3,097
Views
144
CrossRef citations to date
0
Altmetric
SPECIAL SECTION ON STEM CELLS: REVIEW ARTICLE

Mesenchymal stem cells

Pages 469-479 | Published online: 08 Jul 2009

Abstract

About 40 years ago Friedenstein described stromal cells in the bone marrow that were spindle shaped and proliferate to form colonies. These cells attach to plastic and are able to differentiate under defined in vitro conditions into multiple cell types present in many different tissues, e.g. osteoblasts, chondroblasts, adipocytes, etc. Later on these cells, obtained from postnatal bone marrow, were called mesenchymal stem cells (MSC) or stromal stem cells. Recently the presence of somewhat similar cells has been demonstrated in many other tissues too. In spite of extensive attempts to characterize these cells we are still lacking definitive in vivo markers of MSC although retrospective functional data strongly support the existence of common adult stem cells that have the capacity to differentiate along various specific differentiation lineages. Since MSC can be rather easily isolated from the bone marrow and can also be expanded in vitro they have become a prime target for researchers of tissue regeneration. These cells have now been extensively used for transplantation experiments in animals and also for some therapeutic trials in humans. However, much new research is needed to learn enough on the molecular mechanisms of MSC differentiation to evaluate their full capacity for tissue regeneration.

Introduction

Most mesenchymal tissues are remodelled throughout life. Complete regeneration of damaged tissue is an extreme example of remodelling. There are remarkable differences in the regeneration capacity between different species as well as different tissues in the same individual. Notwithstanding the tissue, the remodelling process in adults requires a continuous supply of new cells suggesting the existence of cells, either local or circulating, which have retained their capacity for proliferation and differentiation. It is of interest that the German pathologist Julius Cohnheim, back in 1867 Citation1 suggested that tissue repair in mammals was dependent on cells coming from the bloodstream. Cohnheim's hypothesis was either denied or forgotten for decades but now there are numerous in vivo and in vitro experiments demonstrating that the bone marrow (source of circulating cells), in addition to haematopoetic stem cells (HSC), contain a population (or populations) of stromal cells that under suitable conditions are able to differentiate towards a number of specific mesenchymal cell types including fibroblasts, osteoblasts, chondroblasts, adipocytes etc. Citation2–6. These cells are now widely called mesenchymal stem cells (MSC) and could be found even in the bone marrow of very old individuals although their number may decrease with age Citation7, Citation8.

Since the original demonstration of MSC in bone marrow, several other tissues have also been shown to contain cells that have several features in common with MSC in the bone marrow. Although we still have rather modest understanding of the details of their proliferation and differentiation, they have become an important tool of tissue regeneration studies and are presently considered as potential candidates for several clinical applications Citation9. Some clinical trials using MSC have already been initiated and encouraging results have been reported, for instance, in osteogenesis imperfecta Citation10, Citation11 and metachromatic leukodystrophy Citation12. MSC transplants have also been used to enhance engraftment of heterologous bone marrow transplants Citation13. In this review I will first discuss some issues concerning the biology of MSC and then evaluate their potential clinical value in regenerative medicine and in some other indications.

Key messages

  • Mesenchymal stem cells (MSC) are multipotent adult stem cells present in bone marrow and having capacity to differentiate towards osteoblasts, chondroblasts, adipocytes and some other cell types.

  • The potential plasticity and self‐renewal capacity of MSC offer a huge potential for clinical tissue regeneration.

  • Some clinical trials using MSC have given encouraging results.

Development of MSC concept

In a series of elegant papers Friedenstein and his colleagues working in Moscow during 1960s and 70s described clonal and plastic adherent stromal cells from bone marrow Citation2–6, Citation14–17. Early studies were able to demonstrate that these cells formed fibroblast colony forming units (F‐CFU) but also had the capacity to differentiate towards osteoblasts, chondroblasts and adipocytes under defined in vitro culture conditions. Later in vitro studies in several laboratories confirmed the plasticity and genuine multipotency of bone marrow stromal cells with cultures originated from cloned single cells. It was also evident from the early experiments with MSC that only minor changes in the culture medium were needed to change their differentiation from one lineage to another supporting further the idea that these cells represent truly a plastic cell population.

The plasticity and the importance of tissue microenvironment for MSC differentiation are also demonstrated in various in vivo animal experiments. Liechty et al. Citation18, for instance, injected human bone marrow derived MSC into the circulation of foetal sheep early in gestation. hMSC engrafted and persisted in multiple tissues for as long as 13 months after transplantation. In addition, transplanted human cells underwent site‐specific differentiation into chondrocytes, adipocytes, myocytes and cardiomyocytes, bone marrow stromal cells and thymic stroma in their respective tissues.

Beginning from Friedenstein's original experiments much effort has been focused on the direct identification of MSC from the bone marrow and more recently also from other tissue sources. Although there has been major progress we are, unfortunately, still lacking a defined panel of well characterized surface markers for MSC in order to identify them unambiguously in situ. Several surface markers have been studied and some of them, like STRO‐1, HOP‐26, CD49a and SB‐10/CD166 differentiate MSC from most of the other cell types Citation19. Thus the final definition of MSC is still based on retrospective functional experiments where certain populations of cells or cultures originated from a single stromal cell have been demonstrated to differentiate towards multiple tissue lineages.

Before unquestionable identification of MSC in situ could be done it would be very difficult to answer unambiguously the remaining open questions concerning nature, function and potential therapeutic value of MSC. Firstly, is there one and only one mesenchymal stem cell pool in an adult organism and is this pool a direct descendent of embryonal stem cells? Secondly, what is the hierarchy of MSC differentiation and are these cells able to change lineages even after a substantial differentiation along a specific lineage? Thirdly, how much does the cellular microenvironment determine the cell fate and to what extent, if any, the cell fate is already imprinted during the early development of an individual? All these and many other questions remain to be answered before the concept of MSC has been clarified completely.

Tissue distribution and isolation of MSC

In addition to bone marrow, MSC‐like cells have been shown to be present in a number of other adult and foetal tissues, including circulating blood Citation20, Citation21, cord blood Citation22, Citation23, placenta Citation24, amniotic fluid Citation25, heart Citation26, skeletal muscle Citation27, adipose tissue Citation28, synovial tissue Citation29 and pancreas Citation30. These are only some examples in order to demonstrate diversity of potential MSC‐containing tissues. Putting together the present data one may assume that basically all organs containing connective tissue also contain MSC.

