BBACAN-87929; No. of pages: 15; 4C: 4, 6, 10, 11 Biochimica et Biophysica Acta xxx (2013) xxx–xxx

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta journal homepage: www.elsevier.com/locate/bbacan

1

Review

4Q1

Pedro Barcellos-de-Souza a,b,⁎, Valentina Gori c, Franco Bambi c, Paola Chiarugi a b c

O

a

Department of Experimental and Clinical Biomedical Sciences, University of Florence, Tuscany Tumor Institute and Center for Research, Transfer and High Education DenoTHE, Florence, Italy CAPES Foundation, Ministry of Education of Brazil, Brasília, DF, Brazil Azienda Ospedaliero Universitaria Meyer, Florence, Italy

R O

5 6 7 8

a r t i c l e

i n f o

a b s t r a c t

Article history: Received 23 June 2013 Received in revised form 15 October 2013 Accepted 18 October 2013 Available online xxxx

D

P

Tumor progression is a multistep phenomenon in which tumor-associated stromal cells perform an intricate cross-talk with tumor cells, supplying appropriate signals that may promote tumor aggressiveness. Among several cell types that constitute the tumor stroma, the discovery that bone marrow-derived mesenchymal stem cells (BM-MSC) have a strong tropism for tumors has achieved notoriety in recent years. Not only are the BMMSC recruited, but they can also engraft at tumor sites and transdifferentiate into cells such as activated fibroblasts, perivascular cells and macrophages, which will perform a key role in tumor progression. Whether the BM-MSC and their derived cells promote or suppress the tumor progression is a controversial issue. Recently, it has been proposed that proinflammatory stimuli can be decisive in driving BM-MSC polarization into cells with either tumor-supportive or tumor-repressive phenotypes (MSC1/MSC2). These considerations are extremely important both to an understanding of tumor biology and to the putative use of BM-MSC as “magic bullets” against tumors. In this review, we discuss the role of BM-MSC in many steps in tumor progression, focusing on the factors that attract BM-MSC to tumors, BM-MSC differentiation ability, the role of BM-MSC in tumor support or inhibition, the immunomodulation promoted by BM-MSC and metastatic niche formation by these cells. © 2013 The Authors. Published by Elsevier B.V. All rights reserved.

T

E

Keywords: Bone marrow-derived mesenchymal stem cells Tumor progression Tumor tropism Evasion from tumor site Cancer-associated fibroblasts Metastatic niche

C

9 10 11 12 13 14 16 15 17 18 19 20 21 22 23 24

R

Origin and characterization of BM-MSC . . . . . . . . . . . . . . Recruitment of BM-MSC to tumor sites . . . . . . . . . . . . . . Fate of BM-MSC in tumor microenvironments . . . . . . . . . . . 3.1. BM-MSC transdifferentiation into CAF-like cells . . . . . . . 3.2. Acquisition of a vascular-like phenotype by BM-MSC . . . . 3.3. Hematopoietic transdifferentiation by BM-MSC . . . . . . . 4. The role of BM-MSC in tumor growth . . . . . . . . . . . . . . . 4.1. Tumor progression evidence . . . . . . . . . . . . . . . 4.1.1. Role of BM-MSC-derived CAF in tumor progression . 4.1.2. BM-MSC-induced tumor neovascularization . . . . 4.2. Anti-tumor progression evidence . . . . . . . . . . . . . 5. BM-MSC and evasion from tumor sites . . . . . . . . . . . . . . 5.1. BM-MSC modulation of innate immunity . . . . . . . . . . 5.2. Adaptive immunity modulation by BM-MSC . . . . . . . . 6. BM-MSC and the metastatic niche . . . . . . . . . . . . . . . . 7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . Abbreviations . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

N C O

R

1. 2. 3.

U

45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63

Contents

25 26 27 28 29 30 31 32 33 34 35 36 37 38 40 39

E

42 41 44 43

F

3

Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players☆

2

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . .

