PubMed] [Google Scholar]
- 17.Barbaric I, Gokhale PJ, Andrews PW. High-content screening of small compounds on human embryonic stem cells. Biochemical Society transactions. 2010;38:1046–1050. doi: 10.1042/BST0381046. [DOI] [PubMed] [Google Scholar]
- 18.Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors. The Biochemical journal. 2000;351:95–105. doi: 10.1042/0264-6021:3510095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bain J, Plater L, Elliott M, Shpiro N, Hastie CJ, McLauchlan H, Klevernic I, Arthur JS, Alessi DR, Cohen P. The selectivity of protein kinase inhibitors: a further update. The Biochemical journal. 2007;408:297–315. doi: 10.1042/BJ20070797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Strickland S, Mahdavi V. The induction of differentiation in teratocarcinoma stem cells by retinoic acid. Cell. 1978;15:393–403. doi: 10.1016/0092-8674(78)90008-9. [DOI] [PubMed] [Google Scholar]
- 21.Andrews PW. Retinoic acid induces neuronal differentiation of a cloned human embryonal carcinoma cell line in vitro. Developmental biology. 1984;103:285–293. doi: 10.1016/0012-1606(84)90316-6. [DOI] [PubMed] [Google Scholar]
- 22.Jakob H, Dubois P, Eisen H, Jacob F. [Effects of hexamethylenebisacetamide on the differentiation of embryonal carcinoma cells] . C R Acad Sci Hebd Seances Acad Sci D. 1978;286:109–111. [PubMed] [Google Scholar]
- 23.Friend C, Scher W, Holland JG, Sato T. Hemoglobin synthesis in murine virus-induced leukemic cells in vitro: stimulation of erythroid differentiation by dimethyl sulfoxide. Proceedings of the National Academy of Sciences of the United States of America. 1971;68:378–382. doi: 10.1073/pnas.68.2.378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ding S, Wu TY, Brinker A, Peters EC, Hur W, Gray NS, Schultz PG. Synthetic small molecules that control stem cell fate. Proceedings of the National Academy of Sciences of the United States of America. 2003;100:7632–7637. doi: 10.1073/pnas.0732087100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chen S, Do JT, Zhang Q, Yao S, Yan F, Peters EC, Scholer HR, Schultz PG, Ding S. Self-renewal of embryonic stem cells by a small molecule. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:17266–17271. doi: 10.1073/pnas.0608156103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ying QL, Wray J, Nichols J, Batlle-Morera L, Doble B, Woodgett J, Cohen P, Smith A. The ground state of embryonic stem cell self-renewal. Nature. 2008;453:519–523. doi: 10.1038/nature06968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Li W, Zhou H, Abujarour R, Zhu S, Young Joo J, Lin T, Hao E, Scholer HR, Hayek A, Ding S. Generation of human-induced pluripotent stem cells in the absence of exogenous Sox2. Stem cells. 2009;27:2992–3000. doi: 10.1002/stem.240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zhu S, Wurdak H, Wang J, Lyssiotis CA, Peters EC, Cho CY, Wu X, Schultz PG. A small molecule primes embryonic stem cells for differentiation. Cell stem cell. 2009;4:416–426. doi: 10.1016/j.stem.2009.04.001. [DOI] [PubMed] [Google Scholar]
- 29.Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M, Studer L. Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27:275–280. doi: 10.1038/nbt.1529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Shi Y, Do JT, Desponts C, Hahm HS, Scholer HR, Ding S. A combined chemical and genetic approach for the generation of induced pluripotent stem cells. Cell stem cell. 2008;2:525–528. doi: 10.1016/j.stem.2008.05.011. [DOI] [PubMed] [Google Scholar]
- 31.Shi Y, Desponts C, Do JT, Hahm HS, Scholer HR, Ding S. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small-molecule compounds. Cell stem cell. 2008;3:568–574. doi: 10.1016/j.stem.2008.10.004. [DOI] [PubMed] [Google Scholar]