The bone marrow is still by far the best characterized source of MSC and almost all that is known about their differentiation is based on studies with marrow‐derived MSC. Actually, in order be to critical enough, one should wait for more detailed comparison between the bone marrow and other tissue‐derived MSC before even categorizing them under the same name. This cautiousness is needed although there are already some comparative studies including several different tissues with a number of phenotypic criteria.

One of the major practical problems in MSC research is the relatively low number of self‐renewing cells in studied tissues. In the bone marrow the frequency of MSC among other cells, including haematopoietic stem cells and various differentiated stages of all other possible lineages, has been estimated to be about 10 MSC per one million of all bone marrow cells Citation31. Due to the lack of definitive markers for MSC it has not been possible to count exact densities of these cells among the marrow cell population.

Several different modifications from the original method of Friedenstein have been developed to enrich MSC from bone marrow cell suspension. In addition to plastic adherence, medium selection and single‐cell cloning, these include different sedimentation procedures and more recently various affinity‐based cell sorting methods. Also attempts to up‐scale the production of cells in vitro for transplantation purposes have been under extensive study Citation32. However, all the isolation methods used suffer from lack of absolute specificity as long as there is not a definitive phenotypic marker(s) for MSC. As mentioned earlier several tissues have been demonstrated to contain cells that are able to proliferate and differentiate into various skeletal cell phenotypes. In a recent study of Musina et al. Citation33 MSC were isolated from human bone marrow, adipose tissue, skin, placenta, and thymus and compared for the presence of surface markers (CD10, CD13, CD31, CD44, CD90, CD105) without observing any differences between different tissue sources. To compare the properties of human mesenchymal stem cells present in different tissues Sakaguchi et al. Citation34 isolated cells from bone marrow, synovium, periosteum, skeletal muscle, and adipose tissue and studied their colony forming capacity and differentiation under defined conditions. In studies of osteogenesis, the rate of alizarin red‐positive colonies was highest in bone marrow‐, synovium‐ and periosteum‐derived cells, respectively, whereas synovium‐derived cells had the greatest ability for chondrogenesis. In adipogenesis experiments, the frequency of Oil Red O‐positive colonies was highest in synovium‐ and adipose tissue‐derived cells. These and some other results suggest that MSC from different tissues may have differences in their differentiation capacity even if cultured in exactly the same microenvironment.

Some of these tissue sources besides bone marrow, like peripheral or cord blood, adult adipose tissue and skeletal muscle may turn out to be important sources of MSC for transplantation since they are relatively easy to obtain. Although several studies have indicated that many tissues contain cells that are able to differentiate towards osteogenic and some other phenotypes, it remains to be seen if these ‘MSC’ represent similar cell populations or if they are already further differentiated than marrow derived stromal cells. So many different tissues have now been shown to contain mesenchymal cells that are able to differentiate into different mesenchymal lineages that it may well be that all mesenchymal tissues retain their ‘stem cell capacity’ through most of the individual's life span. This would be promising news for medical tissue regeneration. It remains to be seen if these ‘peripheral MSC populations’ have any tissue‐specific properties that may favour tissue‐selective use of them.

Differentiation of MSC and their niche

Embryonal stem (ES) cells from inner cell mass of early embryo are considered to be pluripotent cells and are thus able to differentiate towards all different specific cell types of an adult organism. MSC are considered to be only multipotent cells with clearly more restricted capacity for differentiation than ES cells. Most likely this is the case, but since MSC are present in several different adult tissues and in vitro studies have demonstrated that they are able to differentiate into various cell types, it may well be that the plasticity of MSC has been underestimated Citation35. Some cellular markers that were thought to be expressed only in ES cells have also been shown to be expressed in MSC.

A proper microenviroment for cellular differentiation in vivo most likely involves both soluble factors as well as multiple cell‐cell and cell‐matrix contacts. So how the plasticity of MSC is then modulated and what are the extra‐ and intracellular signalling pathways that are operating when MSC undergo differentiation towards specific cell lineages? This remains a major challenge of adult stem cell research since it is important to define the correct microenviroment for each lineage in order to create, for instance, proper conditions for in vitro cell expansion. Detailed molecular dissections of the signalling pathways and of transcriptional regulation determining different cellular fates are needed. Both of these are now under extensive investigation and many details have already been solved although the whole picture of MSC differentiation is still far from clear.

Effects of several different hormones, vitamins, growth factors and cytokines on MSC proliferation and differentiation have been tested applying in vitro differentiation assay, studying mainly osteogenic Citation36–42, chondrogenic Citation43–45 or adipogenic differentiation Citation46, Citation47. Self‐renewal capacity is a fundamental feature of all stem cells. It would thus be important to identify growth factors that not only promote proliferation but also retain self‐renewal capacity of MSC and to maintain their multilineage potential. On the basis of the present data it is far too early to conclude which growth factors are essential to retain the self‐renewal capacity. However, several studies seem to indicate that members of fibroblast growth factor (FGF)‐family, especially FGF‐2, play a crucial role in this. Gene transfer of growth factors applying either viral or other types of carriers has been used to create microenvironment to support differentiation to a specific lineage. Enhanced differentiation towards osteoblasts has been obtained e.g. with over‐expression of bone morphogenetic proteins. Cartilage type of differentiation was greatly enhanced by over‐expression of transforming growth factor (TGF)β in the bone marrow‐derived MSC Citation48.

When MSC are plated on plastic at low density, the cultures display a lag phase of about 3 days before they enter a phase of rapid exponential growth. Gregory et al. Citation49 found that as the cultures leave the lag phase, they secrete high levels of dickkopf‐1 (Dkk‐1), an inhibitor of the canonical Wnt signalling pathway. Biological role of the phenomenon was then confirmed by addition of recombinant Dkk‐1 into cultures toward the end of the lag period and, indeed, it increased proliferation and simultaneously decreased the concentration of β‐catenin. If the antibodies to Dkk‐1 were increased during the early log phase it decreased cellular proliferation, and also serum starvation was associated with the decreased expression of Dkk‐1 in MSC. Synthetic peptides derived from Dkk‐1 were found to enhance the proliferation of human MSC in culture. In peptide overlay assays on human MSC lysates these peptides bound a 184‐kDa protein, most likely of low density lipoprotein receptor related protein (LRP)6 Citation50, Citation51. This observation opens an interesting possibility to develop Wnt antagonists to enhance MSC production both in vitro and perhaps also in vivo.