0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0

64 ☆ This is an open-access article distributed under the terms of the Creative Commons Attribution-NonCommercial-No Derivative Works License, which permits non-commercial use, distribution, and reproduction in any medium, provided the original author and source are credited. ⁎ Corresponding author at: Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50-50134, Florence, Italy. Tel.: +39 055 2751247; fax: +39 055 2751201. E-mail address: [email protected] (P. Barcellos-de-Souza). 0304-419X/$ – see front matter © 2013 The Authors. Published by Elsevier B.V. All rights reserved. http://dx.doi.org/10.1016/j.bbcan.2013.10.004

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

It has been described that BM-MSC show a strong tropism towards injured tissues because the intravenous delivery of BM-MSC results in their migration to specific sites of injury [9]. Moreover, endogenous BM-MSC are mobilized in response to inflammation or injury, thus increasing their numbers in the bloodstream and targeting specific tissues via active mechanisms [10]. Tumors are often considered to be “wounds that never heal” because they continuously produce inflammatory mediators that maintain the

89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117

121 122 123 124 125 126 127

C

87 88

E

85 86

R

83 84

R

81 82

O

79 80

C

77 78

N

75 76

U

73 74

t1:4

Aminopeptidase N Integrin β1 chain Hyaluronan receptor Integrin α1-5 chain ICAM-1 Lymphocyte function-associated antigen 3 Integrin β3 chain Transferrin receptor Ecto-5′-nucleotidase Thy-1 Endoglin; TGFβRIII VCAM-1 PDGFR-β MCAM (MUC18) Raph blood group ALCAM p75; NGFR

t1:5 t1:6 t1:7 t1:8 t1:9 t1:10 t1:11 t1:12 t1:13 t1:14 t1:15 t1:16 t1:17 t1:18 t1:19 t1:20 t1:21 t1:22 t1:23

F

120

71 72

Synonym

CD13 [150,151] CD29 [150] CD44 [150,151] CD49 [152] CD54 [153,154] CD58 [154] CD61 [155] CD71 [153] CD73 [6] CD90 [6] CD105 [6] CD106 [151,154] CD140b [155,156] CD146 [154,156,157] CD151 [158] CD166 [153,154] CD271 [156,159] HLA-ABC [154] Stro-1 [160]

O

2. Recruitment of BM-MSC to tumor sites

70

Marker

R O

119

68 69

tumor microenvironment and assist with tumor progression [11]. In the last 10 years, it has been acknowledged that BM-MSC target to and engraft at sites of tumor development, which is a complex response that involves an intricate multistep process. The BM-MSC must be able to sense the signaling molecules released from the distant altered tissues, to move from their niche in the bone marrow into the circulation, and then to migrate into the target tissue. Once incorporated into the tumor microenvironment, the BM-MSC may differentiate into functional cells that participate in microenvironment homeostasis. It is likely that the selective tropism of BM-MSC to both tumors and sites of injury is due to the upregulation of similar inflammatory mediators in both circumstances. Many reports describe that BM-MSC can home and engraft to different types of solid tumors, including breast [12–15], lung [16,17], pancreatic [18], colon [19,20], ovarian [21–23] and prostate carcinomas [24,25], melanoma [26], glioma [27,28], Kaposi's sarcoma [29], and osteosarcoma [30] among other primary and metastatic tumors. The attraction of BM-MSC to tumor sites has been experimentally characterized by the injection by different routes of ex vivo-expanded BM-MSC and the subsequent cell-tracking utilizing a variety of labeling probes based on fluorescence, bioluminescence, positron emission, and magnetic resonance [31]. To support the biological role of endogenous BM-MSC migration to tumors, reports in which the bone marrow of sub-lethally irradiated SCID mice is repopulated with labeled bone marrow cells have concluded that these cells can efficiently home to the stroma of distally established tumors [32,33]. This idea highlights the concept that bone marrow-derived cells are significant components of the tumor stroma [21,32,34–36]. Due to their tropism to inflammatory sites, the chemotactic responses of BM-MSC are generally considered to resemble those of immune cells. Consistent with this, inflammatory cytokines are strongly involved in modulating the mobilization of the BM-MSC in the bone marrow niche and the further trafficking and homing of those cells to tumor sites. BM-MSC express a set of chemokine receptors, such as CCR1, -2, -4, -7 to -10, CXCR1, -2, -4 to -6, and CXC3 [37–40] and migrate in vitro in response to a number of molecules, e.g., CCL1 to -5, -7, -17, -19, -21 to -23, -25, -27, -28, CXCL-5, -8, -12, -13, -16 and CX3CL-1 (for details, see [41]). In addition to the chemokine receptors, it has also been described that BM-MSC express cell surface markers and receptors associated with migration such as growth hormone receptors, adhesion molecules and Toll-like receptors (TLR) [41]. These molecular mechanisms are similar to the leukocyte migratory apparatus, which

