2,740
Views
138
CrossRef citations to date
0
Altmetric
Review

Mesenchymal stem cells in tumor development

Emerging roles and concepts

&
Pages 220-230 | Published online: 01 May 2012

Abstract

Mesenchymal stem cells (MSCs) are multipotent progenitor cells that participate in the structural and functional maintenance of connective tissues under normal homeostasis. They also act as trophic mediators during tissue repair, generating bioactive molecules that help in tissue regeneration following injury. MSCs serve comparable roles in cases of malignancy and are becoming increasingly appreciated as critical components of the tumor microenvironment. MSCs home to developing tumors with great affinity, where they exacerbate cancer cell proliferation, motility, invasion and metastasis, foster angiogenesis, promote tumor desmoplasia and suppress anti-tumor immune responses. These multifaceted roles emerge as a product of reciprocal interactions occurring between MSCs and cancer cells and serve to alter the tumor milieu, setting into motion a dynamic co-evolution of both tumor and stromal tissues that favors tumor progression. Here, we summarize our current knowledge about the involvement of MSCs in cancer pathogenesis and review accumulating evidence that have placed them at the center of the pro-malignant tumor stroma.

Introduction

The study of tumor pathogenesis has, for many years, largely focused on the accumulation of genetic or epigenetic alterations intrinsic to cancer cells, while almost entirely disregarding the vital contributions of the tumor stroma.Citation1 However, in the past decade or so, studies focused on understanding the complex crosstalk between cancer cells and the heterogeneous milieu of tumor stromal cells have allowed for an increased appreciation of the critical nature of some of these interactions, not only in supporting, but also in driving tumor growth and progression.Citation2 We now recognize that developing tumors can mobilize a variety of cell types from both local and distant niches via secreted chemical factors derived from the cancer cells themselves or from neighboring cells disrupted by a growing neoplasm.Citation3 These recruited cells significantly alter the composition of the tumor milieu and set into motion a complex series of interactions which result in the co-evolution of both cancer and stromal compartments.Citation4,Citation5

The stroma of solid cancers contains a variety of mesenchymal cell types, such as endothelial cells, lymphocytes, macrophages, neutrophils and cancer-associated fibroblasts, whose contributions to tumor development have now been extensively characterized and are the subjects of accompanying reviews in this Special Focus. More recently appreciated, however, are the contributions of a class of multipotent mesenchymal progenitor cells found to reside within the tumor microenvironment called mesenchymal stem cells (MSCs) (e.g., refs. Citation6Citation8). In the last few years, MSCs have been demonstrated to play important roles in tumor pathogenesis and are for this reason the subject of intense investigation.

MSCs are a heterogeneous class of self-renewing, multipotent progenitor cells that reside primarily in the bone marrow, but can also be found in a variety of other tissues throughout the body.Citation9-Citation14 They display a number of remarkable properties, such as the tendency to home to sites of injury, the capacity to suppress immune reactions and the ability to aid in the repair and regeneration of damaged tissues.Citation15-Citation17 Accordingly, MSCs have been explored widely for their applications in regenerative medicine and as delivery vehicles for use in gene therapy.Citation18

In the context of cancer, MSCs are becoming increasingly recognized as important stromal facilitators of tumor development. Indeed, MSCs display avid tropism for developing tumors, akin to their abilities to home to wounded tissuesCitation19 and are integral components of the cancer stroma in experimental as well as in clinical settings (e.g., refs. Citation6, Citation7 and Citation20Citation22). Furthermore, numerous studies have now demonstrated that human MSCs enhance tumor growth and/or metastatic progression in neoplasias arising from a wide range of tissues.Citation23-Citation34 Although the specifics of the mechanisms by which MSCs enhance tumor progression and metastasis are only beginning to be understood, their reported influences can now be generally classified into four broad functional categories. First, MSCs within the tumor stroma may exert direct paracrine influences on the cancer cells, promoting tumor proliferation, invasion and metastasis (e.g., ref. Citation21). Second, tumor-associated MSCs may exert indirect pro-malignant actions by promoting tumor angiogenesis through recruitment of endothelial progenitor cells and by facilitating the formation and maturation of tumor vasculature (e.g., refs. Citation35 and Citation36). Third, by virtue of their progenitor status, MSCs may respond to the panoply of signals and cues present in the tumor microenvironment by differentiating into other types of stromal cells. For example, MSCs have been reported to differentiate into cancer-activated fibroblasts (CAFs), which can in turn influence tumor development.Citation37,Citation38 Fourth, stromal MSCs may exert immunomodulatory properties that protect the tumor cells from detection and destruction by the adaptive immune system, functions that can be manifested through the direct or indirect actions of MSCs on various immune cells (reviewed in ref. Citation39). Because of these activities, MSCs are becoming increasingly accepted as important contributors to tumor progression, likely representing a critical component of the tumor-associated stroma. Here, we briefly expand on these themes and summarize recent efforts in elucidating the mechanisms by which these stromal stem cells contribute to tumor pathogenesis.

Identification and Characterization of MSCs

The initial characterization of MSCs dates to the observations of Friedenstein and colleagues, who cultured and propagated bone-marrow-derived non-hematopoietic cells that had the potential to give rise to bone and cartilageCitation40 (reviewed in ref. Citation41). These findings were reproduced by further studies in the subsequent yearsCitation42-Citation48 and paved the way toward the definitive identification of multipotent stem cells within heterogeneous human MSC cultures.Citation41,Citation49 These cells exhibited phenotypic properties that were unambiguously distinct from those of hematopoietic stem cells (HSCs) or endothelial progenitor cells.Citation50

MSCs are classically characterized by their tri-lineage differentiation potential into osteoblasts, chondrocytes and adipocytes.Citation51,Citation52 However, they also possess additional differentiation potentials and have been shown to give rise to myoblasts, endothelial cells, pericytes or fibroblasts and have more recently been reported to generate non-mesenchymal cells as well, such as epithelial cells, hepatocytes or even neuronal cells.Citation49,Citation53-Citation56 Phenotypically, they are plastic-adherent fibroblastic cells that express the cell surface antigens CD29, CD44, CD49, CD73, CD90, CD105, CD106, CD140b, CD166 and STRO-1, but lack key hematopoietic markers such as CD11b, CD14, CD19, CD31, CD34, CD45 and CD133.Citation52,Citation57 Using these markers, researchers have been able to isolate MSCs or MSC-like cells from multiple tissues, including peripheral blood, adipose tissue, umbilical cord blood, fetal liver, lung, amniotic fluid, synovial fluid and gingival tissue.Citation9,Citation13,Citation14,Citation58-Citation61 Genotypically, MSCs derived from different tissue sources appear to exhibit different expression levels of the embryonic-stem-cell-associated pluripotency factors NANOG, OCT-4 and/or SOX2.Citation62,Citation63 However, the functional contribution of such transcription factors to the proliferative and differentiation capacities of MSCs is still a matter of active debate (e.g., refs. Citation64 and Citation65).

MSCs in Wound Responses

MSCs play important roles in maintaining normal tissue homeostasis under resting conditions. These activities include the regulation of vital processes, such as hematopoiesis,Citation49,Citation66,Citation67 the preservation of blood vessel integrityCitation68 or bone maintenance.Citation69 More appreciated, however, are the functions of MSCs in cases of wound healing and tissue repair.Citation70 In these respects, current models suggest that MSCs are dispatched from their niches in response to systemic signals derived from injured tissues (e.g., ref. Citation71). The subsequent integration of the mobilized MSCs within these tissues is thought to provide an environment conducive to tissue rejuvenation and wound closure.

The “healing” functions of MSCs appear to be manifested via three key activities. First, an ability, by virtue of their plasticity, to differentiate on site to replenish tissues lost during injury. This is especially apparent in osteochondral disorders, where the differentiation of MSCs into chondrocytes or osteoblasts may, at least partly, contribute to the repair of these tissues (reviewed in ref. Citation48). Second, MSCs may exert their actions through the release of trophic factors that contribute to tissue regeneration by stimulating the activation of local tissue-specific stem cells. For instance, MSCs have been suggested to enhance the proliferation of local cardiac stem cells in heart infarction models.Citation72 Third and importantly, MSCs appear to promote immunosuppressive environments capable of inhibiting the functions of the adaptive immune system. In fact, MSCs cause the formation of T-regulatory cells (T-regs) in the context of inflammationCitation73 and have been shown to inhibit other aspects of immune cell functions, such as antibody releaseCitation74 and dendritic cell maturation.Citation75,Citation76 While these aforementioned activities entail the local engraftment of MSCs in sites of wounding, new evidence indicates that MSCs may not even need to engraft locally at the wounded sites in order to enhance tissue repair, suggesting that they may also exert actions at a distance.Citation77-Citation79

Due to their increased avidity to wounds in experimental models, as well as their differentiation versatility, MSCs are being heavily explored for applications in regenerative medicineCitation80 and hold great clinical promise for the treatment of a number of diseases in a wide variety of tissues, including osteochondral diseases,Citation81,Citation82 cardiovascular diseases,Citation83,Citation84 liver disease,Citation85 renal diseases,Citation86 spinal cord injuries,Citation87 immunosuppression to benefit organ transplantionCitation88 and neurodegenerative diseases.Citation89,Citation90

MSC Homing to Tumors

Growing tumors continuously remodel local tissue architecture and generate chronic inflammatory responses similar to those evoked by open wounds.Citation19 Accordingly, it is believed that MSCs migrate into tumors in a manner similar to the way they migrate into injured tissues.Citation91 Indeed, MSCs introduced into the systemic circulation of tumor-bearing animals exhibit avid and preferential homing to cancerous growths, with limited homing to other tissues.Citation20,Citation21 This preferential migration has now been convincingly demonstrated in a number of xenograft models, such as melanoma,Citation92 ovarian carcinoma,Citation93 breast carcinomasCitation21 and hepatocellular carcinomasCitation94 and has been described in detail using enhanced detection methods for tracking injected MSCs.Citation22 Importantly, endogenous MSCs have been recovered from the stroma of both experimental xenograft tumorsCitation21 and human tumors,Citation6,Citation7 suggesting that cancer development entails the continuous recruitment of MSCs, which may maintain steady-state levels within tumor stroma.