Perhaps the best characterized lineages presently are osteogenic and adipogenic differentiation. It is clear that osteoblasts and adipocytes originate from the same MSC through the activation of specific transcription programmes. Key transcription factors to direct MSC into osteogenic and adipogenic lineages are runt homology domain transcription factor (Runx)2 and peroxisome proliferators‐activated receptor γ (PPARγ), respectively. The activation of Runx2 and down‐regulation of PPARγ directs MSC into osteogenic lineage and reciprocal transcriptional event leads into adipogenic lineage Citation52. Recent study of Hong et al. Citation53 nicely demonstrates that lineage determination could be regulated by another transcription factor TAZ (transcription coactivator with PDZ‐binding motif ). Both of the above mentioned key transcription factors, Runx2 and PPARγ, are under the regulation of TAZ which co‐activates Runx2 dependent osteogenic gene expression and simultaneously represses PPARγ‐dependent adipogenic gene expression. In the future we will most likely see a number of these molecular rheostats that are important modulators of cellular fate and lineage differentiation. Identification of such molecules will further help to clarify signalling pathways that are needed to differentiate MSC into various specific lineages. This information is urgently needed since it could be used to create optimal microenvironment for tissue specific lineages during tissue regeneration for medical purposes.

It has been known for a long time that the number of MSC in the bone marrow declines dramatically with aging. Whether the remaining cells also lose their capacity to proliferate and differentiate and finally to repair tissue damage has not been studied well enough yet. In some studies these issues have been addressed and MSC from aging individuals seem to retain their capacity at least for bone formation Citation54 and tendon regeneration Citation55. However, recent microarray studies demonstrate differences in gene expression profiles between foetal and adult MSC providing one explanation why foetal MSC have higher proliferative capacity and are less lineage committed than adult MSC Citation56. Further comparative studies both at the functional as well as gene expression level are needed to find out how much aging as such affects the capacity of MSC; and especially to detect if some tissue microenvironments are better than others to protect the MSC population during aging. Baxter et al. Citation57 studied the effect of in vitro expansion on the rate of telomere loss and replicative capacity of MSC. They observed that even minimal expansion induced a rapid aging of MSC, with losses equivalent to about half of their total replicative lifespan. Liu et al. Citation58 isolated MSC from telomerase (‐/‐) mice and found that they failed to differentiate into adipocytes and chondrocytes, even at early passages, whereas wild‐type MSC under the same conditions were capable of differentiation. This suggests that telomerase activity of MSC is not required only for self‐replication but is also needed for differentiation.

Potential clinical applications of MSC

Since MSC are multipotent and their numbers can be expanded in culture, there has been much interest in their clinical potential for tissue repair and gene therapy. A number of studies have now also demonstrated the migration and multi‐organ engraftment potential of MSC in animal models and in various human organ transplantations. In addition, immunomodulatory properties of MSC and their supportive functions for haematopoietic stem cells have also opened up possibilities for interesting new avenues for their use in the treatment of autoimmune diseases and graft rejection, respectively. Although understanding of mechanisms behind MSC differentiation and cell fate determination is still incomplete and the molecular processes that drive, for instance, engraftment are complex, many clinical applications are now emerging for MSC transplantations. Some of these will be briefly discussed below. For more detailed reviews see other reviews in the present issue of Annals of Medicine.

Tissue regeneration

The potential plasticity and self‐renewal capacity of MSC offers a huge potential for clinical tissue regeneration. Although animal experiments using many different disease models have been successful and promising, the use of MSC for clinical indications is still in its infancy when compared to the common clinical use of HSC.

Several theories have been presented of how bone marrow‐derived mesenchymal stem cells could induce tissue regeneration. The first possibility is that they act like genuine mesenchymal stem cells and could replace stem cell population in the specific tissue and mimic normal growth when building up new connective tissue. The second possibility is that MSC or some of their descendents are able to transdifferentiate to specific cell types in a specific microenvironment and form not only mesenchymal tissues but also ectodermal and/or endodermal components. There is also a third possibility, which could explain tissue regeneration in some specific occasions, namely formation of heterokaryons with MSC and tissue‐specific cells. In addition, it is also possible that transplanted MSC only produce paracrine factors that are able to stimulate tissue specific cells to proliferate or at least protect tissue from further damage giving better possibility for the tissue to recover. All these and perhaps also some other mechanisms should be kept in mind when MSC‐induced tissue regeneration is considered.

Up until now most of the tissue regeneration studies involving MSC (naïve or gene‐manipulated) have focused on development of new bone to heal large bone defects together with some supporting scaffolds Citation59–61. Obvious major limitations for the use MSC in the healing of site‐specific large bone defects are the need for the relatively high number of cells and identification of optimal supportive biomaterials to construct ‘a living implant’. Several novel approaches to improve conventional tissue culture conditions in order to get a higher number of cells and to prevent loss of their differentiation capacity have been applied. These include studies focusing on the role of serum source and serum treatment Citation62, Citation63 and use of osteogenic growth and differentiation factors, like bone morphogenetic proteins and fibroblast growth factors and some others Citation64–66. Over‐expression of telomerase in MSC leads also to enhanced proliferation Citation67. The advantage of creating a correct niche for proliferation and differentiation of MSC has stimulated the experimental use of various protein matrices to support lineage‐specific development Citation68. Increasing knowledge of the molecular mechanisms of stem cell differentiation may also give new pharmacological possibilities, such as intervention of mevalonate pathway by statins or mimics of Wnt‐signalling, and like inhibitors of glycogen synthetase kinase 3β, to enhance osteogenetic properties of MSC Citation69, Citation70. In spite of many promising novel strategies we are still lacking a satisfactory standard protocol for in vitro mesenchymal stem cell expansion and optimal biomaterials to prepare living implants for bone repair.

Perhaps the most exciting clinical studies concerning regeneration have been conducted in patients suffering from an inherited bone disease called osteogenesis imperfecta or brittle‐bone disease. These patients suffer multiple fractures already in utero. Direct transplantation of allogeneic MSC to a patient Citation11 as well as gene therapy using transfection of autologous MSC with a gene construct and application of these ‘corrected’ cells have been used with promising results Citation71.