T

118

Recent years have witnessed numerous publications about mesenchymal stem cells (MSC). This is mainly due to the characteristics of these cells, particularly their ability to proliferate ex vivo, their potential for multilineage differentiation and their immunomodulatory properties. These distinctive features, together with an outstanding tropism for tumor sites, have highlighted MSC as putative genetically engineered vehicles that specifically deliver therapeutic agents into tumors. Nonetheless, an important role for both endogenous and exogenous MSC in tumor progression has been described. In addition to their tumor-homing ability, these cells can engraft and differentiate into tumor-associated stromal cells, ultimately leading to different consequences. MSC are widely distributed in a variety of adult tissues such as the umbilical cord, Wharton's Jelly, bone marrow, adipose tissue, peripheral blood and the lungs. In such tissues, MSC are either constantly present or their pool is replenished by the migration of bone marrow-derived MSC (BM-MSC) [1,2]. It has recently been demonstrated that MSC are also present in the placenta and fetal tissues [1]. After an ex vivo expansion, the MSC isolated from diverse tissues have similar properties and minor differences, which may be due to their microenvironment of origin. Nevertheless, they can exert different effects depending on their biological contexts [3]. The bone marrow is the most extensively studied source of MSC. Despite an invasive surgical procedure needed for BM-MSC recovery, a relatively low cell yield (0.001%–0.01%) is obtained, which is inversely correlated with the age of the donor [4]. Despite many limitations, the BM-MSC population is the gold standard for MSC clinical applications and in vitro experiments [5]. There is still a wide perception in the literature that MSC, particularly BM-MSC, represent a phenotypically heterogeneous population of cells; therefore, great confusion still exists among cell biologists regarding the identity of human BM-MSC. The International Society for Cell Therapy has proposed the following criteria for human BM-MSC characterization: (a) the adherence to plastic in standard culture conditions; (b) the expression of surface molecules such as CD73, CD90, and CD105 in the absence of CD34, CD45, HLA-DR, CD14 or CD11b, CD79a, or CD19, as assessed by fluorescence-activated cell sorter analysis; and (c) the capacity to differentiate into osteoblasts, adipocytes, and chondroblasts in vitro [6]. It has been shown that BM-MSC do not express endothelial markers such as CD31, CD34 and von Willebrand Factor (vWF). They express a large number of adhesion molecules (e.g., CD44 and integrins), some stromal cell markers (e.g., Src Homology-2, -3 and -4) and some cytokine receptors (e.g., Interleukin (IL)-1R and the tumor necrosis factor (TNF)-αR) [7]. Table 1 summarizes the currently used markers for BM-MSC characterization. Recent studies have suggested that MSC are capable of differentiating into endodermal and ectodermal lineages, including the lungs, retinal pigment, skin, sebaceous duct cells, renal tubular cells, neural cells, hepatocytes, and pancreatic islets [8]. Furthermore, in vitro functional assays are needed to demonstrate the multilineage differentiation of expanded cells, and a demonstration of pluripotency is necessary to discriminate between bona fide MSC and their precursors.