The prevalent model of MSC recruitment into tumors describes their mobilization from systemic niches, ostensibly in the bone marrow,Citation95 and their subsequent homing to cancer growths in response to chemotactic agents emitted by cancer cells. Although much remains to be proven about the molecular details underlying such a model, accumulating research has begun cataloging the molecules that control the avidity of MSCs to tumor sites. The list of soluble factors governing MSC recruitment to tumors is growing at a fast pace. Early on, due to the high affinity of HSCs and their leukocyte progeny for wound and tumor sites, investigators looked to the already well-characterized processes of HSC and leukocyte homing for clues driving MSC migration.Citation96,Citation97 Indeed, factors involved in HSC and immunocyte recruitment, such as monocyte chemotactic protein-1 (MCP-1),Citation98 stromal-cell derived factor (SDF-1),Citation99 cyclophilin B and hepatoma-derived growth factor (HDGF),Citation100 urokinase plasminogen activator (uPA),Citation101,Citation102 interleukin (IL-)6,Citation103 basic fibroblast growth factor (bFGF)Citation104 and vascular endothelial growth factor (VEGF)Citation104 have all been implicated in driving the tropism of bone-marrow-derived MSCs to tumors. It is believed that an active inflammatory response is necessary to allow MSC recruitment to tumor sites.Citation91 Many such factors, produced as a result of tissue damage or growing neoplasms, exhibit chemoattractant properties toward MSCs in vitro and include bFGF,Citation105 VEGF,Citation106 platelet-derived growth factor (PDGF), insulin-like growth factor (IGF) and variety of other growth factors such as transforming growth factor (TGFβ),Citation107 chemokines and cytokines,Citation108 cathelicidin (LL-37)Citation109 and even complement components C3a and C5a.Citation110 Non-neural cholinergic mediatorsCitation111 and dopamine derived from innervating sympathetic nerves have also recently been implicated in MSC homing to wound sites.Citation112 It is important to note, however, that the inhibition of the actions of any one single factor does not appear to be sufficient to completely disrupt MSC homing to tumors, suggesting that multiple concerted mechanisms cooperate in regulating their tropism to tumor growths.Citation113

MSCs derived from non-bone-marrow sources also possess the abilities to home to tumors and appear to respond to chemotactic signals emitted by cancer cells just as well as bone-marrow-derived MSCs. For example, umbilical-cord-derived MSCs home to medulloblastoma cells in response to matrix metalloproteases (MMP)-2Citation99 and adipose-derived stromal cells can contribute to the malignancy of cancer cell lines derived from a number of different tissues.Citation26-Citation29,Citation114 These observations challenge the current dogma that tumor-associated MSCs are derived solely from bone marrow sources and raise the notion that other MSC-rich tissues (such as adipose tissue) may also contribute to the pools of certain tumor stromal MSCs.

A critical component of the homing of MSCs to cancer sites is their ability to execute transmigration through the endothelial cells of the vessel wall. How MSCs interact specifically with endothelial cells is an area that is receiving a lot of attention and is starting to be illuminated with increasing molecular clarity. The identification of the adhesion molecules and surface receptors that guide the vascular adhesion of MSCs has been aided by comparisons to the similar activities of HSCs and leukocytes (reviewed in ref. Citation115). Like hematopoietic cells, MSCs appear to utilize E-selectin for vascular adhesion, but lack other hematopoietic cell adhesion molecules such as L-selectin, β2 integrins and platelet endothelial cell adhesion molecule-1 (PECAM-1)/CD34 that facilitate the rolling of HSCs on vasculature and their subsequent transmigration through the endothelial wall.Citation116 Alternatively, other sets of adhesion molecules, such as endothelial-cell-expressed P-selectin, which can interact with CD44 on the surface of MSCs,Citation117 or vascular cell adhesion molecule-1 (VCAM-1),Citation118 C3a, C5aCitation110 and C-X-C motif chemokine 5 (CXCL5)Citation119 have all been shown to be important for MSC extravasation across endothelial barriers. Finally, a prominent mechanism reported to control the efficiency of transendothelial migration of MSCs is shear stress. Indeed, low-shear conditions cause the downregulation of certain chemokine receptors, such as C-X-C motif chemokine receptors (CXCR) 3 and 6 and C-C chemokine receptors (CCR) 6 and 9 in cells crossing the aortic endothelial barrier,Citation120 while high shear-stress conditions appear to cause the upregulation of integrins in MSCs adhering on endothelial ligands.Citation116

While the specifics regarding MSC homing to tumor sites are still being investigated, new evidence suggests that stromal cells also participate in MSC recruitment into tumors. For example, cancer-associated fibroblasts (CAFs) reported to be derived from MSCs in the context of gastric cancers have been shown to further recruit primitive MSCs from the bone marrow via the secretion of SDF-1.Citation37,Citation38 This raises the notion that the homing and integration of MSCs into tumors may initiate a vicious cycle, causing further recruitment of MSCs, thereby maintaining their numbers in tumor stroma and sustaining their contributions to tumor pathogenesis.

MSCs in Tumor Pathogenesis

The contributions of tumor-associated MSCs to cancer development have been the subject of intense investigations. While certain studies suggest that MSCs play tumor-suppressive roles (e.g., ref. Citation121), they are outnumbered by an overwhelming literature that has incriminated MSCs in serving tumor-promoting functions and in a wide range of cancer models. Indeed, MSCs have now been demonstrated to serve pro-malignant roles in a number of epithelial cancer subtypes, such as breast,Citation21,Citation27 colon,Citation25,Citation30 lung,Citation26,Citation32 skinCitation23,Citation24,Citation29 or prostate.Citation28,Citation31 MSCs have also been shown to drive hematopoietic malignancies, such as multiple myeloma,Citation122,Citation123 and appear to sustain leukemia/lymphoma development.Citation124 Finally, MSCs have recently been found to promote the progression of certain mesenchymal cancers as well, such as osteosarcomas, which are thought themselves to arise, in part, from the neoplastic transformation of MSC lineagesCitation34 (discussed in more detail below).

As their roles in tumor pathogenesis are still being characterized in detail, several major mechanisms through which MSCs contribute to tumor development are emerging. First, MSCs exert direct actions on the cancer cells through the secretion of a variety of bioactive molecules whose paracrine actions influence the phenotype of the cancer cells. Second, the immunosuppressive properties of MSCs, as described above, alter the local composition of immunocytes and derail immune reactions that are mounted against malignant cells, therefore providing an immune-privileged environment for neoplastic cells. Third, MSCs can influence tumor vascularization by exacerbating tumor angiogenesis. Fourth and finally, as progenitor cells, tumor-associated MSCs have been reported to differentiate within the tumor microenvironment and act as local sources for other tumor stromal cells, such as tumor-associated fibroblasts (TAFs). In the following sections, we will summarize the current knowledge pertaining to each of these mechanisms and explore in more detail the varied functions of tumor-associated MSCs in tumor biology.

Direct actions of MSCs on cancer cells

MSCs produce a plethora of molecules, such as chemokines, cytokines and growth factors, which act in a paracrine fashion on their respective receptors on the surface of cancer cells, thereby regulating tumor growth and/or progression. For example, MSC-derived chemokines, such as CXCL1, CXCL2 or CXCL12/SDF-1, have been shown to foster cancer cell proliferation in a number of cancer models via their actions on their respective CXCR2 and CXCR4 receptors on cancer cells.Citation125,Citation126 Similarly, cytokines secreted by MSCs, which include IL-6 and IL-8, have been demonstrated to enhance cancer cell malignancy in the context of several cancers, such as breastCitation6 or colorectal.Citation127 Finally, MSC-derived growth factors, such as epidermal growth factor (EGF), act on their cognate receptors on the surface of cancer cells, enhancing tumorigenesis in the setting of breast cancer.Citation8

While the list of MSC-derived peptide messengers is expanding, it is important to note that MSCs can also provide additional agents/materials that can act on the cancer cells in a paracrine fashion. In these respects, new research suggests that nucleic acids are effectively transferred from MSCs to cancer cells, potentially via mRNA- and/or microRNA-rich microvesiclesCitation128 or through gap junctions established following physical interactions between the two cell types.Citation129 These observations suggest that cancer cells effectively respond to the innate abilities of MSCs to produce and secrete a rich and vast array of bioactive molecules that can influence the course of tumor development.

In concert with the rich set of bioactive molecules they “normally” generate, MSCs respond to the contextual signals emanating from the cancer cells in their microenvironment by initiating de novo secretion of soluble factors that impact tumor pathogenesis.Citation21 Of note in this regard is the de novo ability of MSCs to produce the chemokine CCL5 upon stimulation by breast cancer cells.Citation21 Here, CCL5 mRNA levels (and subsequently protein levels as well) in the MSCs are induced by > 1,000 times upon their physical contact with the cancer cells. CCL5 then acts in a paracrine fashion on the cancer cells, fostering metastasis by increasing their ability to extravasate into lung parenchyma in experimental models.Citation21 Interestingly, not all cancer cells possessed the ability to instigate CCL5 from the MSCs, lending credence to the notion that idiosyncratic differences exist between cancer cells (and the way in which they respond to interactions with MSCs) even within the same tumor subtype. Similarly, the interaction of MSCs with lung cancer cell lines was found to cause stanniocalcin-1 (STC-1) secretion by the activated MSCs, which in turn upregulates uncoupling protein 2 to protect the neighboring cancer cells from ROS-induced apoptosis.Citation130,Citation131 Taken together, these findings raise the notion that contextual signals control the manner of MSC activation and, accordingly, may regulate their functions within the tumor microenvironment (discussed in more detail below).