In addition of bone, articular cartilage has been the target for intensive cellular therapy efforts, and local delivery of mesenchymal stem cells has been explored as a therapeutic approach in different animal models of osteoarthritis (for review see, for example, (72)). Since regeneration of articular cartilage is very poor, development of MSC‐based methods for regeneration of this tissue would be of enormous medical importance. In an elegant study of Ichinose et al. Citation73 cultured bone marrow cells in alginate beads with a chondrogenesis‐induction medium containing TGF‐β3. Proliferating cells first differentiated to typical matrix producing chondroblasts and gradually during the subsequent culture became hypertrophic with typical gene expression profile of hypertrophic chondrocytes. Interestingly, these cartilage nodules finally calcified and simultaneously started to express osteoblastic marker genes mimicking endochondral ossification. It remains to be seen if the authors were able to create conditions for transdifferentiation of chondrocytes to osteoblasts or if these cells differentiated from the remaining stem cells in the presence of correct microenvironment. These findings indicate that three‐dimensional culture could be a convenient in vitro model to study different steps of endochondral bone formation all the way from MSC via cartilage to bone.

Therapeutic applications of adult mesenchymal stem cells are approaching clinical use also in several fields other than bone and cartilage regeneration. Promising new tissue targets among others seem to be, for instance, tendons, ligaments, menisci, and other connective tissues Citation74, Citation75. Cartilage, bone and ligaments have also been tried to engineer using bioreactors. Human MSC, highly porous protein scaffolds and bioreactors with perfusion cartridges were used to produce specific conditions for lineage specific differentiation Citation76. In each case, the scaffold and bioreactor were designed to mimic some aspects of the environment present in native tissues. These and some other studies have demonstrated that also physical parameters, like loading and fluid flow, could be important to create a differentiation niche for MSC.

One of the most exciting potential applications for the clinical use of MSC is regeneration of the heart muscle after myocardial infarct. It has been shown that multipotent mesenchymal stem cells are present within the connective tissues of the heart and may thus be useful for local tissue regeneration if somehow inducible Citation77. However, more interest is now focused on either systemic or local implantation of bone marrow‐derived allogeneic or autologous MSC into damaged myocardium Citation78. Experiments using several different animal models have thus clearly shown that engraftment of MSC induces at least transient progress of the healing process in the damaged myocardium. However, there is much controversy concerning the mechanism(s). First of all, it remains to be seen if bone marrow‐derived stem cells could differentiate autonomously into muscle cells or if tissue regeneration is reached via cellular fusion. Second, what is the exact origin of cardiomyocyte progenitors in the bone marrow? Are they a subpopulation of MSC or HSC and does actual transdifferentiation take place? Earlier studies suggested that HSC can transdifferentiate into cardiomyocytes, however, more recent studies have challenged the origin of myocardium‐engrafted cardiomyocytes being HSC Citation79. Thirdly, more recent studies have suggested that the protective effect of MSC in myocardial infarction is mainly due to their paracrine effects, meaning that transplanted cells secrete soluble factors that are able to protect cells from tissue damage Citation80, Citation81. This effect does not need an actual replacement of cardiomyoblasts with MSC‐derived cells. In conclusion, several in vivo studies indicate that MSC transplantation improves cardiac function in animal models of cardiomyopathy and ischaemic damage. It remains to be seen if this is direct tissue regeneration via injected MSC or if it is mainly through induction of myogenesis and angiogenesis from local progenitors or mainly by inhibition of myocardial fibrosis by paracrinic factors produced by cardiac progenitors Citation82. It is interesting to note here that a recent study of Ma et al. Citation83 also suggested a paracrinic role for transplanted MSC in the healing of chemically damaged cornea in rats. In addition to the studies mentioned, MSC have been applied in a number of other experimental tissue regeneration studies. Thus there are good reasons to believe that at least some of these experimental models will produce new potential applications for further clinical practice.

MSC as targeting vehicles

Animal studies have shown the engraftment of MSC to different tissues and further suggested that it may be enhanced by local tissue damage and stress. This has opened up a possibility that in addition to direct tissue regeneration, MSC could also be used as targeting vehicles in gene therapy with various therapeutic molecules. At present there are experimental data and animal models to test MSC‐based gene therapy for both malignant and non‐malignant indications.

It has been shown that intra‐tumour injection of MSC into malignant gliomas caused significant inhibition of tumour growth and increased the survival of glioma‐bearing rats. Gene‐modification of MSC by infection with an adenoviral vector encoding human interleukin‐2 (IL‐2) clearly enhanced the anti‐tumour effect and prolonged the survival of tumour‐bearing animals Citation84. Injection of MSC over‐expressing interferon β suppressed the growth of pulmonary metastases, presumably through the local production of interferon β in the tumour microenvironment Citation85. These and other related studies demonstrate that gene therapy employing MSC as a targeting vehicle would be a promising therapeutic approach for some treatment‐resistant tumours. In addition, they may also have a direct capacity to restrict malignant growth.

Transfection of MSC with potentially therapeutic genes has been tried also in many other disease models. They have turned out to be potentially useful gene delivery vehicles of gene therapy e.g. for Parkinson's disease Citation86. Genetically modified mesenchymal stem cells have also been engrafted into the heart to develop a biological pacemaker Citation87. Kurozumi et al. Citation88 transfected telomerized human MSC with the brain‐derived neurotrophic factor (BDNF) gene using a fibre‐mutant adenovirus vector and reported that such treatment improved recovery in a rat transient middle cerebral artery occlusion model. In conclusion, several studies have demonstrated that MSC could be transfected with therapeutic molecules and these gene‐modified cells retain their capacity for engraftment into damaged tissues or tumours.

Immunomodulatory role of MSC

A number of different studies have demonstrated that MSC avoid allogeneic rejection in humans and in different animal models Citation89, Citation90. This opens up a much broader use of MSC for tissue transplantation purposes than use of autologous cell sources only. Reasons for this are not totally clear but it has been shown that MSC often lack major histocompatibility complex (MHC)‐II and co‐stimulatory molecules Citation91. Mechanisms may well be similar to that behind maternal tolerance of the foetal allograft. MSC have been shown to have a direct immunosuppressive effect on T‐cell proliferation in vitro. It has also been shown that MSC induced apoptosis in activated T‐cells but had no effect on resting T‐cells Citation92. In addition, MSC may also have some capacity to modulate the differentiation, maturation and function of dendritic cells Citation93. This may be partially due to the fact that MSC decreased their capacity to produce cytokines, for instance interleukin (IL)‐12 Citation94. These immunosuppressive properties of MSC have also been tested in various in vivo models and clinical situations. Co‐injection of MSC has clearly prolonged the rejection of mismatched skin grafts in animals Citation95 and it has been shown that MSC were homing to several damaged tissues after radiation‐induced multi‐organ failure Citation96. Treatment of severe acute graft‐versus‐host diseases have been tried with transplantation of MSC with striking results suggesting that immunosuppressive properties of MSC may be of interest when developing new strategies to treat these complicated and difficult conditions Citation97. Some of the recent experimental Citation98 and clinical reports Citation99 are also encouraging and suggest that transplantation of MSC may have a role even in the treatment of autoimmune diseases. However, immunosuppressive effects may also have unwanted consequences since it has been shown, for instance that co‐injection of MSC with B16 melanoma cells favoured the tumour growth in allogeneic recipient mice Citation100. Although co‐transplantation of MSC with HSC has given very promising results we obviously need more experimental and clinical data in order to determine the value of co‐transplantation of MSC in different clinical conditions.