Table 1 t1:1 Markers expressed by BM-MSC. The principal markers that are used for the standard char- t1:2 acterization of BM-MSC are listed in the table. t1:3

P

66 67

1. Origin and characterization of BM-MSC

D

Q665 Q2

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

E

2

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235

272

After reaching the tumor site, BM-MSC engraft into the signalreleasing microenvironment and differentiate into functional stromal cells [31]. It is well established that BM-MSC can give rise to mesenchymal lineages such as chondrocytes, adipocytes and osteocytes [6]. They can also differentiate into myofibroblast-like cells, allowing the formation of connective tissue components and, ultimately, tissue repair [9,53]. Finally, the role of BM-MSC in the tumor stroma has become increasingly important because their ability to differentiate into carcinoma-associated fibroblasts (CAF) and other tumor-associated cells such as endothelial and pericyte-like cells, as well as the newly recognized macrophage-like cells [31,54] has been described.

273 274

3.1. BM-MSC transdifferentiation into CAF-like cells

284

It has been shown that engrafted bone marrow cells can develop into α-smooth muscle actin (SMA)-expressing myofibroblasts, usually recognized as CAF, in the tumor microenvironment [32,33]. In particular, BM-MSC co-cultured in vitro with cancer cells can be activated and may have a CAF-like phenotype. The markers used to define the CAF-like phenotype are divided into four groups: 1) the fibroblast activation markers, which include fibroblast specific protein (FSP, also named S100A4) and fibroblast activation protein (FAP); 2) the aggressiveness markers thrombospondin-1, tenascin-C and stromelysin; 3) the pro-vasculogenesis markers (desmin-1, α-SMA and VEGF-AA); and 4) the growth factors that support tumor growth and inflammation (EGF, HGF, IL-6, bFGF) [21,34–36].

285

O

F

3. Fate of BM-MSC in tumor microenvironments

R O

189 190

P

187 188

236 237

D

185 186

conditioning provides BM-MSC with higher migratory ability and increases the expression of c-Met, the major receptor for the HGF/scatter factor [48]. Moreover, the BM-MSC enhanced migration under conditions of hypoxia can be due to the increased expression of 3BP2/ SH3BP2 (Abl SH3-binding protein 2), an adaptor/scaffold protein that regulates leukocyte differentiation and motility, via HIF-mediated transcription. These data reinforce the resemblance between leukocyte and BM-MSC migratory machineries [47]. Furthermore, a hypoxic environment results in the generation of reactive oxygen species in the surrounding cells, which can increase genetic damage, leading to cell necrosis and tissue damage and consequently to inflammation [49]. Thus, hypoxia can maintain and amplify the inflammatory process in tumors, resulting in the production of chemokines and proteases that both attract immune cells and recruit BM-MSC. In solid tumors, cell death via the necrotic pathway can be a recurrent event, as malignant cells often show apoptosis resistance that is useful for enduring the impaired oxygen and nutrient supplies and the constant attack by immune cells. Therefore, the debris generated by cell necrosis is recognized by the immune cells through damageassociated molecular patterns (DAMP), thereby promoting inflammation and priming the adaptive immune responses. DAMP are a heterogeneous group of endogenous molecules, which include hyaluronan fragments, heat shock proteins, the S100 protein family, amyloid-β fibrils, uric acid, cytokines (IL-1α and IL-33) and the DNA-binding molecule high-mobility group box (HMGB)-1. Recent studies have linked DAMP with the chemoattraction of BM-MSC and have identified HMGB-1 as a crucial DAMP mediator [50,51]. When oxidized, HMGB-1 loses many of the functions typical of native HMGB-1, such as subcellular localization, interaction with DNA, and proinflammatory activities [52]. Interestingly, oxidized DAMP showed a significantly reduced induction of BM-MSC migration [51]. Although the direct oxidation of DAMP due to hypoxia has not been clearly reported, under the severe hypoxic conditions of a tumor microenvironment, it is likely that the DAMP are protected from oxidation and thereby they may act as important chemoattractants for BM-MSC.