The transcriptional, secretory and phenotypic changes induced in MSCs following their interactions with cancer cells likely underlie some of the tumor-beneficial effects they potentiate while contributing to the dynamic evolution of the tumor stroma. However, it is important to note that cancer cells also undergo phenotypic and transcriptional changes as a result of their interactions with MSCs. A gamut of different MSC-instigated signal transduction pathways have been studied and they regulate vital machineries in the cancer cells, such as cell cycle/proliferation,Citation8,Citation125,Citation129 but also several phenotypes associated with malignancy, such as motility,Citation21,Citation125,Citation126,Citation132 invasionCitation21,Citation133 and metastasis.Citation21,Citation27,Citation30,Citation134

In this regard, three important MSC-fostered cancer phenotypes deserve particular mention: (1) the ability of MSCs to regulate cancer stem cells (CSCs)—cells with increased tumor-initiating abilities (reviewed in ref. Citation135), (2) their ability to enhance the resistance of cancer cells to chemotherapy and (3) the ability of MSCs to drive the metastatic predilection of cancer cells to certain tissues, such as the bone. Indeed, recent studies have found that labeled human MSCs injected into mouse tibias homed to sites of growing orthotopic breast cancer xenografts and increased the population of cancer stem cells (CSCs) via the paracrine actions of IL-6 and CXCL7.Citation6 Similarly, tumor-associated MSCs were found to enhance the generation of cancer stem cells in ovarian carcinoma, in this case, via BMP2 signaling.Citation7 Along similar lines, MSCs have been described to play a role in cancer-cell-resistance to classical chemotherapy. For example, MSCs were found to protect lung cancer cells from apoptosis as a result of oxidative or chemotherapeutic stress.Citation130 Furthermore, endogenous human MSCs were found to protect tumor cells against chemotherapeutic agents by secreting chemoprotectant polyunsaturated fatty acids in response to activation by platinum analog,Citation136 an action they exert at a distance, without the need to engraft within tumors. Finally, strong evidence suggests that MSCs may preferentially drive tumor metastasis to the bone (e.g., ref. Citation137), through mechanisms that involve, in part, MSC-derived IL17BR.Citation33 Although the molecular underpinnings of the abilities of MSCs to regulate these three phenotypes have not been fully determined, some evidence suggests that the mechanisms underlying such activities may at least be interrelated. Indeed, CSCs and cancer cells resident in bone exhibit increased resistance to chemotherapy and are refractory to tumor irradiation.Citation138

Immunosuppressive properties of MSCs

MSCs act as immune modulators with reported activities in suppressing both innate and adaptive immune responses (reviewed in refs. Citation77 and Citation139). Indeed, their ability to quell the immune system is so potent, it has been exploited to reduce the severity of graft vs. host disease (GvHD).Citation140-Citation142 MSCs appear to modulate immune responses via differential influences they exert on the proliferative capacities of immune cells. For example, MSCs inhibit the proliferation and maturation of B-cellsCitation74 as well as natural killer cells,Citation143-Citation145 while exhibiting protective activities toward other cells, such as neutrophils.Citation146 In similar respects, MSCs have been shown to directly inhibit both CD4+ and CD8+ T-cell proliferation,Citation147 suggesting that MSCs can influence the behavior of almost all of the key immune activities involved in tumor development.

The exact molecular details of how MSCs exert these immunomodulatory functions are only partially characterized. Several mechanisms of action appear to involve their innate or stimulated abilities to secrete cytokines and chemokines, which suppress immune cell proliferation or activation.Citation77,Citation148 For example, MSC-derived interferon-gamma was found to be sufficient in suppressing T-cell effector functions in a number of settings.Citation149 Furthermore, stimulation of Toll-like receptor (TLR)-3 and -4 present on the surface of MSCs leads to the production of IL-6, IL-8 and CXCL10, which were critical for the suppression of T-cell proliferation.Citation150 MSCs may also indirectly quell T-cell proliferation by influencing dendritic cell (DC) proliferation and maturation.Citation75,Citation76,Citation151-Citation153 This is accomplished, in part, by MSCs driving DC differentiation into a subtype incapable of stimulating T-cell expansion.Citation148

Finally and perhaps most importantly, MSCs can direct T-cells to differentiate into T-reg, which, in the case of tumors, can confer growth and metastatic advantages.Citation139,Citation154,Citation155 These advantages are manifested through T-reg-mediated suppression of normal innate and adaptive immune responses, including the direct inactivation of effector T-cells and NK cells (reviewed in ref. Citation156). Although the mechanisms underlying the MSC mediated generation of T-regs is still under investigation, this process appears to involve MSC-secreted TGFβ1.Citation157 Taken together the immune-suppressive effects of MSCs are rather extensive and perhaps unrivaled by any other stromal cell type and constitute crucial protective mechanisms by which MSCs contribute to tumor growth and progression.

MSCs in tumor angiogenesis

MSCs play multifaceted roles in fostering tissue revascularization after injury and appear to serve similar pro-angiogenic functions in the setting of tumor development (reviewed in ref. Citation158). Indeed, MSCs (or their reported progeny; discussed below) may promote tumor neo-angiogenesis via the secretion/production of angiogenic factors, such as VEGF-A, angiopoietins, EGF, keratinocyte growth factor (KGF), IGF-1 and galectin-1Citation159,Citation160 and are involved in the recruitment of endothelial cells and in promoting the maturation of newly-formed blood vessels (e.g., refs. Citation32 and Citation161). Furthermore, MSCs themselves have been shown to be able to differentiate into endothelial-like cells and share cell surface markers with pericytes, thereby modulating the tumor vasculature.Citation162,Citation163 Conversely, vascular endothelial cells have been recently reported to convert into cells with characteristic properties of MSCs,Citation164 suggesting that endothelial-to-mesenchymal or mesenchymal-to-endothelial transformations may be possible in the context of tumorigenesis. These observations highlight important roles for MSCs in supporting tumor vascularization and suggest that targeting MSCs may be a viable avenue in anti-angiogenesis-based cancer therapy.

MSCs as progenitors for tumor stroma

As mentioned above, the crosstalk operating between cancer cells and tumor-associated MSCs changes not only the cancer cells, but the MSCs as well. In these respects, the tumor microenvironment may affect, in addition to the properties of tumor-associated MSCs, their differentiation capacities. In fact, some evidence suggests that MSCs trans-differentiate within tumors to give rise to cancer associated fibroblasts (CAFs; see refs. Citation165 and Citation166).

CAFs have been increasingly appreciated in the last ten years as the importance of the stroma in tumor development has become more evident (e.g., refs. Citation167 and Citation168). They have been associated with poor patient prognosisCitation169,Citation170 and are implicated in serving a multitude of pro-malignant activities, including, for example, the ability to enhance angiogenesis and to foster ECM remodeling (reviewed in ref. Citation171). The notion that MSCs generate CAF populations is highly interesting and holds the potential of shedding much needed light on the origin(s) of CAFs and on the fate of MSCs once recruited into the tumor microenvironment. A cautionary note, however, is that much of the markers associated with CAFs (such as SMA) are already present, to a certain extent, in the MSC populations. Accordingly, it remains to be determined whether the described conversion of MSCs to CAFs is due to the selection of pre-existing cells (e.g., by TGFβCitation165) or whether it results from the bona fide stable differentiation of MSCs into CAFs. Improved and unique functional determinations for the CAF phenotype, along with improved and unique markers characterizing MSCs will be instrumental in clarifying the potential inter-relationship of CAFs to MSCs.

MSCs as Cells-of-Origin for Cancer

MSCs may represent the cell-of-origin for certain sarcomas, such as Ewing’s family sarcomas (reviewed in ref. Citation172). Indeed, a number of observations suggested that the expression of the fusion product between FLI-1 (a member of the E-26 family of transcription factors or ETS genes) and the EWS DNA-binding domain, can cause the transformation of MSCs into Ewing sarcoma cells.Citation173 First, established sarcoma lines in which EWS-FLI-1 was silenced reverted back to a transcriptional program resembling that of MSCs.Citation174 Second, revertants regained a progenitor status and were capable of differentiating into osteoblasts and adipocytes, corroborating their origins as MSCs. Third, this activity appears to be restricted to the highly tumorigenic cancer-stem-cell-rich CD133-positive sarcoma cells, which expresses a series of stem-cell-associated genes, such as NANOG and OCT-4.Citation175 Along the same lines, the transformation of human MSCs by ectopic expression of other sarcoma-associated oncogenes, such as the synovial sarcoma translocated gene product (SYT-SSX1) or the FUS-CHOP gene product, generated cells with the ability to form synovial sarcomasCitation176 or myxoid liposarcomas,Citation177 respectively. Together, these observations suggest that MSCs may very well lie at the origin of multiple sarcomas and that they may be the targets of oncogenic transformation in human disease.

It is noteworthy to add that MSCs have also been directly incriminated in initiating epithelial cancers. Of note is the report by Houghton and colleagues who showed that bone-marrow-derived MSCs initiate gastric cancers by fusing with mucosal cells in a setting of Helicobacter pylori infection.Citation178 Whether MSCs also give rise to other cancers is yet undetermined.

Final Notes and Future Perspectives

The involvement of MSCs in tumor biology has attracted increased attention as of late and the knowledge regarding their biological attributes, their influence on cancer cells and their roles in human cancer pathogenesis is mounting at a fast pace. With this renewed interest comes the promise that such ongoing efforts will bring forth further insights into the still-enigmatic biology of these progenitor cells and the manners with which they impact human cancer development.

In these regards, some of the most important questions pertain to the origin(s) of tumor-associated MSCs. Indeed, whether tumor-associated stromal MSCs derive primarily from bone marrow niches or whether they arrive just as frequently from local reservoirs (similar in many ways to bone-marrow- and tissue-resident pools of hematopoietic stem/progenitor cells) is still unaddressed. This is of particular interest as MSCs derived from different sources do display some differences in the detailed mechanisms through which they support tumor development. Accordingly, the possibility that tumors may harbor MSCs derived from different anatomical origins raises the notion of whether these stromal stem cells contribute to tumor heterogeneity by fostering the evolution of cancer cells they come in contact with along different molecular paths.

Two additional important and interrelated questions that remain to be fully addressed are the degree of plasticity of MSCs once present in the tumor microenvironment and the lack of unique markers with which to distinguish them apart from other stromal cells within tumors. Although increased attention is now focused on the involvement of MSCs in tumor pathogenesis, the field is still complicated by the disparate methodologies still used across many labs to isolate MSCs from (healthy) donors, which contribute to such isolates being invariably different from one another across labs and clinics. The identification of unique MSC markers would, ostensibly, enable researchers to more uniformly determine the differentiation/stem states of MSC preparations derived from different tissues and importantly, would permit the attribution of their influences on tumor initiation and progression to the respective representation of such differentiated and/or stem cells within the original cultures. An attempt at placing a hierarchical relation between different MSCs has been performed recently and has proven to bear functional consequences on tumor development.Citation5

In closing, MSCs appear to reside at the center of a complex crosstalk of interactions that drive the co-evolution of non-transformed stromal cells and neoplastic cells alike. These pathways now include a mounting number of interacting players and processes, summarized in . The critical players essential for this crosstalk are only just beginning to be identified and have already been shown to control key features of cancer malignancy, such as stemness and metastasis. As they are, novel and beneficial anti-neoplastic approaches based on interdicting the crosstalk between MSCs and cancer cells will undoubtedly emerge.