Safety of MSC transplantation and malignant growth

Stem cells' ability for self‐renewal includes a potential possibility for unlimited proliferation and growth. This immediately prompts two questions: First, can transplanted MSC become malignant raising thus a major concern about the safety of adult stem cell therapies? Second, are tumours as such derived from either local or bone marrow‐derived stem cells?

Until recently MSC transplantations were thought to be without major risk of malignant transformation. However, several lines of evidence now suggest that in vitro expanded cells may be the target of neoplastic transformation. Serakinci et al. Citation101 transduced human mesenchymal stem cells with the telomerase gene to investigate the neoplastic potential of adult stem cells. The human telomerase reverse transcriptase (hTERT)‐transduced line developed loss of contact inhibition, anchorage independence and formed tumours in mice. In the following experiments hTERT‐transduced cell lines showed highly variable tumorigenecity. Han et al. Citation102 was also able to establish a tumour cell line from mutated human embryonic bone marrow mesenchymal stem cells (MSC). They further showed that nucleostemin may play an important role in both tumorigenesis and transforming human embryonic bone marrow mesenchymal stem cells into tumour cells. Finally, Rubio et al. Citation103 demonstrated that although MSC can be managed safely during the standard ex vivo expansion period (6–8 weeks), they can undergo spontaneous transformation during long‐term in vitro culture (4–5 months). On the other hand, several clinical and experimental trials have been performed without any signs of transformation. However, the above mentioned studies clearly arouse serious concerns on the use of in vitro expanded MSC.

The role bone marrow MSC in the progression of cancer metastases deserves some attention, too. It is well known that especially breast cancer metastases in bone marrow are associated with increased bone resorption and enhancement of osteoclast formation from HSC population. Previously it was thought that this was mainly due to the fact that breast cancer cells secrete osteoclast stimulating factors. However, more recent data suggest also that MSC in bone marrow may be the primary target of cancer cells‐produced cytokines and mediate the stimulatory effect to osteoclast progenitors. In the case of prostate cancer metastasis it is even more obvious that their metastases directly stimulate MSC to differentiate into osteoblasts. Thus different tumour cells may induce specific changes in the bone marrow microenvironment and modify stem cell plasticity accordingly.

According to cancer stem cell theory epithelial tumours are thought to originate from transformation of tissue stem cells. If cancer stem cells could in fact be directly recruited from bone marrow by some non‐specific stimuli, such as chronic inflammation, this would be an interesting possibility. According to results of Houghton et al. Citation104 this could the case. They showed that chronic infection of C57BL/6 mice with helicobacter induces repopulation of the stomach with bone marrow derived stem cells. These cells then progressed through metaplasia and dysplasia to cancer.