E

183 184

T

181 182

C

179 180

E

177 178

R

176

R

174 175

N C O

172 173

suggests that BM-MSC migration occurs in a leukocyte-similar fashion. This similarity to leukocyte motility is also reported in a study that demonstrates the coordinated rolling and adhesion of BM-MSC on endothelial cells [42]. Although the signals released by tumors as well as the molecular mechanisms involved in the mobilization of BM-MSC are currently poorly understood, a number of studies using in vivo and in vitro approaches have identified some key players in the tumor microenvironment scenario, which range from cytokines to growth factors. Despite some conflicting results, which are probably the consequence of different experimental conditions and/or variability in the BM-MSC heterogeneity, a consistent panel of molecules involved in the promotion of BM-MSC migration towards different types of tumor is currently being identified (Fig. 1). Accordingly, inflammatory cytokines and chemokines released by tumors, including IL-6, CXCL8/IL-8, CCL2/MCP-1, CXCL1, CXCL2, CXCL12/SDF-1, transforming growth factor (TGF)-β and Neurotropin3, acknowledged to recruit leukocytes to sites of injury, have been implicated in regulating BM-MSC mobilization and migration to tumors (Fig. 1). Growth factors such as epidermal growth factor (EGF), hepatocyte growth factor (HGF), bFGF and platelet derived-growth factor (PDGF)-BB have been shown to induce BM-MSC migration, and the effects of these factors can be stronger when they are combined [41]. Recently, the post-transcriptional regulation of SDF-1 mRNA expression by the microRNA (miR) pair 126/126* was identified as a critical mechanism in the BM-MSC recruitment by breast cancer cells. The tissue sample analyses demonstrated that breast cancer patients, with a highly methylated T-2 promoter in the Egfl7 gene, express low levels of miR-126. As the SDF-1 mRNA is a target for the miR126/126* complex, this epigenetic modification allows high expression of SDF-1, which is responsible for the attraction of BM-MSC [15]. In both inflammatory and tumor sites, the activation of extracellular matrix (ECM) proteases, which degrade the ECM and allow cell migration into these locations, is a common event. BM-MSC migrate more efficiently to high aggressive tumors of different origins that show a higher expression of the urokinase plasminogen activator (uPA) and uPA receptor than to less aggressive tumors, evidencing uPA as a chemoattractant molecule. This mechanism may also involve the participation of high levels of proinflammatory cytokines, such as IL-6, IL-8 and MCP-1, which may be correlated with the uPA and soluble uPA receptor expression [16]. Other inflammatory cytokines (TGF-β, TNF-α, IL-1β) are able to increase metalloproteinase (MMP) activity in BMMSC, which is crucial for promoting cell migration [43]. In line with this, MMP-1 activity mediates BM-MSC tropism towards human glioma cells through the cleavage of the protease-activated receptor expressed on the BM-MSC surface [44]. Another similarity between inflammatory and tumor sites is the prompt angiogenic response that commonly occurs. Vascular endothelial growth factor (VEGF)-AA and IL-8 are involved in the recruitment of endothelial cells and in the coordination of an angiogenic process. Of note, these molecules also promote tumor homing for BM-MSC (Fig. 1). Certainly, these findings underscore the importance of the angiogenic scenario in the recruitment of BM-MSC towards tumor sites. As a tumor grows, its blood supply can be insufficient, thereby creating regions of low oxygen concentration that are characterized by variable hypoxia. A decrease in oxygen availability is frequent occurrence within tumor microenvironment, thus promoting the activation of hypoxia-inducible factors (HIF), a family of transcription factors that induce the transcription of several genes, such as VEGF, TNF-α and proinflammatory cytokines, which in turn are able to activate nuclear factor (NF)-κB [45]. NF-κB is a prominent proinflammatory factor that has a prominent role in inducing the transcription of a large number of chemokines and growth factors, including CCL2, -3, -5, CXCL2 and -8 that are implicated in BM-MSC migration. Indeed, BM-MSC migrate in vitro towards the hypoxic environment, mainly through the MT1MMP-1 axis modulated by HIF-1 [46,47]. In addition, hypoxic pre-