Figure 1. MSCs in tumor pathogenesis. Initial systemic factors released by tumor cells or by the disrupted surrounding tissues cause MSC mobilization and recruitment into tumors; MSCs cross vessel walls and home into cancers. MSC-derived trophic factors, including growth factors, chemokines and cytokines influence cancer cell phenotypes. The crosstalk between MSCs and cancer cells fosters remodeling of the tumor microenvironment and impacts other stromal cell types, including immune cells, enhances angiogenesis and/or permeabilization of the vasculature, causes cancer cell growth, local invasion and metastasis. Systemic factors derived from these interactions may influence distal sites/tissues, including secondary colonization sites.

Figure 1. MSCs in tumor pathogenesis. Initial systemic factors released by tumor cells or by the disrupted surrounding tissues cause MSC mobilization and recruitment into tumors; MSCs cross vessel walls and home into cancers. MSC-derived trophic factors, including growth factors, chemokines and cytokines influence cancer cell phenotypes. The crosstalk between MSCs and cancer cells fosters remodeling of the tumor microenvironment and impacts other stromal cell types, including immune cells, enhances angiogenesis and/or permeabilization of the vasculature, causes cancer cell growth, local invasion and metastasis. Systemic factors derived from these interactions may influence distal sites/tissues, including secondary colonization sites.
Abbreviations:
bFGF=

basic fibroblast growth factor

CXCL_=

C-X-C motif chemokine

CXCR_=

C-X-C motif chemokine receptor

CCL_=

C-C motif chemokine

CCR_=

C-C motif chemokine receptor

CAF or TAF=

cancer-associated fibroblast or tumor-associated fibroblast

EGF=

epidermal growth factor

ETS=

e-twenty-six family of transcription factors

GvHD=

graft versus host disease

HDGF=

hepatoma-derived growth factor

HSC=

hematopoietic stem cell

IGF=

insulin-like growth factor

IL-=

interleukin

KGF=

keratinocyte growth factor

LL-37=

cathelicidin

MCP-1=

monocyte chemotactic protein-1

MMP-2=

matrix metalloproteinase-2

MSC=

mesenchymal stem cell

PECAM-1=

platelet endothelial cell adhesion molecule-1 (CD31)

PDGF=

platelet-derived growth factor

SDF-1=

stromal-cell derived factor-1

TGFβ=

transforming growth factor beta

uPA=

urokinase plasminogen activator

VEGF=

vascular endothelial growth factor

VCAM-1=

vascular cell adhesion molecule-1

Acknowledgments

Work in the Karnoub laboratory is supported by funds from the Beth Israel Deaconess Medical Center (A.E.K.), the Sydney Kimmel Cancer Research Foundation (A.E.K.) and the Susan G. Komen for the Cure (A.E.K.). B.C. is a recipient of a 2012 postdoctoral fellowship from the American Cancer Society. A.E.K. is a 2010 Kimmel Scholar, a recipient of a Career Development Award from the Prostate and Breast Cancer Research Program of the Beth Israel Deaconess Medical Center and a recipient of a Career Catalyst Research Award from the Susan G. Komen for the Cure.