- 32.Lee EK, Bae GU, You JS, Lee JC, Jeon YJ, Park JW, Park JH, Ahn SH, Kim YK, Choi WS, Kang JS, Han G, Han JW. Reversine increases the plasticity of lineage-committed cells toward neuroectodermal lineage. The Journal of biological chemistry. 2009;284:2891–2901. doi: 10.1074/jbc.M804055200. [DOI] [PubMed] [Google Scholar]
- 33.Anastasia L, Sampaolesi M, Papini N, Oleari D, Lamorte G, Tringali C, Monti E, Galli D, Tettamanti G, Cossu G, Venerando B. Reversine-treated fibroblasts acquire myogenic competence in vitro and in regenerating skeletal muscle. Cell death and differentiation. 2006;13:2042–2051. doi: 10.1038/sj.cdd.4401958. [DOI] [PubMed] [Google Scholar]
- 34.Shan SW, Tang MK, Chow PH, Maroto M, Cai DQ, Lee KK. Induction of growth arrest and polycomb gene expression by reversine allows C2C12 cells to be reprogrammed to various differentiated cell types. Proteomics. 2007;7:4303–4316. doi: 10.1002/pmic.200700636. [DOI] [PubMed] [Google Scholar]
- 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. Molecular biology of the cell. 2002;13:4279–4295. doi: 10.1091/mbc.E02-02-0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nakahara H, Dennis JE, Bruder SP, Haynesworth SE, Lennon DP, Caplan AI. In vitro differentiation of bone and hypertrophic cartilage from periosteal-derived cells. Experimental cell research. 1991;195:492–503. doi: 10.1016/0014-4827(91)90401-f. [DOI] [PubMed] [Google Scholar]
- 37.Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. Journal of cell science. 2000;113:1161–1166. doi: 10.1242/jcs.113.7.1161. [DOI] [PubMed] [Google Scholar]
- 38.Graf T. Differentiation plasticity of hematopoietic cells. Blood. 2002;99:3089–3101. doi: 10.1182/blood.v99.9.3089. [DOI] [PubMed] [Google Scholar]
- 39.Devine SM, Bartholomew AM, Mahmud N, Nelson M, Patil S, Hardy W, Sturgeon C, Hewett T, Chung T, Stock W, Sher D, Weissman S, Ferrer K, Mosca J, Deans R, Moseley A, Hoffman R. Mesenchymal stem cells are capable of homing to the bone marrow of nonhuman primates following systemic infusion. Experimental hematology. 2001;29:244–255. doi: 10.1016/s0301-472x(00)00635-4. [DOI] [PubMed] [Google Scholar]
- 40.Devine SM, Cobbs C, Jennings M, Bartholomew A, Hoffman R. Mesenchymal stem cells distribute to a wide range of tissues following systemic infusion into nonhuman primates. Blood. 2003;101:2999–3001. doi: 10.1182/blood-2002-06-1830. [DOI] [PubMed] [Google Scholar]
- 41.Ghosh-Choudhury N, Abboud SL, Mahimainathan L, Chandrasekar B, Choudhury GG. Phosphatidylinositol 3-kinase regulates bone morphogenetic protein-2 (BMP-2)-induced myocyte enhancer factor 2A-dependent transcription of BMP-2 gene in cardiomyocyte precursor cells. The Journal of biological chemistry. 2003;278:21998–22005. doi: 10.1074/jbc.M302277200. [DOI] [PubMed] [Google Scholar]
- 42.Hwang KC, Kim JY, Chang W, Kim DS, Lim S, Kang SM, Song BW, Ha HY, Huh YJ, Choi IG, Hwang DY, Song H, Jang Y, Chung N, Kim SH, Kim DW. Chemicals that modulate stem cell differentiation. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:7467–7471. doi: 10.1073/pnas.0802825105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP. Osteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vitro. Journal of cellular biochemistry. 1997;64:295–312. [PubMed] [Google Scholar]
- 44.Castelo-Branco G, Rawal N, Arenas E. GSK-3β inhibition/beta-catenin stabilization in ventral midbrain precursors increases differentiation into dopamine neurons. Journal of cell science. 2004;117:5731–5737. doi: 10.1242/jcs.01505. [DOI] [PubMed] [Google Scholar]
- 45.Augello A, De Bari C. The regulation of differentiation in mesenchymal stem cells. Human gene therapy. 2010;21:1226–1238. doi: 10.1089/hum.2010.173. [DOI] [PubMed] [Google Scholar]
