search
for
 About Bioline  All Journals  Testimonials  Membership  News


Journal of Postgraduate Medicine
Medknow Publications and Staff Society of Seth GS Medical College and KEM Hospital, Mumbai, India
ISSN: 0022-3859 EISSN: 0972-2823
Vol. 53, Num. 2, 2007, pp. 121-127

Journal of Postgraduate Medicine, Vol. 53, No. 2, April-June, 2007, pp. 121-127

Review Article

Adult mesenchymal stem cells: Differentiation potential and therapeutic applications

Wolfson Centre for Stem Cells, Tissue Engineering and Modelling ('STEM'), University of Nottingham, Nottingham NG7 2UH
Correspondence Address:Wolfson Centre for Stem Cells, Tissue Engineering and Modelling ('STEM'), University of Nottingham, Nottingham NG7 2UH, virginie.sottile@nottingham.ac.uk

Date of Submission: 31-Oct-2006
Date of Decision: 19-Dec-2006
Date of Acceptance: 17-Jan-2007

Code Number: jp07044

Abstract

Adult mesenchymal stem cells (MSCs) are a population of multipotent cells found primarily in the bone marrow. They have long been known to be capable of osteogenic, adipogenic and chondrogenic differentiation and are currently the subject of a number of trials to assess their potential use in the clinic. Recently, the plasticity of these cells has come under close scrutiny as it has been suggested that they may have a differentiation potential beyond the mesenchymal lineage. Myogenic and in particular cardiomyogenic potential has been shown in vitro . MSCs have also been shown to have the ability to form neural cells both in vitro and in vivo , although the molecular mechanisms underlying these apparent transdifferentiation events are yet to be elucidated. We describe here the cellular characteristics and differentiation potential of MSCs, which represent a promising stem cell population for future applications in regenerative medicine.

Keywords: Adipogenesis, chondrogenesis, mesenchymal stem cells, osteogenesis, regenerative medicine

Adult stem cells′ is the collective term used to describe postnatal stem cells which, as opposed to embryonic or fetal stem cells, persist throughout life and fulfill functions to repair or replace cells within certain tissues in response to traumatic events or natural cell turnover. Among tissues identified to harbor stem cells throughout postnatal life, bone marrow has been studied for many years as a source of both hematopoietic stem cells (HSCs) and mesenchymal stem cells (MSCs). Mesenchymal stem cells reside in the stromal fraction of the bone marrow, which provides the cellular microenvironment supporting hematopoeisis. Mesenchymal stem cells were first described as bone-forming progenitors from the stromal fraction of rats by Friedenstein and Petrakova in 1966[1] and Friedenstein went on to pioneer in vitro culture methods for the isolation and differentiation of MSCs.[2] MSCs have subsequently been shown to differentiate into a number of mesenchymal cell types including osteoblasts, chondrocytes and adipocytes.[3]

Sources of Primary MSCs

MSCs are typically isolated from the stromal fraction of adult bone marrow. In fresh bone marrow, MSCs account for only 0.01-0.0001% of nucleated marrow cells.[4],[5] Murine MSCs are classically obtained from the femurs and tibias of mice by flushing the marrow out of the bones with culture medium and transferring the resultant cell suspension in culture. Human MSCs can be similarly obtained from healthy volunteers by taking aspirates of bone marrow from the iliac crest and expanding on tissue-culture plastic.[6] Over recent years, MSC-like cells have also been identified in a number of different tissues.[2] Cells exhibiting MSC morphology and cellular characteristics have been isolated from adult peripheral blood,[7] adipose tissue,[8] skin tissue,[9] trabecular bone,[10] as well as fetal blood, liver, bone marrow[11] and lung.[12] Further MSC-like populations have been discovered in umbilical cord blood[13] and within the chorionic villi of the placenta.[14] Amniotic fluid has also been cited as a source of MSCs, with potential far-reaching implications for such areas as prenatal diagnosis and gene therapy.[15]

Isolation of Primary MSCs

It is notable that, contrary to most biological systems, human MSCs are better characterized than animal MSCs. This is probably due to the fact that MSCs are easily isolated and expanded from adult human tissue collected from healthy volunteers. Mesenchymal stem cells have nevertheless been isolated from a number of other species. Along with human MSCs, the better characterized cultures are those of rat[16] and mouse[17] origin, although therapeutic potential in large animal models has been investigated with MSCs from horse,[18] cow,[19] pig,[20] dog,[21] sheep[22] and baboon.[23] Three main approaches have been described for the isolation of MSCs and can either be used independently or combined together to obtain a more homogeneous culture. The traditional isolation method relies on the fact that MSCs selectively adhere to plastic surfaces, whereas hematopoietic cells do not and can therefore be removed through medium changes.[24] Whilst this eliminates most contaminating cells, the remaining heterogeneity of the culture progressively decreases by serial passaging and after a number of passages the culture is enriched in the self-renewing fraction, the stem cells. Another published isolation protocol involves centrifugation over a Percoll gradient, which separates cell populations based on their density and allows the enrichment of nucleated cells.[4] However, both methods are quite nonspecific and an approach that is now increasingly being used, resorts to sorting of bone marrow populations by flow cytometry (FACS), based on MSC reactivity to a number of antibodies. This can either be achieved by positively selecting for expressed antigens or by a process of immunodepletion of cells expressing hematopoietic and/or other lineage antigens. For instance, antibodies against CD34, a surface marker found on hematopoietic cells, are frequently used to identify and remove nonmesenchymal cells from a marrow culture.[3] Since there is no single specific marker available to unequivocally identify the MSC, different groups have opted for a variety of marker combinations. MSCs appear relatively stable as primary cultures[25],[26] although spontaneous transformation events have been observed in long-term cultures.[27]