U

170 171

3

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271

275 276 277 278 279 280 281 282 283

286 287 288 289 290 291 292 293 294 295 296

4

N

C

Glioma Breast CCL-2 (MCP-1)13, IL-844,61, CXCL144, CXCL244, PDGF-BB27, EGF27, SDF-1α27, uPA16, SDF-115, IL-614,29 TGF-β161, Neurotropin61, VEGF28,61

O

R

R

Pancreatic PDGF17, EGF17, VEGF17

Osteosarcoma SDF-130

E

C

Ovarian LL-3723

T

E

Prostate uPA-uPAR axis16, CXCL1625

Lung uPA-uPAR axis16

D

CAF Resting fibroblast

Cancer cell

Invasive cancer cell

Macrophage Bone marrow derived cell

P

Lymphocyte/

R O

Endothelial cell

neutrophil Epithelial cell

Blood vessell

O

Fig. 1. Principal mediators identified in the BM-MSC tropism to tumors. BM-MSC recruitment to several tumors with different etiologies has been reported in the last 10 years. The main molecules involved in this phenomenon are described above.

F

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

U

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

361 362

3.3. Hematopoietic transdifferentiation by BM-MSC

373

311

3.2. Acquisition of a vascular-like phenotype by BM-MSC

374 375

312

The understanding of the importance BM-MSC in a tumor microenvironment is further extended by reports showing their transdifferentiation into endothelial-like cells, which suggests that they maintain their progenitor cell potential and, consequently, participate in functional vasculature structures. Hence, BM-MSC incorporated into tumor sites can transdifferentiate into cells with pro-vascular characteristics, e.g., endothelial cells and pericytes, although conflicting results still raise doubts about their true ability to adopt a classical endothelial phenotype. As mentioned above, the work by Kidd et al. is in agreement with other reports in which MSC isolated from adipose tissue were described to have undergone endothelial differentiation [55,56]. In regard to BMMSC, different investigations have reached different conclusions. It has been reported that murine bone marrow-derived cells expressing CD31+, an endothelial cell surface marker, were present within and around the cancer in a xenograft model of pancreatic cancer [32]. However, those data do not elucidate whether there was transdifferentiation of the BM-MSC or the engraftment of other bone marrow cells, such as multipotent progenitor cells. Annabi and collaborators reported that murine BM-MSC, which were formerly CD31−, contributed to the formation of highly vascularized tumors in vivo, and ultimately those cells differentiated into CD31+ cells [46]. In vitro results have shown that human BM-MSC cultured in the presence of VEGF could be differentiated into endothelial-like cells, expressing the endothelial-specific markers VEGFR-1, -2 and vWF, although they remained negative for CD31 or CD34 [57]. Moreover, culturing BM-MSC in endothelial cell growth supplements, and exposing those cells to shear force and extracellular matrix stimuli provided both phenotypic and functional endothelial-like characteristics to BM-MSC [58]. In models of melanoma and lung adenocarcinoma, after tumor engraftment, some BM-MSC were closely integrated into the tumor vasculature and expressed CD31+ but not α-SMA, which is often used as a pericyte marker, suggesting a direct support of the tumor vasculature, similar to an endothelial phenotype [17]. A different biological scenario, however, can lead to the prevalence of a pericyte-like differentiation rather than an endothelial-like one. Recent reports have identified that BM-MSC do not differentiate into endothelial cells in vitro; however, reactivity to endothelin-1 in vivo, which is a similarity with perivascular cell precursors, has been reported [18,59]. Rat BM-MSC integrated into tumor vessel walls expressed pericyte markers such as α-SMA, NG2, PDGFR-β, but not endothelial cell markers. The pericyte marker expression profile and the perivascular location of the engrafted BM-MSC indicate that these cells act as pericytes within tumors [60]. Human BM-MSC treated with glioblastoma conditioned medium express CD 151, desmin, α-SMA, nestin, and NG2, whereas no expression of the vWF or myosin could be detected, indicating a differentiation into pericyte-like cells, instead of endothelial or smooth muscle cells. From a morphologic point of view, the BM-MSC capillary-like networks substantially differ from human