References

  • Bissell MJ, Hines WC. Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med 2011; 17:320 - 9; http://dx.doi.org/10.1038/nm.2328; PMID: 21383745
  • Egeblad M, Nakasone ES, Werb Z. Tumors as organs: complex tissues that interface with the entire organism. Dev Cell 2010; 18:884 - 901; http://dx.doi.org/10.1016/j.devcel.2010.05.012; PMID: 20627072
  • Arendt LM, Rudnick JA, Keller PJ, Kuperwasser C. Stroma in breast development and disease. Semin Cell Dev Biol 2010; 21:11 - 8; http://dx.doi.org/10.1016/j.semcdb.2009.10.003; PMID: 19857593
  • Bhowmick NA, Neilson EG, Moses HL. Stromal fibroblasts in cancer initiation and progression. Nature 2004; 432:332 - 7; http://dx.doi.org/10.1038/nature03096; PMID: 15549095
  • Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S, et al. TGF-beta signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science 2004; 303:848 - 51; http://dx.doi.org/10.1126/science.1090922; PMID: 14764882
  • Liu S, Ginestier C, Ou SJ, Clouthier SG, Patel SH, Monville F, et al. Breast cancer stem cells are regulated by mesenchymal stem cells through cytokine networks. Cancer Res 2011; 71:614 - 24; http://dx.doi.org/10.1158/0008-5472.CAN-10-0538; PMID: 21224357
  • McLean K, Gong Y, Choi Y, Deng N, Yang K, Bai S, et al. Human ovarian carcinoma–associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J Clin Invest 2011; 121:3206 - 19; http://dx.doi.org/10.1172/JCI45273; PMID: 21737876
  • Yan XL, Fu CJ, Chen L, Qin JH, Zeng Q, Yuan HF, et al. Mesenchymal stem cells from primary breast cancer tissue promote cancer proliferation and enhance mammosphere formation partially via EGF/EGFR/Akt pathway. Breast Cancer Res Treat 2012; 132:153 - 64; http://dx.doi.org/10.1007/s10549-011-1577-0; PMID: 21584665
  • Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 2000; 109:235 - 42; http://dx.doi.org/10.1046/j.1365-2141.2000.01986.x; PMID: 10848804
  • Young HE, Duplaa C, Young TM, Floyd JA, Reeves ML, Davis KH, et al. Clonogenic analysis reveals reserve stem cells in postnatal mammals: I. Pluripotent mesenchymal stem cells. Anat Rec 2001; 263:350 - 60; http://dx.doi.org/10.1002/ar.1112; PMID: 11500811
  • Young HE, Steele TA, Bray RA, Hudson J, Floyd JA, Hawkins K, et al. Human reserve pluripotent mesenchymal stem cells are present in the connective tissues of skeletal muscle and dermis derived from fetal, adult, and geriatric donors. Anat Rec 2001; 264:51 - 62; http://dx.doi.org/10.1002/ar.1128; PMID: 11505371
  • De Ugarte DA, Morizono K, Elbarbary A, Alfonso Z, Zuk PA, Zhu M, et al. Comparison of multi-lineage cells from human adipose tissue and bone marrow. Cells Tissues Organs 2003; 174:101 - 9; http://dx.doi.org/10.1159/000071150; PMID: 12835573
  • 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; http://dx.doi.org/10.1186/ar130; PMID: 11056678
  • 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; http://dx.doi.org/10.1182/blood.V98.8.2396; PMID: 11588036
  • 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; PMID: 15758422
  • Xia X, Chen W, Ma T, Xu G, Liu H, Liang C, et al. Mesenchymal stem cells administered after liver transplantation prevent acute graft-versus-host disease in rats. Liver Transpl 2012; 18:696 - 706; http://dx.doi.org/10.1002/lt.23414; PMID: 22344929
  • Lin Y, Hogan WJ. Clinical Application of Mesenchymal Stem Cells in the Treatment and Prevention of Graft-versus-Host Disease. Adv Hematol 2011; 2011:427863; http://dx.doi.org/10.1155/2011/427863; PMID: 22190941
  • Prockop DJ, Gregory CA, Spees JL. 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; http://dx.doi.org/10.1073/pnas.1834138100; PMID: 13679583
  • Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med 1986; 315:1650 - 9; PMID: 3537791
  • Studeny M, Marini FC, Dembinski JL, Zompetta C, Cabreira-Hansen M, Bekele BN, 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; http://dx.doi.org/10.1093/jnci/djh299; PMID: 15523088
  • Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, Bell GW, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 2007; 449:557 - 63; http://dx.doi.org/10.1038/nature06188; PMID: 17914389
  • Kidd S, Spaeth E, Dembinski JL, Dietrich M, Watson K, Klopp A, et al. Direct evidence of mesenchymal stem cell tropism for tumor and wounding microenvironments using in vivo bioluminescent imaging. Stem Cells 2009; 27:2614 - 23; http://dx.doi.org/10.1002/stem.187; PMID: 19650040
  • 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; http://dx.doi.org/10.1182/blood-2003-04-1193; PMID: 12881305
  • Djouad F, Bony C, Apparailly F, Louis-Plence P, Jorgensen C, Noël D. Earlier onset of syngeneic tumors in the presence of mesenchymal stem cells. Transplantation 2006; 82:1060 - 6; http://dx.doi.org/10.1097/01.tp.0000236098.13804.0b; PMID: 17060855
  • Zhu W, Xu W, Jiang R, Qian H, Chen M, Hu J, et al. Mesenchymal stem cells derived from bone marrow favor tumor cell growth in vivo. Exp Mol Pathol 2006; 80:267 - 74; http://dx.doi.org/10.1016/j.yexmp.2005.07.004; PMID: 16214129
  • Yu JM, Jun ES, Bae YC, Jung JS. Mesenchymal stem cells derived from human adipose tissues favor tumor cell growth in vivo. Stem Cells Dev 2008; 17:463 - 73; http://dx.doi.org/10.1089/scd.2007.0181; PMID: 18522494
  • Muehlberg FL, Song YH, Krohn A, Pinilla SP, Droll LH, Leng X, et al. Tissue-resident stem cells promote breast cancer growth and metastasis. Carcinogenesis 2009; 30:589 - 97; http://dx.doi.org/10.1093/carcin/bgp036; PMID: 19181699
  • Lin G, Yang R, Banie L, Wang G, Ning H, Li LC, et al. Effects of transplantation of adipose tissue-derived stem cells on prostate tumor. Prostate 2010; 70:1066 - 73; http://dx.doi.org/10.1002/pros.21140; PMID: 20232361
  • Kucerova L, Matuskova M, Hlubinova K, Altanerova V, Altaner C. Tumor cell behaviour modulation by mesenchymal stromal cells. Mol Cancer 2010; 9:129; http://dx.doi.org/10.1186/1476-4598-9-129; PMID: 20509882
  • Shinagawa K, Kitadai Y, Tanaka M, Sumida T, Kodama M, Higashi Y, et al. Mesenchymal stem cells enhance growth and metastasis of colon cancer. Int J Cancer 2010; 127:2323 - 33; http://dx.doi.org/10.1002/ijc.25440; PMID: 20473928
  • Prantl L, Muehlberg F, Navone NM, Song YH, Vykoukal J, Logothetis CJ, et al. Adipose tissue-derived stem cells promote prostate tumor growth. Prostate 2010; 70:1709 - 15; http://dx.doi.org/10.1002/pros.21206; PMID: 20564322
  • Suzuki K, Sun R, Origuchi M, Kanehira M, Takahata T, Itoh J, et al. Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization. Mol Med 2011; 17:579 - 87; http://dx.doi.org/10.2119/molmed.2010.00157; PMID: 21424106
  • Goldstein RH, Reagan MR, Anderson K, Kaplan DL, Rosenblatt M. Human bone marrow-derived MSCs can home to orthotopic breast cancer tumors and promote bone metastasis. Cancer Res 2010; 70:10044 - 50; http://dx.doi.org/10.1158/0008-5472.CAN-10-1254; PMID: 21159629
  • Tsukamoto S, Honoki K, Fujii H, Tohma Y, Kido A, Mori T, et al. Mesenchymal stem cells promote tumor engraftment and metastatic colonization in rat osteosarcoma model. Int J Oncol 2012; 40:163 - 9; PMID: 21971610
  • Koike N, Fukumura D, Gralla O, Au P, Schechner JS, Jain RK. Tissue engineering: creation of long-lasting blood vessels. Nature 2004; 428:138 - 9; http://dx.doi.org/10.1038/428138a; PMID: 15014486
  • Au P, Tam J, Fukumura D, Jain RK. Bone marrow-derived mesenchymal stem cells facilitate engineering of long-lasting functional vasculature. Blood 2008; 111:4551 - 8; http://dx.doi.org/10.1182/blood-2007-10-118273; PMID: 18256324
  • Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, Alexe G, Mesirov JP, et al. Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res 2008; 68:4331 - 9; http://dx.doi.org/10.1158/0008-5472.CAN-08-0943; PMID: 18519693
  • Quante M, Tu SP, Tomita H, Gonda T, Wang SS, Takashi S, et al. Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell 2011; 19:257 - 72; http://dx.doi.org/10.1016/j.ccr.2011.01.020; PMID: 21316604
  • Bianchi G, Borgonovo G, Pistoia V, Raffaghello L. Immunosuppressive cells and tumour microenvironment: focus on mesenchymal stem cells and myeloid derived suppressor cells. Histol Histopathol 2011; 26:941 - 51; PMID: 21630223
  • Friedenstein AJ, Gorskaja JF, Kulagina NN. Fibroblast precursors in normal and irradiated mouse hematopoietic organs. Exp Hematol 1976; 4:267 - 74; PMID: 976387
  • Prockop DJ. Marrow stromal cells as stem cells for nonhematopoietic tissues. Science 1997; 276:71 - 4; http://dx.doi.org/10.1126/science.276.5309.71; PMID: 9082988
  • Golde DW, Hocking WG, Quan SG, Sparkes RS, Gale RP. Origin of human bone marrow fibroblasts. Br J Haematol 1980; 44:183 - 7; http://dx.doi.org/10.1111/j.1365-2141.1980.tb01200.x; PMID: 6990960
  • Piersma AH, Ploemacher RE, Brockbank KG, Nikkels PG, Ottenheim CP. Migration of fibroblastoid stromal cells in murine blood. Cell Tissue Kinet 1985; 18:589 - 95; PMID: 4064102
  • Piersma AH, Brockbank KG, Ploemacher RE, Ottenheim CP. Recovery of hemopoietic stromal progenitor cells after lethal total-body irradiation and bone marrow transplantation in mice. Transplantation 1985; 40:198 - 201; http://dx.doi.org/10.1097/00007890-198508000-00018; PMID: 2862723
  • Piersma AH, Brockbank KG, Ploemacher RE, van Vliet E, Brakel-van Peer KM, Visser PJ. Characterization of fibroblastic stromal cells from murine bone marrow. Exp Hematol 1985; 13:237 - 43; PMID: 2580729
  • Mardon HJ, Bee J, von der Mark K, Owen ME. Development of osteogenic tissue in diffusion chambers from early precursor cells in bone marrow of adult rats. Cell Tissue Res 1987; 250:157 - 65; http://dx.doi.org/10.1007/BF00214667; PMID: 3652158
  • Beresford JN, Bennett JH, Devlin C, Leboy PS, Owen ME. Evidence for an inverse relationship between the differentiation of adipocytic and osteogenic cells in rat marrow stromal cell cultures. J Cell Sci 1992; 102:341 - 51; PMID: 1400636
  • Bruder SP, Fink DJ, Caplan AI. Mesenchymal stem cells in bone development, bone repair, and skeletal regeneration therapy. J Cell Biochem 1994; 56:283 - 94; http://dx.doi.org/10.1002/jcb.240560303; PMID: 7876320
  • 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; http://dx.doi.org/10.1126/science.284.5411.143; PMID: 10102814
  • Delorme B, Charbord P. Culture and characterization of human bone marrow mesenchymal stem cells. Methods Mol Med 2007; 140:67 - 81; http://dx.doi.org/10.1007/978-1-59745-443-8_4; PMID: 18085203