Characterization of MSCs

Markers used by different groups to identify the MSC fraction from human bone marrow include, but are not limited to, CD13, CD29, CD31, CD44, CD54, CD63, CD73, CD105, CD106, CD140b, CD166 and Stro1.[3],[28],[29],[30],[31],[32] Comparisons of the various combinations used by different investigators show that the majority of subsets include either CD29, CD105 or both. We have established that over 95% of cells from the human MSC culture used in our lab express both CD29 and CD105 [Figure - 1]B. Although these markers have been used by various groups, there is still no general consensus on the optimal marker combination for MSCs. Some of this conjecture may be due to variations in sample origin, culture techniques and media composition among laboratories or differences in the age of the donors from which the MSCs were obtained and used for immunophenotyping. Because different antibody subsets are likely to selectively isolate slightly different cell types, comparison and evaluation of published data arising from different groups can be difficult. For instance, some groups report a degree of heterogeneity in their cultures after isolation and purification, with occasional description of a subset of small rounded cells among the more common fibroblast-like phenotype[33] [Figure - 1]A. This ambiguity begs the question of what MSCs are: do they purely represent the proliferating fibroblastic-like progenitors from the bone marrow stroma or do they include all cells capable of forming mesenchymal tissue? In the absence of a specific cell marker, MSCs may well incorporate a number of different cell populations all potentially variable in their phenotypic and growth characteristics, with mesenchymal differentiation as a common denominator.

Differentiation Potential

MSCs are usually grown as a monolayer culture in medium typically containing 10% fetal calf serum at 37°C in a humid environment containing 5% CO 2 . As for many other adult stem cells, MSCs are traditionally considered to only be capable of differentiating into cell types of their own original lineage, i.e., mesenchymal derivatives. We and many other groups have shown MSCs to be capable of forming osteoblasts, chrondrocytes and adipocytes both in vitro[34] and in vivo .[35] The ability of clonally expanded cells to form these three distinct cell types remains the only reliable functional criterion available to identify the genuine MSC and distinguish it from preosteoblast, preadipocyte or prechondrocytic cells which each only give rise to one cell type.[36]

Osteogenic potential

Osteogenic differentiation of MSCs is induced in vitro by treating a monolayer culture with a pro-osteogenic cocktail. Standardly used differentiation medium consists of dexamethasone, ascorbic acid-2-phosphate and beta-glycerophosphate.[37] Mineralized deposits can appear after a week, but the treatment is often maintained for up to three weeks in order to maximize the number and size of mineralizing nodules. Additional calcium is also used to increase in vitro mineralization.[10] Early signs of osteogenesis include an increase in bone-specific alkaline phosphatase activity, which can be measured enzymatically.[38] At the end of the treatment, cells can be fixed and stained with either Alizarin-Red S solution[10] [Figure - 1]C or silver nitrate for von Kossa staining,[37] which highlight the calcium phosphate deposits. Quantitative measurements of mineral deposition can then be obtained by colorimetry.[10] Furthermore, osteogenic differentiation is accompanied by the expression of genes such as osterix, cbfa1, osteopontin, osteocalcin, bone sialoprotein,[39],[40] which can be monitored at the RNA and protein level.

Chondrogenic potential

Chondrogenic differentiation is traditionally achieved by first forcing aggregation of 200,000 to 300,000 MSCs in chondrogenic medium, in order to generate a micromass pellet culture. Common to most culture protocols is the presence of dexamethasone, ascorbic acid phosphate and ITS+ supplement, which consists of bovine insulin, transferrin, selenous acid, linoleic acid and bovine serum albumin.[10] Further additives to the medium include sodium pyruvate, proline, L-glutamine and TGF-beta1,[37],[41] a growth factor involved in chondrogenesis in vivo .[42] Over the period of treatment, there is an upregulation of chondrogenic markers such as collagen II, collagen XI, aggrecan, perlecan and syndecan.[19],[43] After two to three weeks in culture, micromass pellets can then be fixed and embedded for sectioning and subsequent staining with Safranin-O, Toluidine blue or Alcian blue to highlight acid mucopolysaccharides, glycosaminoglycans and proteoglycans respectively.[10],[37],[41]

Adipogenic potential

Formation of mature adipocytes occurs following treatment of MSCs with a medium supplemented with dexamethasone, isobutylmethylxanthine, insulin and a PPARgamma agonist such as BRL 49653[10] with twice weekly medium changes for three weeks. Alternatively, MSCs can be exposed to three cycles of a treatment which alternates three days of culture in induction medium followed by two days in maintenance medium.[37] In this instance, the induction medium contains indomethacin instead of BRL 49653 and the maintenance medium contains insulin. The appearance of adipocytes containing lipid-filled droplets can be demonstrated by staining with oil red O [Figure - 1]D and by RT-PCR detection of adipsin, aP2 and PPARgamma expression.[3],[44] Quantitative data can be obtained by enzymatic dosage of the Glycerol-3-phosphate dehydrogenase, a marker of the mature adipocyte.[45] Furthermore, an elegant technique developed by Gimble allows the quantitation of adipocytes by flow cytometry in a MSC culture stained with the lipophilic dye nile red.[46]

Myogenic potential

In recent years, the extent of MSC multipotency has been frequently discussed, with many authors suggesting that MSCs have a differentiation potential broader than initially thought. When treated with the demethylating agent 5-azacytidine, MSCs have been demonstrated to have the ability to form muscle cells.[47] Myogenic potential has also been observed in a xenogeneic in vitro model where human MSCs are co-cultured with murine skeletal myocytes.[48]