Recently, as already demonstrated for adipose tissue-MSC [63], it has been identified that BM-MSC can undergo hematopoietic differentiation and achieve macrophage-like phenotypic and functional aspects [54]. After either direct or indirect co-culture with nasopharyngeal carcinoma and lung adenocarcinoma cells, BM-MSC expressed hematopoietic markers such as CD11b, CD34 and CD45. Moreover, BM-MSC conditioned by co-culturing with these cancer cells expressed the mature monocyte/macrophage markers CD68, CSF1R, and MRC1. On the contrary, they had diminished expression of the pluripotent stem cell markers OCT3/4, SOX2, and Nanog. The acquisition of a macrophagelike phenotype is reinforced by a morphology alteration to roundshaped cells, the expression of CXCL5, G-CSF, GM-CSF, CXCL1, IL-7, CCL7, CXCL6, and CCL10 and enhanced macrophage effector functions such as a phagocytic capability and nitric oxide production. The BMMSC hematopoietic transdifferentiation is dependent on a decreased transient influx of Ca2+ [54]. Although interesting, this new role for tumor engrafted BM-MSC needs further investigations into (a) the molecular signals that promote the ability of BM-MSC to transdifferentiate into hematopoietic cells; and (b) the role of the macrophage-like BM-MSC in tumor progression. Although BM-MSC plasticity has been well characterized, more studies are needed to elucidate the role of a diverse biological background in influencing the behavior of the BM-MSC. As BM-MSC have been shown to give rise to many tissues, migrating BM-MSC may represent a source of pluripotent cells that are constantly available for tumor stroma formation and for promoting the developing cancer-related inflammatory environment (Fig. 2). 4. The role of BM-MSC in tumor growth

401

In the tumor microenvironment, the BM-MSC, and their subsequently differentiated cells with distinct phenotypes, interact with the tumor cells and with the other stromal cells via a number of signaling molecules, resulting in a complex crosstalk. The factors that are produced by the BM-MSC directly, or that are produced by others cells in the tumor microenvironment after BM-MSC contact, can range from mitogens, ECM proteins, and angiogenic and inflammatory mediators and are therefore potential regulators of tumor growth and spread. The outcome of this intricate signaling network is not yet clear because BMMSC have been shown to have both stimulating and inhibiting effects on tumor progression (Fig. 3). It is conceivable that the biological context, i.e., the tumor histotypes or their local microenvironments may be critical to setting the balance between the pro- and anti-survival effects elicited by the BM-MSC.

402

4.1. Tumor progression evidence

416

During the past decade, it has been acknowledged that tumor progression is driven not only by alterations in the cancer cells but also by contributions of the cancer-associated stroma [64]. To evaluate the BM-MSC ability to induce tumor growth in vivo, assays that utilize co-

417

319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360

O

R O

P

317 318

D

315 316

E

313 314

T

308 309

C

306 307

E

304 305

R

303

R

301 302

N C O

299 300

F

endothelial cells because the tube network formed by the pericyte cells without an endothelial lining has finer branched networks, reflecting the early participation of pericytes in the angiogenic sprouting [59,61]. In fact, it must be considered that the BM-MSC exhibit a remarkable similarity to pericytes, not only because these two cell types are of a mesenchymal origin and have a resemblance in their protein marker profiles but also because they can perform analogous functions in supporting a tumor vascular network. Furthermore, as BM-MSC can be a source of pericytes, the latter can also acquire an MSC phenotype [62]. Thus, BM-MSC transdifferentiation into pericytes seems to be a plausible and recurrent event in the tumor microenvironment.