References

  • Cohnheim J. Ueber Entzündung und Eiterung. (In German.) Pathol Anat Physiol Klin Med. (Archiv für pathologische Anatomie und Physiologie und für klinische Medicin) 1867; 40: 1–79
  • Friedenstein A. J., Piatetzky‐Shapiro I. I., Petrakova K. V. Osteogenesis in transplants of bone marrow cells. J Embryol Exp Morphol 1996; 3: 381–90
  • Friedenstein A. J., Petrakova K. V., Kurolesova A. I., Frolova G. P. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968; 2: 230–47
  • Friedenstein A. J., Chailakhjan R. K., Lalykina K. S. The development of fibroblast colonies in monolayer cultures of guinea pig bone marrow and spleen cells. Cell Tissue Kinet 1970; 4: 393–403
  • Friedenstein A. J., Gorskaja J. F., Kulagina N. N. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol 1976; 4: 267–274
  • Friedenstein A. J., Chailakhyan R. K., Gerasimov U. V. Bone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet 1987; 20: 263–272
  • Caplan A. I. Mesenchymal stem cells. J Orthop Res 1991; 5: 641–50
  • Caplan A. I. The mesengenic process. Clin Plast Surg 1994; 3: 429–35
  • Prockop D. J., Gregory C. A., Spees J. L. One strategy for cell and gene therapy: Harnessing the power of adult stem cells to repair tissues. Proc Natl Acad Sci U S A 2003; 100((Suppl 1))11917–23
  • Horwitz E. M., Gordon P. L., Koo W. K., Marx J. C., Neel M. D., McNall R. Y., et al. Isolated allogeneic bone marrow‐derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: Implications for cell therapy of bone. Proc Natl Acad Sci U S A 2002; 99: 8932–37
  • Le Blanc K., Gotherstrom C., Ringden O., Hassan M., McMahon R., Horwitz E., et al. Fetal mesenchymal stem‐cell engraftment in bone after in utero transplantation in a patient with severe osteogenesis imperfecta. Transplantation 2005; 79: 1607–14
  • Koc O. N., Day J., Nieder M., Gerson S. L., Lazarus H. M., Krivit B. Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS‐IH). Bone Marrow Transplant 2002; 30: 215–22
  • Koc O. N., Gerson S. L., Cooper B. W., Dyhouse S. M., Haynesworth S. E., Caplan A. I., 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
  • Friedenstein A. J., Deriglasova U. F., Kulagina N. N., Panasuk A. F., Rudakowa S. F., Luria E. A., et al. Precursors for fibroblasts in different populations of heamtopoietic cells as detected by the in vitro colony assay method. Exp Hematol 1974; 2: 83–92
  • Friedenstein A. J., Chailakhyan R. K., Latsinik N. V., Panasyuk A. F., Keiliss‐Borok I. V. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation 1974; 17: 331–40
  • Friedenstein A. J., Gorskaja J. F., Kulagina N. N. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol 1976; 4: 267–74
  • Friedenstein A. J. Stromal mechanisms of bone marrow: cloning in vitro and retransplantation in vivo. Haematol Blood Transfus 1980; 25: 19–29
  • Liechty K. W., MacKenzie T. C., Shaaban A. F., Radu A., Moseley A. M., Deans R., et al. Human mesenchymal stem cells engraft and demonsrate site‐specific differentiation after in utero transplantation in sheep. Nat Med 2000; 6: 1282–86
  • Stewart K., Monk P., Walsh S., Jefferiss C. M., Letchford J., Beresford J. N. STRO‐1, HOP‐26 (CD63), CD49a and SB10 (CD166) as markers of primitive human marrow stromal cells and their more differentiated progeny: a comparative investigation in vitro. Cell Tissue Res 2003; 313: 281–90
  • Luria E. A., Panasvuk A. F., Friedenstein A. Y. Fibroblast colony formation from monolayer cultures of blood cells. Transfusion 1971; 11: 345–49
  • Kuznetsov S. A., Mankani M. H., Gronthos S., Satomura K., Bianco P., Robey P. G. Circulating skeletal stem cells. J Cell Biol 2001; 153: 1133–40
  • BIeback K., Kern S., Kluter H., Eichler H. Critical parameters for the isolation of mesenchymal stem cells from umbilical cord blood. Stem Cells 2004; 22: 625–34
  • Romanov Y. A., Svintsitskaya V. A., Smirnov V. N. Searching for alternative sources of postnatal human mesenchymal stem cells: candidate MSC‐like cells from umbilical cord. Stem Cells 2003; 21: 105–10
  • Igura K., Zhang X., Takahashi K., Mitsuru A., Yamaguchi S., Takashi T. A. Isolation and characterization of mesenchymal progenitor cells from chorionic villi of human placenta. Cytotherapy 2004; 6: 543–53
  • Tsai M. S., Lee J. L., Chang Y. J., Hwang S. M. Isolation of human multipotent mesenchymal stem cells from second‐trimester amniotic fluid using a novel two‐stage culture protocol. Hum Reprod 2004; 19: 1450–56
  • Warejcka D. J., Harvey R., Taylor B. J., Young H. E., Lucas P. A. A population of cells isolated from rat heart capable of differentiation into several mesodermal phenotypes. J Surg Res 1996; 62: 233–42
  • Young H. E., Mancini M. L., Wright R. P., Smith J. C., Black AC J. r., Reagan C. R., et al. Mesenchymal stem cells reside within the connective tissues of many organs. Dev Dyn 1995; 202: 137–44
  • Katz A. J., Tholpady A., Tholpady S. S., Shang H., Ogle R. C. Cell surface and transcriptional characterization of human adipose‐derived adherent stromal (hADAS) cells. Stem Cells 2005; 23: 412–23
  • Fickert S., Fiedler J., Brenner R. E. Identification, quantification and isolation of mesenchymal progenitor cells from osteoarthritic synovium by fluorescence automated cell sorting. Osteoarthritis Cartilage 2003; 11: 790–800
  • Hu Y., Liao L., Wang Q., Ma L., Ma G., Jiang X., et al. Isolation and identification of mesenchymal stem cells from human fetal pancreas. J Lab Clin Med 2003; 141: 342–9
  • Baksh D., Song L., Tuan R. S. Adult mesenchymal stem cells: characterization, differentiation, and application in cell and gene therapy. J Cell Mol Med 2004; 8: 301–16
  • Sotiropoulou P. A., Perez S. A., Salagianni M., Baxevanis C. N., Papamichail M. Characterization of the optimal culture conditions for clinical scale production of human mesenchymal stem cells. Stem Cells, 2005. In press
  • Musina R. A., Bekchanova E. S., Sukhikh G. T. Comparison of mesenchymal stem cells obtained from different human tissues. Bull Exp Biol Med 2005; 139: 504–9
  • 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
  • Prindull G. Hypothesis: cell plasticity, linking embryonal stem cells to adult stem cell reservoirs and metastatic cancer cells?. Exp Hematol 2005; 33: 738–46
  • Zhou S., Turgeman G., Harris S. E., Leitman D. C., Komm B. S., Bodine P. V., et al. Estrogens activate bone morphogenetic protein‐2 gene transcription in mouse mesenchymal stem cells. Mol Endocrinol 2003; 17: 56–66
  • Qu Q., Perala‐Heape M., Kapanen A., Dahllund J., Salo J., Vaananen H. K., et al. Estrogen enhances differentiation of osteoblasts in moue bone marrow culture. Bone 1998; 22: 201–9