Furthermore, MSCs have been shown to form cardiomyocytes,[49] which is being pursued as a potential replacement for the scar tissue formed as a result of myocardial infarction. Murine MSCs were reported to start beating spontaneously two weeks after 5-azacytidine treatment as well as assuming a cardiomyocyte-like morphology. The cells displayed both sinus node-like and ventricular cell-like action potentials and the beating rate was increased by isoproterenol treatment and blocked by a beta1-selective blocker, thus exhibiting signs of physiological cardiomyogenic response. This differentiation was supported by expression of alpha1, alpha1B, alpha1D, beta1 and beta2 adrenergic and M1 and M2 muscarinic receptors.[49] In all cases of MSCs displaying myogenic potential in vivo ,[48],[50] it is not clear how much of this ability is due to de novo myogenesis and how much is due to cell fusion events with resident myoblasts.[51] Whatever the mechanism may be, MSCs appear to provide a positive contribution to areas of muscular or cardiac damage, with no observed detrimental side-effects.[14],[52]

Neural potential?

Perhaps most interesting are the recent reports that under certain conditions MSCs can be made to form neural cells. Most studies showing the formation of both neuronal and glial cells from MSCs have been carried out in vitro ,[53],[54] complemented in a few instances by in vivo data,[55],[56] although in many cases the possibility of cell fusion cannot be formally ruled out. There have been a number of different approaches reported to trigger this apparent transdifferentiation in vitro . Some groups have used chemical treatments such as DMSO,[23],[57] whilst others have opted for the use of growth factors such as fibroblast growth factor (FGF), neuronal growth factor (NGF) and epidermal growth factor (EGF).[53],[54],[58],[59] However, recent evidence suggests that early positive results obtained using DMSO-based protocols are unreliable, as the neural-like morphology and gene expression displayed by MSCs after treatment were in fact due to toxicity.[60],[61] Despite this setback, many other groups have reported the production of MSC-derived neurons, astrocytes,[62] oligodendrocytes[53] and Schwann cells[63] in vitro with varying degrees of efficiency. There is, however, an intense ongoing debate about the nature of these differentiation responses. Are they merely an artefact of the in vitro process or do they represent genuine neural potential from MSCs? As mentioned earlier, the multipotency of the MSC seems to be a fluid and evolving concept as novel culture conditions and differentiation treatments are being developed.

Evaluating MSC Potential for Therapeutic Applications

An ideal source of adult stem cells?

The availability of autologous MSCs, which are easily accessible from patients, makes them a promising source of cells for many clinical applications in the evolving field of regenerative medicine. As well as providing the scaffolding (stromal) fraction of the bone marrow for HSCs to proliferate on, MSCs are thought to play a role in hematopoiesis itself.[4] MSCs have been shown to significantly improve hematopoietic recovery in patients receiving high-dose chemotherapy when compared with autologous blood stem cell transfusion alone.[64] Koc et al[64] co-infused culture-expanded MSCs with autologous blood stem cells in breast cancer patients and observed accelerated hematopoietic recovery.

Furthermore, MSCs represent an advantageous cell type for allogenic transplantation as evidence suggests that MSCs are immune-privileged, with low MHC I and no MHC II expression,[65] therefore reducing risks of rejection and complications for transplantation. In utero transplantation of human MSCs into sheep have shown that grafted cells could integrate into a variety of host tissues without any specific immune response.[66] MSCs have also been found to be immunosuppressive, through a mechanism thought to involve paracrine inhibition of T- and B-cell proliferation[67] (reviewed in[65]) and as such have been used in trials investigating their effects on autoimmune diseases and GVHD.[68],[69] Co-infusion of donor-derived MSCs together with HSCs has been shown to reduce the incidence and severity of GVHD in sibling allografts.[70] It was recently reported that a nine-year-old patient suffering from progressive severe GVHD that was unresponsive to classical therapy was treated with a MSC intravenous transplant from his mother and demonstrated a complete recovery.[68] The hypo-immunogenic properties of MSCs are considered by some to be sufficient to allow transplantation even between individuals who are not HLA-compatible.[71]

Applications for osteochondral repair

MSCs represent one of the few adult stem cell populations already in use in the clinic. MSCs are currently used to develop new therapies for a number of skeletal conditions. The osteogenic potential of MSCs has been utilized to treat cases of defective fracture healing, both alone and in combination with scaffolds to repair large bone defects with a high degree of success.[72] MSCs have also been used for cartilage repair. Autologous MSCs were expanded ex vivo , embedded in a collagen gel and re-implanted into areas of articular cartilage defect in osteoarthritis patients.[73] In this study, formation of hyaline cartilage-like tissue was improved in the experimental group compared to control. Although most applications for tissue repair involve local transplantation of MSCs to directly target the area of injury, systemic transplantation of MSCs has been in place for a long time in hematopoietic stem cell transplants. Recently, children suffering from osteogenesis imperfecta were treated systemically with allogenic MSCs. Transplanted MSCs were shown to migrate to the bone and produce collagen, thus providing a new and efficient route to alleviate the debilitating consequences of this genetic condition.[74]