  • Horwitz EM, Le Blanc K, Dominici M, Mueller I, Slaper-Cortenbach I, Marini FC, et al, International Society for Cellular Therapy. Clarification of the nomenclature for MSC: The International Society for Cellular Therapy position statement. Cytotherapy 2005; 7:393 - 5; http://dx.doi.org/10.1080/14653240500319234; PMID: 16236628
  • Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006; 8:315 - 7; http://dx.doi.org/10.1080/14653240600855905; PMID: 16923606
  • 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; http://dx.doi.org/10.1172/JCI5298; PMID: 10074487
  • Spees JL, Olson SD, Ylostalo J, Lynch PJ, Smith J, Perry A, et al. Differentiation, cell fusion, and nuclear fusion during ex vivo repair of epithelium by human adult stem cells from bone marrow stroma. Proc Natl Acad Sci U S A 2003; 100:2397 - 402; http://dx.doi.org/10.1073/pnas.0437997100; PMID: 12606728
  • Phinney DG, Isakova I. Plasticity and therapeutic potential of mesenchymal stem cells in the nervous system. Curr Pharm Des 2005; 11:1255 - 65; http://dx.doi.org/10.2174/1381612053507495; PMID: 15853682
  • Tropel P, Platet N, Platel JC, Noël D, Albrieux M, Benabid AL, et al. Functional neuronal differentiation of bone marrow-derived mesenchymal stem cells. Stem Cells 2006; 24:2868 - 76; http://dx.doi.org/10.1634/stemcells.2005-0636; PMID: 16902198
  • Roobrouck VD, Ulloa-Montoya F, Verfaillie CM. Self-renewal and differentiation capacity of young and aged stem cells. Exp Cell Res 2008; 314:1937 - 44; http://dx.doi.org/10.1016/j.yexcr.2008.03.006; PMID: 18439579
  • Tsai MS, Lee JL, Chang YJ, Hwang SM. Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two-stage culture protocol. Hum Reprod 2004; 19:1450 - 6; http://dx.doi.org/10.1093/humrep/deh279; PMID: 15105397
  • Djouad F, Bony C, Häupl T, Uzé G, Lahlou N, Louis-Plence P, et al. Transcriptional profiles discriminate bone marrow-derived and synovium-derived mesenchymal stem cells. Arthritis Res Ther 2005; 7:R1304 - 15; http://dx.doi.org/10.1186/ar1827; PMID: 16277684
  • Jones EA, Crawford A, English A, Henshaw K, Mundy J, Corscadden D, et al. Synovial fluid mesenchymal stem cells in health and early osteoarthritis: detection and functional evaluation at the single-cell level. Arthritis Rheum 2008; 58:1731 - 40; http://dx.doi.org/10.1002/art.23485; PMID: 18512779
  • Tomar GB, Srivastava RK, Gupta N, Barhanpurkar AP, Pote ST, Jhaveri HM, et al. Human gingiva-derived mesenchymal stem cells are superior to bone marrow-derived mesenchymal stem cells for cell therapy in regenerative medicine. Biochem Biophys Res Commun 2010; 393:377 - 83; http://dx.doi.org/10.1016/j.bbrc.2010.01.126; PMID: 20138833
  • Greco SJ, Liu K, Rameshwar P. Functional similarities among genes regulated by OCT4 in human mesenchymal and embryonic stem cells. Stem Cells 2007; 25:3143 - 54; http://dx.doi.org/10.1634/stemcells.2007-0351; PMID: 17761754
  • Riekstina U, Cakstina I, Parfejevs V, Hoogduijn M, Jankovskis G, Muiznieks I, et al. Embryonic stem cell marker expression pattern in human mesenchymal stem cells derived from bone marrow, adipose tissue, heart and dermis. Stem Cell Rev 2009; 5:378 - 86; http://dx.doi.org/10.1007/s12015-009-9094-9; PMID: 20058201
  • Pierantozzi E, Gava B, Manini I, Roviello F, Marotta G, Chiavarelli M, et al. Pluripotency regulators in human mesenchymal stem cells: expression of NANOG but not of OCT-4 and SOX-2. Stem Cells Dev 2011; 20:915 - 23; http://dx.doi.org/10.1089/scd.2010.0353; PMID: 20879854
  • Choumerianou DM, Martimianaki G, Stiakaki E, Kalmanti L, Kalmanti M, Dimitriou H. Comparative study of stemness characteristics of mesenchymal cells from bone marrow of children and adults. Cytotherapy 2010; 12:881 - 7; http://dx.doi.org/10.3109/14653249.2010.501790; PMID: 20662612
  • Méndez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010; 466:829 - 34; http://dx.doi.org/10.1038/nature09262; PMID: 20703299
  • Pontikoglou C, Deschaseaux F, Sensebé L, Papadaki HA. Bone marrow mesenchymal stem cells: biological properties and their role in hematopoiesis and hematopoietic stem cell transplantation. Stem Cell Rev 2011; 7:569 - 89; http://dx.doi.org/10.1007/s12015-011-9228-8; PMID: 21249477
  • Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008; 3:301 - 13; http://dx.doi.org/10.1016/j.stem.2008.07.003; PMID: 18786417
  • Guan M, Yao W, Liu R, Lam KS, Nolta J, Jia J, et al. Directing mesenchymal stem cells to bone to augment bone formation and increase bone mass. Nat Med 2012; 18:456 - 62; http://dx.doi.org/10.1038/nm.2665; PMID: 22306732
  • Hocking AM, Gibran NS. Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair. Exp Cell Res 2010; 316:2213 - 9; http://dx.doi.org/10.1016/j.yexcr.2010.05.009; PMID: 20471978
  • Mansilla E, Marín GH, Drago H, Sturla F, Salas E, Gardiner C, et al. Bloodstream cells phenotypically identical to human mesenchymal bone marrow stem cells circulate in large amounts under the influence of acute large skin damage: new evidence for their use in regenerative medicine. Transplant Proc 2006; 38:967 - 9; http://dx.doi.org/10.1016/j.transproceed.2006.02.053; PMID: 16647520
  • Hatzistergos KE, Quevedo H, Oskouei BN, Hu Q, Feigenbaum GS, Margitich IS, et al. Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circ Res 2010; 107:913 - 22; http://dx.doi.org/10.1161/CIRCRESAHA.110.222703; PMID: 20671238
  • Di Ianni M, Del Papa B, De Ioanni M, Moretti L, Bonifacio E, Cecchini D, et al. Mesenchymal cells recruit and regulate T regulatory cells. Exp Hematol 2008; 36:309 - 18; http://dx.doi.org/10.1016/j.exphem.2007.11.007; PMID: 18279718
  • Corcione A, Benvenuto F, Ferretti E, Giunti D, Cappiello V, Cazzanti F, et al. Human mesenchymal stem cells modulate B-cell functions. Blood 2006; 107:367 - 72; http://dx.doi.org/10.1182/blood-2005-07-2657; PMID: 16141348
  • Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R, Fibbe WE. Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells. J Immunol 2006; 177:2080 - 7; PMID: 16887966
  • Djouad F, Charbonnier LM, Bouffi C, Louis-Plence P, Bony C, Apparailly F, et al. Mesenchymal stem cells inhibit the differentiation of dendritic cells through an interleukin-6-dependent mechanism. Stem Cells 2007; 25:2025 - 32; http://dx.doi.org/10.1634/stemcells.2006-0548; PMID: 17510220
  • Prockop DJ, Oh JY. Mesenchymal stem/stromal cells (MSCs): role as guardians of inflammation. Mol Ther 2012; 20:14 - 20; http://dx.doi.org/10.1038/mt.2011.211; PMID: 22008910
  • Prockop DJ, Oh JY. Medical therapies with adult stem/progenitor cells (MSCs): a backward journey from dramatic results in vivo to the cellular and molecular explanations. J Cell Biochem 2012; 113:1460 - 9; PMID: 22213121
  • Prockop DJ. “Stemness” does not explain the repair of many tissues by mesenchymal stem/multipotent stromal cells (MSCs). Clin Pharmacol Ther 2007; 82:241 - 3; http://dx.doi.org/10.1038/sj.clpt.6100313; PMID: 17700588
  • Jung Y, Bauer G, Nolta JA. Concise review: Induced pluripotent stem cell-derived mesenchymal stem cells: progress toward safe clinical products. Stem Cells 2012; 30:42 - 7; http://dx.doi.org/10.1002/stem.727; PMID: 21898694
  • Gao C, Seuntjens J, Kaufman GN, Tran-Khanh N, Butler A, Li A, et al. Mesenchymal stem cell transplantation to promote bone healing. J Orthop Res 2012; 30:1183 - 9; http://dx.doi.org/10.1002/jor.22028; PMID: 22228593
  • 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; http://dx.doi.org/10.1038/6529; PMID: 10086387
  • Li XH, Fu YH, Lin QX, Liu ZY, Shan ZX, Deng CY, et al. Induced bone marrow mesenchymal stem cells improve cardiac performance of infarcted rat hearts. Mol Biol Rep 2012; 39:1333 - 42; http://dx.doi.org/10.1007/s11033-011-0867-2; PMID: 21667244
  • Simpson DL, Boyd NL, Kaushal S, Stice SL, Dudley SC Jr. Use of human embryonic stem cell derived-mesenchymal cells for cardiac repair. Biotechnol Bioeng 2012; 109:274 - 83; http://dx.doi.org/10.1002/bit.23301; PMID: 21837664
  • Sakai Y, Kaneko S. Mesenchymal stem cell therapy on murine model of nonalcoholic steatohepatitis. Methods Mol Biol 2012; 826:217 - 23; http://dx.doi.org/10.1007/978-1-61779-468-1_17; PMID: 22167651
  • Wise AF, Ricardo SD. Mesenchymal stem cells in kidney inflammation and repair. Nephrology (Carlton) 2012; 17:1 - 10; http://dx.doi.org/10.1111/j.1440-1797.2011.01501.x; PMID: 21777348
  • Nakajima H, Uchida K, Guerrero AR, Watanabe S, Sugita D, Takeura N, et al. Transplantation of mesenchymal stem cells promotes an alternative pathway of macrophage activation and functional recovery after spinal cord injury. J Neurotrauma 2012; 29:1614 - 25; http://dx.doi.org/10.1089/neu.2011.2109; PMID: 22233298
  • Roemeling-van Rhijn M, Weimar W, Hoogduijn MJ. Mesenchymal stem cells: application for solid-organ transplantation. Curr Opin Organ Transplant 2012; 17:55 - 62; http://dx.doi.org/10.1097/MOT.0b013e32834ee676; PMID: 22186092
  • Mazzini L, Mareschi K, Ferrero I, Miglioretti M, Stecco A, Servo S, et al. Mesenchymal stromal cell transplantation in amyotrophic lateral sclerosis: a long-term safety study. Cytotherapy 2012; 14:56 - 60; http://dx.doi.org/10.3109/14653249.2011.613929; PMID: 21954839
  • Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, et al. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study. Lancet Neurol 2012; 11:150 - 6; http://dx.doi.org/10.1016/S1474-4422(11)70305-2; PMID: 22236384
  • Spaeth E, Klopp A, Dembinski J, Andreeff M, Marini F. Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene Ther 2008; 15:730 - 8; http://dx.doi.org/10.1038/gt.2008.39; PMID: 18401438
  • Ren C, Kumar S, Chanda D, Chen J, Mountz JD, Ponnazhagan S. Therapeutic potential of mesenchymal stem cells producing interferon-alpha in a mouse melanoma lung metastasis model. Stem Cells 2008; 26:2332 - 8; http://dx.doi.org/10.1634/stemcells.2008-0084; PMID: 18617688
  • Komarova S, Kawakami Y, Stoff-Khalili MA, Curiel DT, Pereboeva L. Mesenchymal progenitor cells as cellular vehicles for delivery of oncolytic adenoviruses. Mol Cancer Ther 2006; 5:755 - 66; http://dx.doi.org/10.1158/1535-7163.MCT-05-0334; PMID: 16546991