  • Qu Q., Harkonen P. L., Vaananen H. K. Comparative effects of estrogen and antiestrogens on differentiation of osteoblasts in mouse bone marrow culture. J Cell Biochem 1999; 73: 500–7
  • Heino T. J., Hentunen T. A., Vaananen H. K. Conditioned medium from osteocytes stimulates the proliferation of bone marrow mesenchymal stem cells and their differentiation into osteoblasts. Exp Cell Res 2004; 294: 458–68
  • Cool S. M., Nurcombe V. The osteoblast‐heparan sulfate axis: control of the bone cell lineage. Int J Biochem Cell Biol 2005; 37: 1739–45
  • Arikawa T., Omura K., Morita I. Regulation of bone morphogenetic protein‐2 expression by endogenous prostaglandingE2 in human mesenchymal stem cells. J Cell Physiol 2004; 200: 400–6
  • Gerstenfeld L. C., Barnes G. L., Shea C. M., Einhorn T. A. Osteogenic differentiation is selectively promoted by morphogenetic signals from chondrocytes and synergized by a nutrient rich growth environment. Connect Tissue Res 2003; 44: 85–91
  • Davidson D., Blanc A., Filion D., Wang H., Plut P., Pfeffer G., et al. Fibroblast growth factor (FGF) 18 signals through FGF receptor 3 to promote chondrogenesis. J Biol Chem 2005; 280: 20509–15
  • Solchaga L. A., Penick K., Porter J. D., Goldberg V. M., Caplan A. I., Welter J. F. FGF‐2 enhances the mitotic and chonrogenic potentials of human adult bone marrow‐derived mesenchymal stem cells. J Cell Physiol 2005; 203: 398–409
  • Schmitt B., Ringe J., Haupl T., Notter M., Manz R., Burmester G. R., et al. BMP2 initiates chondrogenic lineage development of adult human mesenchymal stem cells in high‐density culture. Differentiation 2003; 71: 567–77
  • Janderova L., McNeil M., Murrell A. N., Mynatt R. L., Smith S. R. Human mesenchymal stem cells as an in vitro model for human adipogenesis. Obes Res 2003; 11: 65–74
  • Neubauer M., Fischbach C., Bauer‐Kreisel P., Lieb E., Hacker M., Tessmar J., et al. Basic fibroblast growth factor enhances PPARgamma ligand‐induced adipogenesis of mesenchymal stem cells. FEBS Lett 2004; 577: 277–83
  • Palmer G. D., Steinert A., Pascher A., Gouze E., Gouze J. N., Betz O., et al. Gene‐induced chondrogenesis of primary mesenchymal stem cells in vitro. Mol Ther 2005; 12: 219–28
  • Gregory C. A., Singh H., Perry A. S., Prockop D. J. The Wnt signaling inhibitor dickkopf‐1 is required for reentry into the cell cycle of human adult stem cells from bone marrow. J Biol Chem 2003; 278: 28067–78
  • Gregory C. A., Perry A. S., Reyes E., Conley A., Gunn W. G., Prockop D. J. Dkk‐1‐derived synthetic peptides and lithium chloride for the control and recovery of adult stem cells from bone marrow. J Biol Chem 2005; 280: 2309–23
  • Gregory C. A., Gunn W. G., Reyes E., Smolarz A. J., Munoz J., Spees J. L., et al. How wnt signaling affects bone repair by mesenchymal stem cells from the bone marrow. Ann NY Acad Sci 2005; 1049: 97–106
  • Moerman E. J., Teng K., Lipschitz D. A., Lecka‐Czernik B. Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR‐gamma2 transcription factor and TGF‐beta/BMP signaling pathways. Aging Cell 2004; 3: 379–89
  • Hong J. H., Hwang E. S., McManus M. T., Amsterdam A., Tian Y., Kalmukova R., et al. TAZ, a stranscriptional modulator of mesenchymal stem cell differentiation. Science 2005; 309: 1074–8
  • Laskela H. V., Risteli J., Niskanen S., Koivunen J., Ivaska K. K., Lehenkari P. Osteoblast recruitment from stem cells does not decrease by age at late adulthood. Biochem Biophys Res Commun 2003; 311: 1008–13
  • Dressler M. R., Butler D. L., Boivin G. P. Effects of age on the repair ability of mesenchymal stem cells in rabbit tendon. J Orthop Res 2005; 23: 287–93
  • Gotherstrom C., West A., Liden J., Uzunel M., Lahesmaa R., Blanc Le. Difference in gene expression between human fetal liver and adult bone marrow mesenchymal stem cells. Haematologica 2005; 90: 1017–26
  • Baxter M. A., Wynn R. F., Jowitt S. N., Wraith J. E., Fairbairn L. J., Bellatuono I. Study of telomere length reveals rapid aging of human marrow stromal cells following in vitro expansion. Stem Cells 2004; 22: 675–82
  • Liu L., DiGirolamo C. M., Navarro P. A., Blasco M. A., Keefe D. L. Telomerase deficiency impairs differentiation of mesenchymal stem cells. Exp Cell Res 2004; 294: 1–8
  • Goshima J., Goldberg V. M., Caplan A. I. The osteogenic potential of culture‐expanded rat marrow mesenchymal cells assayed in vivo in calcium phosphate ceramic blocks. Clin Orthop Relat Res 1991; 262: 298–311
  • Caplan A. I. Stem cell delivery vehicle. Biomaterials 1990; 11: 44–6
  • Caplan A. I., Bruder S. P. Mesenchymal stem cells: building blocks for molecular medicine in the 21st century. Trends Mol Med 2001; 7: 259–64
  • Shahdadfar A., Fronsdal K., Haug T., Reinholt F. P., Brinchmann J. E. in vitro expansion of human mesenchymal stem cells: Choice of serum is a determinant of cell proliferation, differentiation, gene expression and transcriptome stability. Stem Cells, 2005; August. Epub ahead of print. In press
  • Bruinink A., Tobler U., Halg M., Grunert J. Effects of serum and serum heat‐inactivation on human bone derived osteoblast progenitor cells. J Mater Sci Mater Med 2004; 15: 497–501
  • Noel D., Gazit D., Bouguet C., Apparailly F., Bony C., Plence P., et al. Short‐term BMP‐2 expression is sufficient for in vivo osteochondral differentiation of mesenchymal stem cells. Stem Cells 2004; 22: 74–85
  • Reddi A. H., Cunningham N. S. Bone induction by osteogenin and bone morphogenetic proteins. Biomaterials 1990; 11: 33–4
  • Terai M., Uyama T., Sugiki T., Li X. K., Umezawa A., Kiyono T. Immortalization of human fetal cells: the life span of umbilical cord blood‐derived cells can be prolonged without manipulating p161NK4a/RB braking pathway. Mol Biol Cell 2005; 16: 1491–9
  • Kratchmarova I., Blagoev B., Haack‐Sorensen M., Kassem M., Mann M. Mechanism of divergent growth factor effects in mesenchymal stem cell differentiation. Science 2005; 308: 1472–7
  • Mauney J. R., Volloch V., Kaplan D. L. Role of adult mesenchymal stem cells in bone tissue engineering applications; current status and future prospects. Tissue Eng 2005; 11: 787–802
  • Mundy G., Garrett R., Harris S., Chan J., Chen D., Rossini G., et al. Stimulation of bone formation in vitro and in rodents by statins. Science 1999; 286: 1946–9
  • Gregory C. A., Gunn W. G., Reyes E. Smolarz AJ, Munoz J, Spees JL, et al. How wnt signaling affects bone repair by mesenchymal stem cells from the bone marrow. Ann NY Acad Sci 2005; 1049: 97–106