Potential applications of MSCs for myocardiac repair

Current clinical trials are investigating the potential of MSCs for the treatment of myocardial infarction.[75] As previously discussed, a number of groups have reported MSC differentiation into cardiomyocytes in vitro . The current in vivo approach consists of injecting undifferentiated MSCs or whole bone marrow directly into the heart and, although the underlying mechanisms remains to be elucidated, significant improvement has been detected.[76],[77] The report by Chen et al[78] demonstrates a significant and sustained improvement in global left-ventricular ejection fraction, suggesting that MSC infusion triggers the formation of new cardiomyocytes and neoangiogenesis in the human heart.[79] It is still unclear whether MSCs act directly by in situ differentiation or fusion with resident myocytes[48] or indirectly through secretion of pro-myogenic factors promoting endogenous myocardial repair, such as VEGF and FGF.[80]

Therapeutic potential beyond mesenchymal lineages

The ability of MSCs to migrate to the site of injury has also been reported following transplantation in the brain. MSCs transplanted into rat striata were seen to migrate across the corpus callosum and populate the striatum, thalamic nuclei and substantia nigra of the 6-OHDA-lesioned hemisphere.[81] Untreated MSCs systemically infused into animals with damaged brain tissue have also been seen to migrate to the trauma site and improve recovery, although whether this is via secretion of neuroprotective factors or by differentiation into neural tissue remains unclear. Whilst it is not disputed that the MSCs appear to serve a positive role in recovery, there is debate as to whether the signs of differentiation observed in situ are genuine or merely a result of cell fusion with resident neural cells. More research is required to determine the reality and precise extent of MSC contribution in brain repair models as such results could have implications for neurodegenerative diseases such as Parkinson′s disease, Alzheimer′s disease or traumatic events such as stroke or spinal cord injury. Although the adult brain contains populations of neural stem cells[82],[83] these are insufficient to replace the massive number of cells needed to treat these conditions. The critical question has been to identify a source of neural progenitor cells for transplantation. The potential use of fetal tissue or differentiated embryonic stem cells suffers limitations due to tissue availability, ethical issues or safety concerns. The identification of an adult population of cells such as MSCs, which can be easily obtained from autologous or donated marrow and can be cultured and manipulated ex vivo, would represent a significant breakthrough in the search for many applications in regenerative medicine.

Conclusion

Mesenchymal stem cells represent a population of cells with the potential to contribute to future treatments for a wide range of acute or degenerative diseases. Significant progress has been made to identify the pharmacological and molecular pathways driving MSC differentiation towards mesenchymal derivatives in vitro and preliminary results indicate that MSCs could be used to generate neural derivatives. Much remains to be done in order to evaluate the physiological relevance of these early observations and to unravel the molecular mechanisms governing their differentiation in vivo . Applications currently under investigation for MSC-based therapies include musculoskeletal and cardiac repair, as well as genetic manipulation of MSCs for gene therapy strategies. Directed differentiation of autologous MSCs towards extra-mesenchymal lineages is an exciting and promising area of stem cell biology, with potential for the repair of tissues where resident stem cells are not accessible, such as the brain. MSCs thus represent an interesting and versatile population of adult stem cells demanding further molecular characterization and functional investigation. Future research will define the extent of their potential as an autologous and allogenic stem cell source for clinical application.

Acknowledgments

We are grateful to Julie Swales and Dr. Adrian Robins for their expert advice on flow cytometry. LJ is funded by the University of Nottingham. VS is grateful to the Anne McLaren Fellowship scheme and the Alzheimer′s Society for their support.