  • Niess H, Bao Q, Conrad C, Zischek C, Notohamiprodjo M, Schwab F, et al. Selective targeting of genetically engineered mesenchymal stem cells to tumor stroma microenvironments using tissue-specific suicide gene expression suppresses growth of hepatocellular carcinoma. Ann Surg 2011; 254:767 - 74, discussion 774-5; http://dx.doi.org/10.1097/SLA.0b013e3182368c4f; PMID: 22042469
  • Kidd S, Spaeth E, Watson K, Burks J, Lu H, Klopp A, et al. Origins of the tumor microenvironment: quantitative assessment of adipose-derived and bone marrow-derived stroma. PLoS One 2012; 7:e30563; http://dx.doi.org/10.1371/journal.pone.0030563; PMID: 22363446
  • Springer TA. Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm. Cell 1994; 76:301 - 14; http://dx.doi.org/10.1016/0092-8674(94)90337-9; PMID: 7507411
  • Chute JP. Stem cell homing. Curr Opin Hematol 2006; 13:399 - 406; http://dx.doi.org/10.1097/01.moh.0000245698.62511.3d; PMID: 17053451
  • Dwyer RM, Potter-Beirne SM, Harrington KA, Lowery AJ, Hennessy E, Murphy JM, et al. Monocyte chemotactic protein-1 secreted by primary breast tumors stimulates migration of mesenchymal stem cells. Clin Cancer Res 2007; 13:5020 - 7; http://dx.doi.org/10.1158/1078-0432.CCR-07-0731; PMID: 17785552
  • Bhoopathi P, Chetty C, Gogineni VR, Gujrati M, Dinh DH, Rao JS, et al. MMP-2 mediates mesenchymal stem cell tropism towards medulloblastoma tumors. Gene Ther 2011; 18:692 - 701; http://dx.doi.org/10.1038/gt.2011.14; PMID: 21368903
  • Lin SY, Yang J, Everett AD, Clevenger CV, Koneru M, Mishra PJ, et al. The isolation of novel mesenchymal stromal cell chemotactic factors from the conditioned medium of tumor cells. Exp Cell Res 2008; 314:3107 - 17; http://dx.doi.org/10.1016/j.yexcr.2008.07.028; PMID: 18722367
  • Gutova M, Najbauer J, Frank RT, Kendall SE, Gevorgyan A, Metz MZ, et al. Urokinase plasminogen activator and urokinase plasminogen activator receptor mediate human stem cell tropism to malignant solid tumors. Stem Cells 2008; 26:1406 - 13; http://dx.doi.org/10.1634/stemcells.2008-0141; PMID: 18403751
  • Pulukuri SM, Gorantla B, Dasari VR, Gondi CS, Rao JS. Epigenetic upregulation of urokinase plasminogen activator promotes the tropism of mesenchymal stem cells for tumor cells. Mol Cancer Res 2010; 8:1074 - 83; http://dx.doi.org/10.1158/1541-7786.MCR-09-0495; PMID: 20663859
  • Rattigan Y, Hsu JM, Mishra PJ, Glod J, Banerjee D. Interleukin 6 mediated recruitment of mesenchymal stem cells to the hypoxic tumor milieu. Exp Cell Res 2010; 316:3417 - 24; http://dx.doi.org/10.1016/j.yexcr.2010.07.002; PMID: 20633553
  • Ritter E, Perry A, Yu J, Wang T, Tang L, Bieberich E. Breast cancer cell-derived fibroblast growth factor 2 and vascular endothelial growth factor are chemoattractants for bone marrow stromal stem cells. Ann Surg 2008; 247:310 - 4; http://dx.doi.org/10.1097/SLA.0b013e31816401d5; PMID: 18216538
  • Schmidt A, Ladage D, Schinköthe T, Klausmann U, Ulrichs C, Klinz FJ, et al. Basic fibroblast growth factor controls migration in human mesenchymal stem cells. Stem Cells 2006; 24:1750 - 8; http://dx.doi.org/10.1634/stemcells.2005-0191; PMID: 16822883
  • Ball SG, Shuttleworth CA, Kielty CM. Vascular endothelial growth factor can signal through platelet-derived growth factor receptors. J Cell Biol 2007; 177:489 - 500; http://dx.doi.org/10.1083/jcb.200608093; PMID: 17470632
  • Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, et al. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med 2009; 15:757 - 65; http://dx.doi.org/10.1038/nm.1979; PMID: 19584867
  • Ponte AL, Marais E, Gallay N, Langonné A, Delorme B, Hérault O, et al. The in vitro migration capacity of human bone marrow mesenchymal stem cells: comparison of chemokine and growth factor chemotactic activities. Stem Cells 2007; 25:1737 - 45; http://dx.doi.org/10.1634/stemcells.2007-0054; PMID: 17395768
  • Coffelt SB, Marini FC, Watson K, Zwezdaryk KJ, Dembinski JL, LaMarca HL, et al. The pro-inflammatory peptide LL-37 promotes ovarian tumor progression through recruitment of multipotent mesenchymal stromal cells. Proc Natl Acad Sci U S A 2009; 106:3806 - 11; http://dx.doi.org/10.1073/pnas.0900244106; PMID: 19234121
  • Schraufstatter IU, Discipio RG, Zhao M, Khaldoyanidi SK. C3a and C5a are chemotactic factors for human mesenchymal stem cells, which cause prolonged ERK1/2 phosphorylation. J Immunol 2009; 182:3827 - 36; http://dx.doi.org/10.4049/jimmunol.0803055; PMID: 19265162
  • Schraufstatter IU, DiScipio RG, Khaldoyanidi SK. Alpha 7 subunit of nAChR regulates migration of human mesenchymal stem cells. J Stem Cells 2009; 4:203 - 15; PMID: 20720594
  • Shome S, Dasgupta PS, Basu S. Dopamine regulates mobilization of mesenchymal stem cells during wound angiogenesis. PLoS One 2012; 7:e31682; http://dx.doi.org/10.1371/journal.pone.0031682; PMID: 22355389
  • Spaeth EL, Marini FC. Dissecting mesenchymal stem cell movement: migration assays for tracing and deducing cell migration. Methods Mol Biol 2011; 750:241 - 59; http://dx.doi.org/10.1007/978-1-61779-145-1_17; PMID: 21618096
  • Zhang Y, Daquinag A, Traktuev DO, Amaya-Manzanares F, Simmons PJ, March KL, et al. White adipose tissue cells are recruited by experimental tumors and promote cancer progression in mouse models. Cancer Res 2009; 69:5259 - 66; http://dx.doi.org/10.1158/0008-5472.CAN-08-3444; PMID: 19491274
  • Henschler R, Deak E, Seifried E. Homing of Mesenchymal Stem Cells. Transfus Med Hemother 2008; 35:306 - 12; http://dx.doi.org/10.1159/000143110; PMID: 21512647
  • Ciuculescu F, Giesen M, Deak E, Lang V, Seifried E, Henschler R. Variability in chemokine-induced adhesion of human mesenchymal stromal cells. Cytotherapy 2011; 13:1172 - 9; http://dx.doi.org/10.3109/14653249.2011.602339; PMID: 21867464
  • Rüster B, Göttig S, Ludwig RJ, Bistrian R, Müller S, Seifried E, et al. Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells. Blood 2006; 108:3938 - 44; http://dx.doi.org/10.1182/blood-2006-05-025098; PMID: 16896152
  • Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, Moorman MA, McIntosh KR, et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci 2003; 10:228 - 41; http://dx.doi.org/10.1007/BF02256058; PMID: 12595759
  • Zhang H, Ning H, Banie L, Wang G, Lin G, Lue TF, et al. Adipose tissue-derived stem cells secrete CXCL5 cytokine with chemoattractant and angiogenic properties. Biochem Biophys Res Commun 2010; 402:560 - 4; http://dx.doi.org/10.1016/j.bbrc.2010.10.090; PMID: 21034724
  • Chamberlain G, Smith H, Rainger GE, Middleton J. Mesenchymal stem cells exhibit firm adhesion, crawling, spreading and transmigration across aortic endothelial cells: effects of chemokines and shear. PLoS One 2011; 6:e25663; http://dx.doi.org/10.1371/journal.pone.0025663; PMID: 21980522
  • Khakoo AY, Pati S, Anderson SA, Reid W, Elshal MF, Rovira II, et al. Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi’s sarcoma. J Exp Med 2006; 203:1235 - 47; http://dx.doi.org/10.1084/jem.20051921; PMID: 16636132
  • Corre J, Labat E, Espagnolle N, Hébraud B, Avet-Loiseau H, Roussel M, et al. Bioactivity and prognostic significance of growth differentiation factor GDF15 secreted by bone marrow mesenchymal stem cells in multiple myeloma. Cancer Res 2012; 72:1395 - 406; http://dx.doi.org/10.1158/0008-5472.CAN-11-0188; PMID: 22301101
  • Xu S, Menu E, De Becker A, Van Camp B, Vanderkerken K, Van Riet I. Bone marrow-derived mesenchymal stromal cells are attracted by multiple myeloma cell-produced chemokine CCL25 and favor myeloma cell growth in vitro and in vivo. Stem Cells 2012; 30:266 - 79; http://dx.doi.org/10.1002/stem.787; PMID: 22102554
  • Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 2010; 464:852 - 7; http://dx.doi.org/10.1038/nature08851; PMID: 20305640
  • Rhodes LV, Antoon JW, Muir SE, Elliott S, Beckman BS, Burow ME. Effects of human mesenchymal stem cells on ER-positive human breast carcinoma cells mediated through ER-SDF-1/CXCR4 crosstalk. Mol Cancer 2010; 9:295; http://dx.doi.org/10.1186/1476-4598-9-295; PMID: 21087507
  • Halpern JL, Kilbarger A, Lynch CC. Mesenchymal stem cells promote mammary cancer cell migration in vitro via the CXCR2 receptor. Cancer Lett 2011; 308:91 - 9; http://dx.doi.org/10.1016/j.canlet.2011.04.018; PMID: 21601983
  • Tsai KS, Yang SH, Lei YP, Tsai CC, Chen HW, Hsu CY, et al. Mesenchymal stem cells promote formation of colorectal tumors in mice. Gastroenterology 2011; 141:1046 - 56; http://dx.doi.org/10.1053/j.gastro.2011.05.045; PMID: 21699785
  • Collino F, Deregibus MC, Bruno S, Sterpone L, Aghemo G, Viltono L, et al. Microvesicles derived from adult human bone marrow and tissue specific mesenchymal stem cells shuttle selected pattern of miRNAs. PLoS One 2010; 5:e11803; http://dx.doi.org/10.1371/journal.pone.0011803; PMID: 20668554
  • Lim PK, Bliss SA, Patel SA, Taborga M, Dave MA, Gregory LA, et al. Gap junction-mediated import of microRNA from bone marrow stromal cells can elicit cell cycle quiescence in breast cancer cells. Cancer Res 2011; 71:1550 - 60; http://dx.doi.org/10.1158/0008-5472.CAN-10-2372; PMID: 21343399
  • Ohkouchi S, Block GJ, Katsha AM, Kanehira M, Ebina M, Kikuchi T, et al. Mesenchymal stromal cells protect cancer cells from ROS-induced apoptosis and enhance the Warburg effect by secreting STC1. Mol Ther 2012; 20:417 - 23; http://dx.doi.org/10.1038/mt.2011.259; PMID: 22146344
  • Liu Y, Han ZP, Zhang SS, Jing YY, Bu XX, Wang CY, et al. Effects of inflammatory factors on mesenchymal stem cells and their role in the promotion of tumor angiogenesis in colon cancer. J Biol Chem 2011; 286:25007 - 15; http://dx.doi.org/10.1074/jbc.M110.213108; PMID: 21592963
  • Shin SY, Nam JS, Lim Y, Lee YH. TNFα-exposed bone marrow-derived mesenchymal stem cells promote locomotion of MDA-MB-231 breast cancer cells through transcriptional activation of CXCR3 ligand chemokines. J Biol Chem 2010; 285:30731 - 40; http://dx.doi.org/10.1074/jbc.M110.128124; PMID: 20650898
  • Martin FT, Dwyer RM, Kelly J, Khan S, Murphy JM, Curran C, et al. Potential role of mesenchymal stem cells (MSCs) in the breast tumour microenvironment: stimulation of epithelial to mesenchymal transition (EMT). Breast Cancer Res Treat 2010; 124:317 - 26; http://dx.doi.org/10.1007/s10549-010-0734-1; PMID: 20087650
  • Albarenque SM, Zwacka RM, Mohr A. Both human and mouse mesenchymal stem cells promote breast cancer metastasis. Stem Cell Res 2011; 7:163 - 71; http://dx.doi.org/10.1016/j.scr.2011.05.002; PMID: 21763624