- 46.Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama H, Pan J, Sano M, Takahashi T, Hori S, Abe H, Hata J, Umezawa A, Ogawa S. Cardiomyocytes can be generated from marrow stromal cells in vitro. The Journal of clinical investigation. 1999;103:697–705. doi: 10.1172/JCI5298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Fukuda K. Application of mesenchymal stem cells for the regeneration of cardiomyocyte and its use for cell transplantation therapy. Hum cell. 2003;16:83–94. doi: 10.1111/j.1749-0774.2003.tb00138.x. [DOI] [PubMed] [Google Scholar]
- 48.Shim WS, Jiang S, Wong P, Tan J, Chua YL, Tan YS, Sin YK, Lim CH, Chua T, Teh M, Liu TC, Sim E. Ex vivo differentiation of human adult bone marrow stem cells into cardiomyocyte-like cells. Biochemical and biophysical research communications. 2004;324:481–488. doi: 10.1016/j.bbrc.2004.09.087. [DOI] [PubMed] [Google Scholar]
- 49.Ding S, Schultz PG. A role for chemistry in stem cell biology. Nat Biotechnol. 2004;22:833–840. doi: 10.1038/nbt987. [DOI] [PubMed] [Google Scholar]
- 50.Martin-Rendon E, Sweeney D, Lu F, Girdlestone J, Navarrete C, Watt SM. 5-Azacytidine-treated human mesenchymal stem/progenitor cells derived from umbilical cord, cord blood and bone marrow do not generate cardiomyocytes in vitro at high frequencies. Vox sanguinis. 2008;95:137–148. doi: 10.1111/j.1423-0410.2008.01076.x. [DOI] [PubMed] [Google Scholar]
- 51.Roura S, Farre J, Hove-Madsen L, Prat-Vidal C, Soler-Botija C, Galvez-Monton C, Vilalta M, Bayes-Genis A. Exposure to cardiomyogenic stimuli fails to transdifferentiate human umbilical cord blood-derived mesenchymal stem cells. Basic research in cardiology. 2010;105:419–430. doi: 10.1007/s00395-009-0081-8. [DOI] [PubMed] [Google Scholar]
- 52.Zhang W, Yan Q, Zeng YS, Zhang XB, Xiong Y, Wang JM, Chen SJ, Li Y, Bruce IC, Wu W. Implantation of adult bone marrow-derived mesenchymal stem cells transfected with the neurotrophin-3 gene and pretreated with retinoic acid in completely transected spinal cord. Brain research. 2010;1359:256–271. doi: 10.1016/j.brainres.2010.08.072. [DOI] [PubMed] [Google Scholar]
- 53.Henderson SE, Santangelo KS, Bertone AL. Chondrogenic effects of exogenous retinoic acid or a retinoic acid receptor antagonist (LE135) on equine chondrocytes and bone marrow-derived mesenchymal stem cells in monolayer culture. American journal of veterinary research. 2011;72:884–892. doi: 10.2460/ajvr.72.7.884. [DOI] [PubMed] [Google Scholar]
- 54.Dingwall M, Marchildon F, Gunanayagam A, Louis CS, Wiper-Bergeron N. Retinoic acid-induced Smad3 expression is required for the induction of osteoblastogenesis of mesenchymal stem cells. Differentiation : research in biological diversity. 2011;82:57–65. doi: 10.1016/j.diff.2011.05.003. [DOI] [PubMed] [Google Scholar]
- 55.Benvenuti S, Cellai I, Luciani P, Deledda C, Baglioni S, Giuliani C, Saccardi R, Mazzanti B, Dal Pozzo S, Mannucci E, Peri A, Serio M. Rosiglitazone stimulates adipogenesis and decreases osteoblastogenesis in human mesenchymal stem cells. Journal of endocrinological investigation. 2007;30:RC26–30. doi: 10.1007/BF03350807. [DOI] [PubMed] [Google Scholar]
- 56.Muruganandan S, Parlee SD, Rourke JL, Ernst MC, Goralski KB, Sinal CJ. Chemerin, a Novel Peroxisome Proliferator-activated Receptor γ (PPARγ) Target Gene That Promotes Mesenchymal Stem Cell Adipogenesis. The Journal of biological chemistry. 2011;286:23982–23995. doi: 10.1074/jbc.M111.220491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Grigoriadis AE, Heersche JN, Aubin JE. Differentiation of muscle, fat, cartilage, and bone from progenitor cells present in a bone-derived clonal cell population: effect of dexamethasone. The Journal of cell biology. 1988;106:2139–2151. doi: 10.1083/jcb.106.6.2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002;298:1912–1934. doi: 10.1126/science.1075762. [DOI] [PubMed] [Google Scholar]