References

1.Friedenstein AJ, Piatetzky S 2nd, Petrakova KV. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol 1966;16:381-90.  Back to cited text no. 1    
2.Friedenstein AJ, Chailakhyan RK, Gerasimov UV. Bone marrow osteogenic stem cells: In vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet 1987;20:263-72.  Back to cited text no. 2  [PUBMED]  
3.Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD , et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143-7.  Back to cited text no. 3  [PUBMED]  [FULLTEXT]
4.Dazzi F, Ramasamy R, Glennie S, Jones SP, Roberts I. The role of mesenchymal stem cells in haemopoiesis. Blood Rev 2006;20:161-71.  Back to cited text no. 4  [PUBMED]  [FULLTEXT]
5.Sakaguchi Y, Sekiya I, Yagishita K, Muneta T. Comparison of human stem cells derived from various mesenchymal tissues: Superiority of synovium as a cell source. Arthritis Rheum 2005;52:2521-9.  Back to cited text no. 5  [PUBMED]  [FULLTEXT]
6.Risbud MV, Shapiro IM, Guttapalli A, Di Martino A, Danielson KG, Beiner JM , et al. Osteogenic potential of adult human stem cells of the lumbar vertebral body and the iliac crest. Spine 2006;31: 83-9.  Back to cited text no. 6  [PUBMED]  [FULLTEXT]
7.Zvaifler NJ, Marinova-Mutafchieva L, Adams G, Edwards CJ, Moss J, Burger JA , et al. Mesenchymal precursor cells in the blood of normal individuals. Arthritis Res 2000;2:477-88.  Back to cited text no. 7  [PUBMED]  [FULLTEXT]
8.Zuk PA, Zhu M, Mizuno H, Huang J, Futrell JW, Katz AJ , et al. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Eng 2001;7:211-28.  Back to cited text no. 8  [PUBMED]  [FULLTEXT]
9.Chunmeng S, Tianmin C. Effects of plastic-adherent dermal multipotent cells on peripheral blood leukocytes and CFU-GM in rats. Transplant Proc 2004;36:1578-81.  Back to cited text no. 9  [PUBMED]  [FULLTEXT]
10.Sottile V, Halleux C, Bassilana F, Keller H, Seuwen K. Stem cell characteristics of human trabecular bone-derived cells. Bone 2002;30:699-704.  Back to cited text no. 10  [PUBMED]  [FULLTEXT]
11.Campagnoli C, Roberts IA, Kumar S, Bennett PR, Bellantuono I, Fisk NM. Identification of mesenchymal stem/progenitor cells in human first-trimester fetal blood, liver and bone marrow. Blood 2001;98:2396-402.  Back to cited text no. 11  [PUBMED]  [FULLTEXT]
12.in 't Anker PS, Noort WA, Scherjon SA, Kleijburg-van der Keur C, Kruisselbrink AB, van Bezooijen RL , et al. Mesenchymal stem cells in human second-trimester bone marrow, liver, lung and spleen exhibit a similar immunophenotype but a heterogeneous multi line age differentiation potential. Haematologica 2003;88:845-52.  Back to cited text no. 12  [PUBMED]  [FULLTEXT]
13.Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 2000;109:235-42.  Back to cited text no. 13  [PUBMED]  [FULLTEXT]
14.Igura K, Zhang X, Takahashi K, Mitsuru A, Yamaguchi S, Takashi TA. Isolation and characterization of mesenchymal progenitor cells from chorionic villi of human placenta. Cytotherapy 2004;6: 543-53.  Back to cited text no. 14  [PUBMED]  
15.Pieternella S, Anker SA, Kleijburg-van der Keur C, Noort WA, Claas FH, Willemze R, et al . Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 2003;102:1548-9.  Back to cited text no. 15    
16.Santa Maria L, Rojas CV, Minguell JJ. Signals from damaged but not undamaged skeletal muscle induce myogenic differentiation of rat bone-marrow-derived mesenchymal stem cells. Exp Cell Res 2004;300:418-26.  Back to cited text no. 16  [PUBMED]  [FULLTEXT]
17.Baddoo M, Hill K, Wilkinson R, Gaupp D, Hughes C, Kopen GC , et al. Characterization of mesenchymal stem cells isolated from murine bone marrow by negative selection. J Cell Biochem 2003;89: 1235-49.  Back to cited text no. 17  [PUBMED]  [FULLTEXT]
18.Ringe J, Haupl T, Sittinger M. Mesenchymal stem cells for tissue engineering of bone and cartilage. Med Klin (Munich) 2003;98: 35-40.  Back to cited text no. 18  [PUBMED]  
19.Bosnakovski D, Mizuno M, Kim G, Takagi S, Okumura M, Fujinaga T. Isolation and multi line age differentiation of bovine bone marrow mesenchymal stem cells. Cell Tissue Res 2005;319:243-53.  Back to cited text no. 19  [PUBMED]  [FULLTEXT]
20.Moscoso I, Centeno A, Lopez E, Rodriguez-Barbosa JI, Santamarina I, Filgueira P , et al. Differentiation " in vitro" of primary and immortalized porcine mesenchymal stem cells into cardiomyocytes for cell transplantation. Transplant Proc 2005;37:481-2.  Back to cited text no. 20  [PUBMED]  [FULLTEXT]
21.Silva GV, Litovsky S, Assad JA, Sousa AL, Martin BJ, Vela D , et al. Mesenchymal stem cells differentiate into an endothelial phenotype, enhance vascular density and improve heart function in a canine chronic ischemia model. Circulation 2005;111: 150-6.  Back to cited text no. 21  [PUBMED]  [FULLTEXT]
22.Rhodes NP, Srivastava JK, Smith RF, Longinotti C. Heterogeneity in proliferative potential of ovine mesenchymal stem cell colonies. J Mater Sci Mater Med 2004;15:397-402.  Back to cited text no. 22  [PUBMED]  [FULLTEXT]