  • Nguyen LV, Vanner R, Dirks P, Eaves CJ. Cancer stem cells: an evolving concept. Nat Rev Cancer 2012; 12:133 - 43; PMID: 22237392
  • Roodhart JM, Daenen LG, Stigter EC, Prins HJ, Gerrits J, Houthuijzen JM, et al. Mesenchymal stem cells induce resistance to chemotherapy through the release of platinum-induced fatty acids. Cancer Cell 2011; 20:370 - 83; http://dx.doi.org/10.1016/j.ccr.2011.08.010; PMID: 21907927
  • Corcoran KE, Trzaska KA, Fernandes H, Bryan M, Taborga M, Srinivas V, et al. Mesenchymal stem cells in early entry of breast cancer into bone marrow. PLoS One 2008; 3:e2563; http://dx.doi.org/10.1371/journal.pone.0002563; PMID: 18575622
  • Coleman RE, Guise TA, Lipton A, Roodman GD, Berenson JR, Body JJ, et al. Advancing treatment for metastatic bone cancer: consensus recommendations from the Second Cambridge Conference. Clin Cancer Res 2008; 14:6387 - 95; http://dx.doi.org/10.1158/1078-0432.CCR-08-1572; PMID: 18927277
  • Zhao S, Wehner R, Bornhäuser M, Wassmuth R, Bachmann M, Schmitz M. Immunomodulatory properties of mesenchymal stromal cells and their therapeutic consequences for immune-mediated disorders. Stem Cells Dev 2010; 19:607 - 14; http://dx.doi.org/10.1089/scd.2009.0345; PMID: 19824807
  • Le Blanc K, Rasmusson I, Sundberg B, Götherström 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; http://dx.doi.org/10.1016/S0140-6736(04)16104-7; PMID: 15121408
  • Ball L, Bredius R, Lankester A, Schweizer J, van den Heuvel-Eibrink M, Escher H, et al. Third party mesenchymal stromal cell infusions fail to induce tissue repair despite successful control of severe grade IV acute graft-versus-host disease in a child with juvenile myelo-monocytic leukemia. Leukemia 2008; 22:1256 - 7; http://dx.doi.org/10.1038/sj.leu.2405013; PMID: 17972946
  • Ringden O, Le Blanc K. Mesenchymal stem cells for treatment of acute and chronic graft-versus-host disease, tissue toxicity and hemorrhages. Best Pract Res Clin Haematol 2011; 24:65 - 72; http://dx.doi.org/10.1016/j.beha.2011.01.003; PMID: 21396594
  • Krampera M, Cosmi L, Angeli R, Pasini A, Liotta F, Andreini A, et al. Role for interferon-gamma in the immunomodulatory activity of human bone marrow mesenchymal stem cells. Stem Cells 2006; 24:386 - 98; http://dx.doi.org/10.1634/stemcells.2005-0008; PMID: 16123384
  • Spaggiari GM, Capobianco A, Abdelrazik H, Becchetti F, Mingari MC, Moretta L. Mesenchymal stem cells inhibit natural killer-cell proliferation, cytotoxicity, and cytokine production: role of indoleamine 2,3-dioxygenase and prostaglandin E2. Blood 2008; 111:1327 - 33; http://dx.doi.org/10.1182/blood-2007-02-074997; PMID: 17951526
  • Prigione I, Benvenuto F, Bocca P, Battistini L, Uccelli A, Pistoia V. Reciprocal interactions between human mesenchymal stem cells and gammadelta T cells or invariant natural killer T cells. Stem Cells 2009; 27:693 - 702; http://dx.doi.org/10.1634/stemcells.2008-0687; PMID: 19096038
  • Raffaghello L, Bianchi G, Bertolotto M, Montecucco F, Busca A, Dallegri F, et al. Human mesenchymal stem cells inhibit neutrophil apoptosis: a model for neutrophil preservation in the bone marrow niche. Stem Cells 2008; 26:151 - 62; http://dx.doi.org/10.1634/stemcells.2007-0416; PMID: 17932421
  • Glennie S, Soeiro I, Dyson PJ, Lam EW, Dazzi F. Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood 2005; 105:2821 - 7; http://dx.doi.org/10.1182/blood-2004-09-3696; PMID: 15591115
  • 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; http://dx.doi.org/10.1182/blood.V99.10.3838; PMID: 11986244
  • Krampera M, Glennie S, Dyson J, Scott D, Laylor R, Simpson E, et al. Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood 2003; 101:3722 - 9; http://dx.doi.org/10.1182/blood-2002-07-2104; PMID: 12506037
  • Liotta F, Angeli R, Cosmi L, Filì L, Manuelli C, Frosali F, et al. Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing Notch signaling. Stem Cells 2008; 26:279 - 89; http://dx.doi.org/10.1634/stemcells.2007-0454; PMID: 17962701
  • Jiang XX, Zhang Y, Liu B, Zhang SX, Wu Y, Yu XD, et al. Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood 2005; 105:4120 - 6; http://dx.doi.org/10.1182/blood-2004-02-0586; PMID: 15692068
  • Ramasamy R, Fazekasova H, Lam EW, Soeiro I, Lombardi G, Dazzi F. Mesenchymal stem cells inhibit dendritic cell differentiation and function by preventing entry into the cell cycle. Transplantation 2007; 83:71 - 6; http://dx.doi.org/10.1097/01.tp.0000244572.24780.54; PMID: 17220794
  • Wehner R, Wehrum D, Bornhäuser M, Zhao S, Schäkel K, Bachmann MP, et al. Mesenchymal stem cells efficiently inhibit the proinflammatory properties of 6-sulfo LacNAc dendritic cells. Haematologica 2009; 94:1151 - 6; http://dx.doi.org/10.3324/haematol.2008.001735; PMID: 19546436
  • Ghannam S, Pène J, Torcy-Moquet G, Jorgensen C, Yssel H. Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J Immunol 2010; 185:302 - 12; http://dx.doi.org/10.4049/jimmunol.0902007; PMID: 20511548
  • Zhao ZG, Xu W, Sun L, You Y, Li F, Li QB, et al. Immunomodulatory function of regulatory dendritic cells induced by mesenchymal stem cells. Immunol Invest 2012; 41:183 - 98; http://dx.doi.org/10.3109/08820139.2011.607877; PMID: 21936678
  • Colombo MP, Piconese S. Regulatory-T-cell inhibition versus depletion: the right choice in cancer immunotherapy. Nat Rev Cancer 2007; 7:880 - 7; http://dx.doi.org/10.1038/nrc2250; PMID: 17957190
  • Patel SA, Meyer JR, Greco SJ, Corcoran KE, Bryan M, Rameshwar P. Mesenchymal stem cells protect breast cancer cells through regulatory T cells: role of mesenchymal stem cell-derived TGF-beta. J Immunol 2010; 184:5885 - 94; http://dx.doi.org/10.4049/jimmunol.0903143; PMID: 20382885
  • Weis SM, Cheresh DA. Tumor angiogenesis: molecular pathways and therapeutic targets. Nat Med 2011; 17:1359 - 70; http://dx.doi.org/10.1038/nm.2537; PMID: 22064426
  • Chen L, Tredget EE, Wu PY, Wu Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 2008; 3:e1886; http://dx.doi.org/10.1371/journal.pone.0001886; PMID: 18382669
  • Burns JS, Kristiansen M, Kristensen LP, Larsen KH, Nielsen MO, Christiansen H, et al. Decellularized matrix from tumorigenic human mesenchymal stem cells promotes neovascularization with galectin-1 dependent endothelial interaction. PLoS One 2011; 6:e21888; http://dx.doi.org/10.1371/journal.pone.0021888; PMID: 21779348
  • Oskowitz AZ, Penfornis P, Tucker A, Prockop DJ, Pochampally R. Drosha regulates hMSCs cell cycle progression through a miRNA independent mechanism. Int J Biochem Cell Biol 2011; 43:1563 - 72; http://dx.doi.org/10.1016/j.biocel.2011.07.005; PMID: 21794839
  • Oswald J, Boxberger S, Jørgensen B, Feldmann S, Ehninger G, Bornhäuser M, et al. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells 2004; 22:377 - 84; http://dx.doi.org/10.1634/stemcells.22-3-377; PMID: 15153614
  • 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; http://dx.doi.org/10.1161/01.CIR.0000151812.86142.45; PMID: 15642764
  • Medici D, Shore EM, Lounev VY, Kaplan FS, Kalluri R, Olsen BR. Conversion of vascular endothelial cells into multipotent stem-like cells. Nat Med 2010; 16:1400 - 6; http://dx.doi.org/10.1038/nm.2252; PMID: 21102460
  • Cho JA, Park H, Lim EH, Lee KW. Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int J Oncol 2012; 40:130 - 8; PMID: 21904773
  • Spaeth EL, Dembinski JL, Sasser AK, Watson K, Klopp A, Hall B, et al. Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression. PLoS One 2009; 4:e4992; http://dx.doi.org/10.1371/journal.pone.0004992; PMID: 19352430
  • Hayward SW, Wang Y, Cao M, Hom YK, Zhang B, Grossfeld GD, et al. Malignant transformation in a nontumorigenic human prostatic epithelial cell line. Cancer Res 2001; 61:8135 - 42; PMID: 11719442
  • Guo X, Oshima H, Kitmura T, Taketo MM, Oshima M. Stromal fibroblasts activated by tumor cells promote angiogenesis in mouse gastric cancer. J Biol Chem 2008; 283:19864 - 71; http://dx.doi.org/10.1074/jbc.M800798200; PMID: 18495668
  • Yazhou C, Wenlv S, Weidong Z, Licun W. Clinicopathological significance of stromal myofibroblasts in invasive ductal carcinoma of the breast. Tumour Biol 2004; 25:290 - 5; http://dx.doi.org/10.1159/000081394; PMID: 15627894
  • Tsujino T, Seshimo I, Yamamoto H, Ngan CY, Ezumi K, Takemasa I, et al. Stromal myofibroblasts predict disease recurrence for colorectal cancer. Clin Cancer Res 2007; 13:2082 - 90; http://dx.doi.org/10.1158/1078-0432.CCR-06-2191; PMID: 17404090
  • Cirri P, Chiarugi P. Cancer-associated-fibroblasts and tumour cells: a diabolic liaison driving cancer progression. Cancer Metastasis Rev 2012; 31:195 - 208; http://dx.doi.org/10.1007/s10555-011-9340-x; PMID: 22101652
  • Lin PP, Wang Y, Lozano G. Mesenchymal Stem Cells and the Origin of Ewing’s Sarcoma. Sarcoma 2011; 2011; http://dx.doi.org/10.1155/2011/276463; PMID: 20953407
  • Kauer M, Ban J, Kofler R, Walker B, Davis S, Meltzer P, et al. A molecular function map of Ewing’s sarcoma. PLoS One 2009; 4:e5415; http://dx.doi.org/10.1371/journal.pone.0005415; PMID: 19404404
  • Tirode F, Laud-Duval K, Prieur A, Delorme B, Charbord P, Delattre O. Mesenchymal stem cell features of Ewing tumors. Cancer Cell 2007; 11:421 - 9; http://dx.doi.org/10.1016/j.ccr.2007.02.027; PMID: 17482132
  • Suvà ML, Riggi N, Stehle JC, Baumer K, Tercier S, Joseph JM, et al. Identification of cancer stem cells in Ewing’s sarcoma. Cancer Res 2009; 69:1776 - 81; http://dx.doi.org/10.1158/0008-5472.CAN-08-2242; PMID: 19208848
  • Cironi L, Provero P, Riggi N, Janiszewska M, Suva D, Suva ML, et al. Epigenetic features of human mesenchymal stem cells determine their permissiveness for induction of relevant transcriptional changes by SYT-SSX1. PLoS One 2009; 4:e7904; http://dx.doi.org/10.1371/journal.pone.0007904; PMID: 19936258
  • Riggi N, Cironi L, Provero P, Suvà ML, Stehle JC, Baumer K, et al. Expression of the FUS-CHOP fusion protein in primary mesenchymal progenitor cells gives rise to a model of myxoid liposarcoma. Cancer Res 2006; 66:7016 - 23; http://dx.doi.org/10.1158/0008-5472.CAN-05-3979; PMID: 16849546
  • Houghton J, Stoicov C, Nomura S, Rogers AB, Carlson J, Li H, et al. Gastric cancer originating from bone marrow-derived cells. Science 2004; 306:1568 - 71; http://dx.doi.org/10.1126/science.1099513; PMID: 15567866

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.