- 59.Jaiswal RK, Jaiswal N, Bruder SP, Mbalaviele G, Marshak DR, Pittenger MF. Adult human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by mitogen-activated protein kinase. The Journal of biological chemistry. 2000;275:9645–9652. doi: 10.1074/jbc.275.13.9645. [DOI] [PubMed] [Google Scholar]
- 60.Hu X, Jin L, Feng L. Erk1/2 but not PI3K pathway is required for neurotrophin 3-induced oligodendrocyte differentiation of post-natal neural stem cells. Journal of neurochemistry. 2004;90:1339–1347. doi: 10.1111/j.1471-4159.2004.02594.x. [DOI] [PubMed] [Google Scholar]
- 61.Koyanagi M, Haendeler J, Badorff C, Brandes RP, Hoffmann J, Pandur P, Zeiher AM, Kuhl M, Dimmeler S. Non-canonical Wnt signaling enhances differentiation of human circulating progenitor cells to cardiomyogenic cells. The Journal of biological chemistry. 2005;280:16838–16842. doi: 10.1074/jbc.M500323200. [DOI] [PubMed] [Google Scholar]
- 62.Song H, Hwang HJ, Chang W, Song BW, Cha MJ, Kim IK, Lim S, Choi EJ, Ham O, Lee CY, Park JH, Lee SY, Choi E, Lee C, Lee M, Lee MH, Kim SH, Jang Y, Hwang KC. Cardiomyocytes from phorbol myristate acetate-activated mesenchymal stem cells restore electromechanical function in infarcted rat hearts. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:296–301. doi: 10.1073/pnas.1015873107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wu X, Ding S, Ding Q, Gray NS, Schultz PG. A small molecule with osteogenesis-inducing activity in multipotent mesenchymal progenitor cells. Journal of the American Chemical Society. 2002;124:14520–14521. doi: 10.1021/ja0283908. [DOI] [PubMed] [Google Scholar]
- 64.Sakamoto S, Kojima F, Igarashi M, Sawa R, Umekita M, Kubota Y, Nakae K, Yamaguchi S, Adachi H, Nishimura Y, Akamatsu Y. Decalpenic acid, a novel small molecule from Penicillium verruculosum CR37010, induces early osteoblastic markers in pluripotent mesenchymal cells. The Journal of antibiotics. 2010;63:703–708. doi: 10.1038/ja.2010.114. [DOI] [PubMed] [Google Scholar]
- 65.Pevsner-Fischer M, Morad V, Cohen-Sfady M, Rousso-Noori L, Zanin-Zhorov A, Cohen S, Cohen IR, Zipori D. Toll-like receptors and their ligands control mesenchymal stem cell functions. Blood. 2007;109:1422–1432. doi: 10.1182/blood-2006-06-028704. [DOI] [PubMed] [Google Scholar]
- 66.Peterson RT, Mably JD, Chen JN, Fishman MC. Convergence of distinct pathways to heart patterning revealed by the small molecule concentramide and the mutation heart-and-soul. Current biology : CB. 2001;11:1481–1491. doi: 10.1016/s0960-9822(01)00482-1. [DOI] [PubMed] [Google Scholar]
- 67.Fukuda K. Use of adult marrow mesenchymal stem cells for regeneration of cardiomyocytes. Bone marrow transplantation. 2003;32(Suppl 1):S25–27. doi: 10.1038/sj.bmt.1703940. [DOI] [PubMed] [Google Scholar]
- 68.Liao J, Hammerick KE, Challen GA, Goodell MA, Kasper FK, Mikos AG. Investigating the role of hematopoietic stem and progenitor cells in regulating the osteogenic differentiation of mesenchymal stem cells in vitro. Journal of orthopaedic research : official publication of the Orthopaedic Research Society. 2011;29:1544–53. doi: 10.1002/jor.21436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Potdar PD, D'Souza SB. Ascorbic acid induces in vitro proliferation of human subcutaneous adipose tissue derived mesenchymal stem cells with upregulation of embryonic stem cell pluripotency markers Oct4 and SOX 2. Hum cell. 2010;23:152–155. doi: 10.1111/j.1749-0774.2010.00095.x. [DOI] [PubMed] [Google Scholar]
- 70.Wu X, Walker J, Zhang J, Ding S, Schultz PG. Purmorphamine induces osteogenesis by activation of the hedgehog signaling pathway. Chemistry & biology. 2004;11:1229–1238. doi: 10.1016/j.chembiol.2004.06.010. [DOI] [PubMed] [Google Scholar]