23.Devine SM, Bartholomew AM, Mahmud N, Nelson M, Patil S, Hardy W , et al. Mesenchymal stem cells are capable of homing to the bone marrow of non-human primates following systemic infusion. Exp Hematol 2001;29:244-55.  Back to cited text no. 23  [PUBMED]  [FULLTEXT]
24.Luria EA, Panasyuk AF, Friedenstein AY. Fibroblast colony formation from monolayer cultures of blood cells. Transfusion 1971;11:345-9.  Back to cited text no. 24  [PUBMED]  
25.Mareschi K, Ferrero I, Rustichelli D, Aschero S, Gammaitoni L, Aglietta M, et al . Expansion of mesenchymal stem cells isolated from pediatric and adult donor bone marrow. J Cell Biochem 2006;97:744-54.  Back to cited text no. 25  [PUBMED]  [FULLTEXT]
26.Bernardo ME, Avanzini MA, Perotti C, Cometa AM, Moretta A, Lenta E, et al . Optimization of in vitro expansion of human multipotent mesenchymal stromal cells for cell-therapy approaches: Further insights in the search for a fetal calf serum substitute. J Cell Physiol 2006.  Back to cited text no. 26    
27.Rubio D, Garcia-Castro J, Martin MC, de la Fuente R, Cigudosa JC, Lloyd AC, et al . Spontaneous human adult stem cell transformation. Cancer Res 2005;65:3035-9.  Back to cited text no. 27  [PUBMED]  [FULLTEXT]
28.Bruder SP, Ricalton NS, Boynton RE, Connolly TJ, Jaiswal N, Zaia J , et al. Mesenchymal stem cell surface antigen SB-10 corresponds to activated leukocyte cell adhesion molecule and is involved in osteogenic differentiation. J Bone Miner Res 1998;13:655-63.  Back to cited text no. 28  [PUBMED]  
29.Gronthos S, Graves SE, Ohta S and Simmons PJ. The STRO-1+ fraction of adult human bone marrow contains the osteogenic precursors. Blood 1994;84:4164-73.  Back to cited text no. 29    
30.Vogel W, Grunebach F, Messam CA, Kanz L, Brugger W, Buhring HJ. Heterogeneity among human bone marrow-derived mesenchymal stem cells and neural progenitor cells. Haematologica 2003;88:126-33.  Back to cited text no. 30  [PUBMED]  [FULLTEXT]
31.Mitchell JB, McIntosh K, Zvonic S, Garrett S, Floyd ZE, Kloster A , et al. Immunophenotype of human adipose-derived cells: Temporal changes in stromal-associated and stem cell-associated markers. Stem Cells 2006;24:376-85.  Back to cited text no. 31  [PUBMED]  [FULLTEXT]
32.Covas DT, Siufi JL, Silva AR, Orellana MD. Isolation and culture of umbilical vein mesenchymal stem cells. Braz J Med Biol Res 2003;36:1179-83.  Back to cited text no. 32  [PUBMED]  [FULLTEXT]
33.Colter DC, Class R, DiGirolamo CM, Prockop DJ. Rapid expansion of recycling stem cells in cultures of plastic-adherent cells from human bone marrow. Proc Natl Acad Sci USA 2000;97:3213-8.  Back to cited text no. 33  [PUBMED]  [FULLTEXT]
34.Muraglia A, Cancedda R, Quarto R. Clonal mesenchymal progenitors from human bone marrow differentiate in vitro according to a hierarchical model. J Cell Sci 2000;113:1161-6.  Back to cited text no. 34  [PUBMED]  [FULLTEXT]
35.Aslan H, Zilberman Y, Kandel L, Liebergall M, Oskouian RJ, Gazit D , et al. Osteogenic differentiation of non-cultured immuno isolated bone marrow-derived CD105+ cells. Stem Cells 2006;24:1728-37.  Back to cited text no. 35  [PUBMED]  [FULLTEXT]
36.Halleux C, Sottile V, Gasser JA, Seuwen K. Multi-lineage potential of human mesenchymal stem cells following clonal expansion. J Musculoskelet Neuronal Interact 2001;2:71-6.  Back to cited text no. 36  [PUBMED]  
37.Okamoto T, Aoyama T, Nakayama T, Nakamata T, Hosaka T, Nishijo K , et al. Clonal heterogeneity in differentiation potential of immortalized human mesenchymal stem cells. Biochem Biophys Res Commun 2002;295:354-61.  Back to cited text no. 37  [PUBMED]  [FULLTEXT]
38.Yoshimura H, Muneta T, Nimura A, Yokoyama A, Koga H, Sekiya I. Comparison of rat mesenchymal stem cells derived from bone marrow, synovium, periosteum, adipose tissue and muscle. Cell Tissue Res 2006; [Epub ahead of print].  Back to cited text no. 38    
39.Lin Y, Liu L, Li Z, Qiao J, Wu L, Tang W , et al. Pluripotency potential of human adipose-derived stem cells marked with exogenous green fluorescent protein. Mol Cell Biochem 2006;291:1-10.  Back to cited text no. 39  [PUBMED]  [FULLTEXT]
40.Friedman MS, Long MW, Hankenson KD. Osteogenic differentiation of human mesenchymal stem cells is regulated by bone morphogenetic protein-6. J Cell Biochem 2006;98:538-54.  Back to cited text no. 40  [PUBMED]  [FULLTEXT]
41.Suva D, Garavaglia G, Menetrey J, Chapuis B, Hoffmeyer P, Bernheim L , et al. Non-hematopoietic human bone marrow contains long-lasting, pluripotential mesenchymal stem cells. J Cell Physiol 2004;198:110-8.  Back to cited text no. 41  [PUBMED]  [FULLTEXT]
42.Emans PJ, Spaapen F, Surtel DA, Reilly KM, Cremers A, van Rhijn LW , et al. A novel in vivo model to study endochondral bone formation: HIF-1alpha activation and BMP expression. Bone 2007;40:409-18.  Back to cited text no. 42  [PUBMED]  [FULLTEXT]
43.Derfoul A, Perkins GL, Hall DJ, Tuan RS. Glucocorticoids promote chondrogenic differentiation of adult human mesenchymal stem cells by enhancing expression of cartilage extracellular matrix genes. Stem Cells 2006;24:1487-95.  Back to cited text no. 43  [PUBMED]  [FULLTEXT]