  • Chamberlain J. R., Schwarze U., Wang P. R., Hirata R. K., Hankenson K. D., Pace J. M., et al. Gene targeting in stem cells from individuals with osteogenesis imperfecta. Science 2004; 303: 1198–201
  • Luyten F. P. Mesenchymal stem cells in osteoarthritis. Curr Opin Rheumatol 2004; 16: 599–603
  • Ichinose S., Yamagata K., Sekiya I., Muneta T., Tagami M. Detailed examination of cartilage formation and endochondral ossification using human mesenchymal stem cells. Clin Exp Pharmacol Physiol 2005; 32: 561–70
  • Pittenger M., Vanguri P., Simonetti D., Young R. Adult mesenchymal stem cells: potential for muscle and tendon regeneration and use in gene therapy. J Musculoskelet Neuronal Interact 2002; 2: 309–20
  • Hui J. H., Ouyang H. W., Hutmacher D. W., Goh J. C., Lee E. H. Mesenchymal stem cells in musculoskeletal tissue engineering: a review of recent advances in National University of Singapore. Ann Acad Med Singapore 2005; 34: 206–12
  • Vunjak‐Novakovic G., Meinel L., Altman G., Kaplan D. Bioreactor cultivation of osteochondral grafts. Orthod Craniofac Res 2005; 8: 209–18
  • Young H. e., Ceballos E. M., Smith J. C., Mancini M. L., Wright R. P., Ragan B. L., et al. Pluripotent mesenchymal stem cells reside within avian connective tissue matrices. In Vitro Cell Dev Biol Anim 1993; 29A: 723–36
  • Amado L. C., Saliaris A. P., Schuleri K. H., St John M., Xie J. S., Cattaneo S., et al. Cardiac repair with intramyocardial injection of allogeneic mesenchymal stem cells after myocardial infarction. Proc Natl Acad Sci U S A 2005; 102: 11474–9
  • Hattan N., Kawaguchi H., Ando K., Kuwabara E., Fujita J., Murata M., et al. Purified cardiomyocytes from bone marrow mesenchymal stem cells produce stable intracardiac grafts in mice. Cardiovasc Res 2005; 65: 334–44
  • Tang Y. L., Zhao Q., Qin X., Shen L., Cheng L., Ge J., et al. Paracrine action enhances the effects of autologous mesenchymal stem cell transplantation on vascular regeneration in rat model of myocardial infarction. Ann Thorac Surg 2005; 80: 229–36
  • Gnecchi M., He H., Liang O. D., Melo L. G., Morello F., Mu H., et al. Paracrine action accounts for marked protection of ischemic heart by Akt‐modified mesenchymal stem cells. Nat Med 2005; 11: 367–8
  • Nagaya N., Kangawa K., Itoh T., Iwase T., Murakami S., Miyahara Y., et al. Transplantation of mesenchymal stem cells improves cardiac function in a rat model of dilated cardiomyopathy. Circulation 2005; 112: 1128–35
  • Ma Y., Xu Y., Xiao Z., Yang W., Zhang C., Song E., et al. Reconstruction of chemically burned rat corneal surface by bone marrow‐derived human mesenchymal stem cells. Stem Cells, 2005; August. Epub ahead of print. In press
  • Nakamura K., Ito Y., Kawano Y., Kurozumi K., Kobune M., Tsuda H., et al. Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther 2004; 11: 1155–64
  • Studeny M., Marini F. C., Dembinski J. L., Zompetta C., Cabreira‐Hansen M., Bekele B. N., et al. Mesenchymal stem cells: potential precursors for tumor stroma and targeted‐delivery vehicles for anticancer agents. J Natl Cancer Inst 2004; 96: 1593–603
  • Lu L., Zhao C., Liu Y., Sun X., Duan C., Ji M., et al. Therapeutic benefit of TH‐engineering mesenchymal stem cells for Parkinson's disease. Brain Res Brain Res Protoc 2005; 15: 46–51
  • Potapova I., Plotnikov A., Lu Z., Danilo P Jr., Valiunas V., Qu J., et al. Human mesenchymal stem cells as a gene delivery system to create cardiac pacemakers. Circ Res. 952–9, 2004;
  • Kurozumi K., Nakamura K., Tamiva T., Kawano Y., Ishii K., Kobune M., et al. Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model. Mol Ther 2005; 11: 96–104
  • Aggarwal S., Pittenger M. F. Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood 2005; 105: 1815–22
  • 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
  • Schoeberlein A., Holzgreve W., Dudler L., Hahn S., Surbek D. V. Tissue‐specific engraftment after in utero transplantation of allogeneic mesenchymal stem cells intosheep fetuses. Am J Obstet Gynecol 2005; 192: 1044–52
  • Plumas J., Chaperot L., Richard M. J., Molens J. P., Bensa J. C., Favrot M. C. Mesenchymal stem cells induce apoptosis of activated T cells. Leukemia 2005; 19: 1597–604
  • Jiang X. X., Zhang Y., Liu B., Zhang S. X., Wu Y., Yu X. D., et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte‐derived dendritic cells. Blood 2005; 105: 4120–6
  • Zhang W., Ge W., Li C., You S., Liao L., Han Q., et al. Effects of mesenchymal stem cells on differentiation, maturation, and function of human monocyte‐derived dendritic cells. Stem Cells Dev 2004; 13: 263–71
  • Bartholomew A., Sturgeon C., Siatskas M., Ferrer K., McIntosh K., Patil S., et al. Mesenchymal stem cells suppress lymphocyte proliferation in vitro and prolong skin graft survival in vivo. Exp Hematol 2002; 30: 42–8
  • Chapel A., Bertho J. M., Bensidhoum M., Fouillard L., Young R. G., Frick J., et al. Mesenchymal stem cells home to injured tissues when co‐infused with hematopoietic cells to treat a radiation‐induced multi‐organ failure syndrome. J Gene Med 2003; 5: 1028–38
  • Troeger A., Meisel R., Moritz T., Dilloo D. Immunotherapy in allogeneic hematopoietic stem cell transplantation – not just a case for effector cells. Bone Marrow Transplant 2005; 35: S59–64
  • Zappia E., Casazza S., Pedemonte E., Benvenuto F., Bonánni I., Gerdoni E. Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T‐cell anergy. Blood 2005; 106: 1755–61
  • Jones O. y., Good R. A., Cahill R. A. Nonmyeloablative allogeneic bone marrow transplantation for treatment of childhood overlap syndrome and small vessel vasculitis. Bone Marrow Transplant 2004; 33: 1061–3
  • Djouad F., Plence P., Bony C., Tropel P., Apparailly F., Sany J., et al. Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals. Blood 2003; 102: 3837–44
  • Serakinci N., Guldberg P., Burns J. S., Abdallah B., Schrodder H., Jensen T., et al. Adult human mesenchymal stem cell as a target for neoplastic transformation. Oncogene 2004; 23: 5095–8
  • Han C., Zhang X., Xu W., Wang W., Qian H., Chen Y. Cloning of the nucleostemin gene and its function in transforming human embryonic bone marrow mesenchymal stem cells into F6 tumor cells. Int J Mol Med 2005; 16: 205–13
  • Rubio D., Garcia‐Castro J., Martin M. C., de la Fuente R., Cigudosa J. C., Lloyd A. C., et al. Spontaneous human adultstem cell transformation. Cancer Res 2005; 65: 3035–9
  • Houghton J., Stoicov C., Nomura S., Rogers A. B., Carlson J., Li H., et al. Gastric cancer originating from bone marrow‐derived cells. Science 2004; 306: 1568–71

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.