44.Fink T, Abildtrup L, Fogd K, Abdallah BM, Kassem M, Ebbesen P , et al. Induction of adipocyte-like phenotype in human mesenchymal stem cells by hypoxia. Stem Cells 2004;22:1346-55.  Back to cited text no. 44  [PUBMED]  [FULLTEXT]
45.Neubauer M, Hacker M, Bauer-Kreisel P, Weiser B, Fischbach C, Schulz MB , et al. Adipose tissue engineering based on mesenchymal stem cells and basic fibroblast growth factor in vitro . Tissue Eng 2005;11:1840-51.  Back to cited text no. 45  [PUBMED]  [FULLTEXT]
46.Gimble JM, Morgan C, Kelly K, Wu X, Dandapani V, Wang CS , et al. Bone morphogenetic proteins inhibit adipocyte differentiation by bone marrow stromal cells. J Cell Biochem 1995;58:393-402.  Back to cited text no. 46  [PUBMED]  
47.Taylor SM, Jones PA. Changes in phenotypic expression in embryonic and adult cells treated with 5-azacytidine. J Cell Physiol 1982;111:187-94.  Back to cited text no. 47  [PUBMED]  
48.Lee JH, Kosinski PA, Kemp DM. Contribution of human bone marrow stem cells to individual skeletal myotubes followed by myogenic gene activation. Exp Cell Res 2005;307:174-82.  Back to cited text no. 48  [PUBMED]  [FULLTEXT]
49.Makino S, Fukuda K, Miyoshi S, Konishi F, Kodama H, Pan J , et al. Cardiomyocytes can be generated from marrow stromal cells in vitro. J Clin Invest 1999;103:697-705.  Back to cited text no. 49  [PUBMED]  [FULLTEXT]
50.Di Rocco G, Iachininoto MG, Tritarelli A, Straino S, Zacheo A, Germani A , et al. Myogenic potential of adipose-tissue-derived cells. J Cell Sci 2006;119:2945-52.  Back to cited text no. 50  [PUBMED]  [FULLTEXT]
51.Goncalves MA, de Vries AA, Holkers M, van de Watering MJ, van der Velde I, van Nierop GP , et al. Human mesenchymal stem cells ectopically expressing full-length dystrophin can complement Duchenne muscular dystrophy myotubes by cell fusion. Hum Mol Genet 2006;15:213-21.  Back to cited text no. 51  [PUBMED]  [FULLTEXT]
52.Bartunek J, Croissant JD, Wijns W, Gofflot S, de Lavareille A, Vanderheyden M , et al. Pre treatment of adult bone marrow mesenchymal stem cells with cardiomyogenic growth factors and repair of the chronically infarcted myocardium. Am J Physiol Heart Circ Physiol 2006; [Epub ahead of print].  Back to cited text no. 52    
53.Hermann A, Gastl R, Liebau S, Popa MO, Fiedler J, Boehm BO , et al. Efficient generation of neural stem cell-like cells from adult human bone marrow stromal cells. J Cell Sci 2004;117:4411-22.  Back to cited text no. 53  [PUBMED]  [FULLTEXT]
54.Alexanian AR. Neural stem cells induce bone-marrow-derived mesenchymal stem cells to generate neural stem-like cells via juxtacrine and paracrine interactions. Exp Cell Res 2005;310: 383-91.  Back to cited text no. 54  [PUBMED]  [FULLTEXT]
55.Arnhold S, Klein H, Klinz FJ, Absenger Y, Schmidt A, Schinkothe T , et al. Human bone marrow stroma cells display certain neural characteristics and integrate in the subventricular compartment after injection into the liquor system. Eur J Cell Biol 2006;85: 551-65.  Back to cited text no. 55  [PUBMED]  [FULLTEXT]
56.Lu J, Moochhala S, Moore XL, Ng KC, Tan MH, Lee LK , et al. Adult bone marrow cells differentiate into neural phenotypes and improve functional recovery in rats following traumatic brain injury. Neurosci Lett 2006;398:12-7.  Back to cited text no. 56  [PUBMED]  [FULLTEXT]
57.Suzuki H, Taguchi T, Tanaka H, Kataoka H, Li Z, Muramatsu K , et al. Neurospheres induced from bone marrow stromal cells are multipotent for differentiation into neuron, astrocyte and oligodendrocyte phenotypes. Biochem Biophys Res Commun 2004;322:918-22.  Back to cited text no. 57  [PUBMED]  [FULLTEXT]
58.Magaki T, Kurisu K, Okazaki T. Generation of bone marrow-derived neural cells in serum-free monolayer culture. Neurosci Lett 2005;384:282-7.  Back to cited text no. 58  [PUBMED]  [FULLTEXT]
59.Bossolasco P, Cova L, Calzarossa C, Rimoldi SG, Borsotti C, Deliliers GL, et al . Neuro-glial differentiation of human bone marrow stem cells in vitro. Exp Neurol 2005;193:312-25.  Back to cited text no. 59  [PUBMED]  [FULLTEXT]
60.Lu P, Blesch A, Tuszynski MH. Induction of bone marrow stromal cells to neurons: Differentiation, trans-differentiation or artifact? J Neurosci Res 2004;77:174-91.  Back to cited text no. 60  [PUBMED]  [FULLTEXT]
61.Neuhuber B, Gallo G, Howard L, Kostura L, Mackay A, Fischer I. Reevaluation of in vitro differentiation protocols for bone marrow stromal cells: Disruption of actin cytoskeleton induces rapid morphological changes and mimics neuronal phenotype. J Neurosci Res 2004;77:192-204.  Back to cited text no. 61  [PUBMED]  [FULLTEXT]
62.Wislet-Gendebien S, Wautier F, Leprince P, Rogister B. Astrocytic and neuronal fate of mesenchymal stem cells expressing nestin. Brain Res Bull 2005;68:95-102.  Back to cited text no. 62  [PUBMED]  [FULLTEXT]
63.Keilhoff G, Goihl A, Langnase K, Fansa H, Wolf G. Transdifferentiation of mesenchymal stem cells into Schwann cell-like myelinating cells. Eur J Cell Biol 2006;85:11-24.  Back to cited text no. 63  [PUBMED]  [FULLTEXT]
64.Koc ON, Gerson SL, Cooper BW, Dyhouse SM, Haynesworth SE, Caplan AI , et al. Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J Clin Oncol 2000;18:307-16.  Back to cited text no. 64  [PUBMED]  [FULLTEXT]
65.Uccelli A, Moretta L, Pistoia V. Immunoregulatory function of mesenchymal stem cells. Eur J Immunol 2006;36:2566-73.  Back to cited text no. 65  [PUBMED]  [FULLTEXT]
66.Liechty KW, MacKenzie TC, Shaaban AF, Radu A, Moseley AM, Deans R , et al. Human mesenchymal stem cells engraft and demonstrate site-specific differentiation after in utero transplantation in sheep. Nat Med 2000;6:1282-6.  Back to cited text no. 66  [PUBMED]  [FULLTEXT]
67.Di Nicola M, Carlo-Stella C, Magni M, Milanesi M, Longoni PD, Matteucci P, et al . Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002;99:3838-43.  Back to cited text no. 67  [PUBMED]  [FULLTEXT]
68.Le Blanc K, Rasmusson I, Sundberg B, Gotherstrom C, Hassan M, Uzunel M , et al. Treatment of severe acute graft-versus-host disease with third party haploidentical mesenchymal stem cells. Lancet 2004;363:1439-41.  Back to cited text no. 68  [PUBMED]  [FULLTEXT]
69.Fang B, Song YP, Liao LM, Han Q, Zhao RC. Treatment of severe therapy-resistant acute graft-versus-host disease with human adipose tissue-derived mesenchymal stem cells. Bone Marrow Transplant 2006;38:389-90.  Back to cited text no. 69  [PUBMED]  [FULLTEXT]
70.Lazarus HM, Koc ON, Devine SM, Curtin P, Maziarz RT, Holland HK , et al. Cotransplantation of HLA-identical sibling culture-expanded mesenchymal stem cells and hematopoietic stem cells in hematologic malignancy patients. Biol Blood Marrow Transplant 2005;11:389-98.  Back to cited text no. 70  [PUBMED]  [FULLTEXT]
71.Le Blanc K, Tammik C, Rosendahl K, Zetterberg E, Ringden O. HLA expression and immunologic properties of differentiated and undifferentiated mesenchymal stem cells. Exp Hematol 2003;31:890-6.  Back to cited text no. 71  [PUBMED]  [FULLTEXT]
72.Quarto R, Mastrogiacomo M, Cancedda R, Kutepov SM, Mukhachev V, Lavroukov A , et al. Repair of large bone defects with the use of autologous bone marrow stromal cells. N Engl J Med 2001;344: 385-6.  Back to cited text no. 72  [PUBMED]  
73.Wakitani S, Imoto K, Yamamoto T, Saito M, Murata N, Yoneda M. Human autologous culture expanded bone marrow mesenchymal cell transplantation for repair of cartilage defects in osteoarthritic knees. Osteoarthritis Cartilage 2002;10:199-206.  Back to cited text no. 73  [PUBMED]  [FULLTEXT]
74.Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M , et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med 1999;5:309-13.  Back to cited text no. 74  [PUBMED]  [FULLTEXT]
75.Stamm C, Westphal B, Kleine HD, Petzsch M, Kittner C, Klinge H , et al. Autologous bone-marrow stem-cell transplantation for myocardial regeneration. Lancet 2003;361:45-6.  Back to cited text no. 75  [PUBMED]  [FULLTEXT]
76.Wollert KC, Meyer GP, Lotz J, Ringes-Lichtenberg S, Lippolt P, Breidenbach C , et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: The BOOST randomized controlled clinical trial. Lancet 2004;364:141-8.  Back to cited text no. 76  [PUBMED]  [FULLTEXT]
77.Fuchs S, Satler LF, Kornowski R, Okubagzi P, Weisz G, Baffour R , et al. Catheter-based autologous bone marrow myocardial injection in no-option patients with advanced coronary artery disease: A feasibility study. J Am Coll Cardiol 2003;41:1721-4.  Back to cited text no. 77  [PUBMED]  [FULLTEXT]
78.Chen SL, Fang WW, Ye F, Liu YH, Qian J, Shan SJ , et al. Effect on left ventricular function of intracoronary transplantation of autologous bone marrow mesenchymal stem cell in patients with acute myocardial infarction. Am J Cardiol 2004;94:92-5.  Back to cited text no. 78  [PUBMED]  [FULLTEXT]
79.Nagaya N, Fujii T, Iwase T, Ohgushi H, Itoh T, Uematsu M , et al. Intravenous administration of mesenchymal stem cells improves cardiac function in rats with acute myocardial infarction through angiogenesis and myogenesis. Am J Physiol Heart Circ Physiol 2004;287:H2670-6.  Back to cited text no. 79  [PUBMED]  [FULLTEXT]
80.Xu M, Uemura R, Dai Y, Wang Y, Pasha Z, Ashraf M. In vitro and in vivo effects of bone marrow stem cells on cardiac structure and function. J Mol Cell Cardiol 2006; [Epub ahead of print].  Back to cited text no. 80    
81.Hellmann MA, Panet H, Barhum Y, Melamed E, Offen D. Increased survival and migration of engrafted mesenchymal bone marrow stem cells in 6-hydroxydopamine-lesioned rodents. Neurosci Lett 2006;395:124-8.  Back to cited text no. 81  [PUBMED]  [FULLTEXT]
82.Gage FH. Mammalian neural stem cells. Science 2000;287: 1433-8.  Back to cited text no. 82  [PUBMED]  [FULLTEXT]
83.Sottile V, Li M, Scotting PJ. Stem cell marker expression in the Bergmann glia population of the adult mouse brain. Brain Res 2006;1099:8-17.  Back to cited text no. 83  [PUBMED]  [FULLTEXT]

Copyright 2007 - Journal of Postgraduate Medicine


The following images related to this document are available:

Photo images

[jp07044f1.jpg]
Home Faq Resources Email Bioline
© Bioline International, 1989 - 2024, Site last up-dated on 01-Sep-2022.
Site created and maintained by the Reference Center on Environmental Information, CRIA, Brazil
System hosted by the Google Cloud Platform, GCP, Brazil