310

Recent reports have shed new light on the importance of the CAFlike derived-BM-MSC in the tumor microenvironment. The exact percentage of cells involved in this phenomenon is highly dependent on the experimental conditions and the model used; in a model of pancreatic cancer, it has been documented that ~20% of the CAF originated from BM-MSC [33], while in an ovarian cancer study, a significant 60– 70% of the FAP+ and FSP+ CAF were reported to have been derived from the bone marrow [36]. In fact, the origins of the stroma-recruited MSC were reported by Kidd et al., and it was proposed that the majority of the pericytes (neuron/glial antigen (NG)2+ and α-SMA+) and endothelial cells (CD31+) are recruited from the tumor adjacent tissue, such as local adipose tissue, whereas the FAP+ and FSP+ CAF-like cells, involved in extracellular matrix remodeling, are attracted from the host bone marrow populations, such as BM-MSC [36].

U

297 298

5

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

363 364 365 366 367 368 369 370 371 372

376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400

403 404 405 406 407 408 409 410 411 412 413 414 415

418 419 420

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

R O

O

F

6

P

Fig. 2. BM-MSC transdifferentiation ability and the role of their derived phenotypes in tumor progression. The BM-MSC potential to transdifferentiate into several cells types is recognized in many reports. The tumor microenvironment is acknowledged to drive the BM-MSC into activated fibroblast-like or perivascular-like differentiated cells.

434 435 436

E

D

the proliferation of osteosarcoma cells [30]. In keeping with a key role for BM-MSC in the formation of distant metastases, BM-MSC injected intravenously into nude mice bearing orthotopic osteosarcomas led to increased metastatic nodules, and CCL5 has been proposed to play the main role [30]. The proliferation of the Saos-2 osteosarcoma can also be induced by IL-6, secreted by BM-MSC through the activation of the STAT3 transcription factor. The IL-6/STAT3 signaling pathway gives rise to a survival signal in Saos-2 cells, thereby increasing cell migration and invasion and promoting the metastasis of osteosarcoma [71]. Additionally, the IL-6 secretion by BM-MSC increases the number of colorectal tumorinitiating cells and promotes tumor formation [69]. BM-MSC also produce TGF-β1 , acknowledged as a tumor inducing cytokine for prostate cancer cells [24]. LL-37 (leucine, leucine-37), a proinflammatory cytokine with chemoattractive properties for immune cells that also has a pro-

T

C

E

432 433

R

430 431

R

428 429

O

426 427

C

424 425

injection of these cells with different types of tumors in immunecompromised animals have been performed by several groups. BMMSC co-injected with cancer cells increased tumor growth in models of B16 melanoma [17,65], colon cancer [20,66], breast cancer [12,67], as well as in osteosarcoma [68], ovarian [21], colorectal [69,70], lung [17], gastric [34] and prostate carcinomas [24,25]. Signaling molecules secreted by BM-MSC have been acknowledged to promote tumor progression. Karnoub and collaborators investigated the tumor onset, growth kinetics and metastasis formation of four different human mammary carcinoma cell lines co-injected subcutaneously with BM-MSC. Although the BM-MSC only increased the primary tumor growth of one cell type (MCF7/Ras), they enhanced the metastatic potential of all the cancer cell lines. CCL5/RANTES, secreted by BMMSC and acting primarily through Akt signaling, was identified as a key factor in allowing metastatic colonization [12]. In addition, CCL5 produced by BM-MSC is also important in mobilizing and increasing

N

422 423

U

421

Motility Proliferation Transdifferentiation into CAF-like cells Invasion EMT Metastasis Apoptosis Inhibition Increase tumor-initiating cells Angiogenesis Transdifferentiation into perivascular-like cells Immunosuppression CCL5, TGF-β, IL-6, IL-10, VEGF, MMP, SDF-1, Neuregulin1

Oncostatin-M

BM-MSC

Tumor growth inhibition Antiangiogenic effect Endothelial cell apoptosis

Tumor mass

Fig. 3. Positive or negative role of BM-MSC in tumor progression. BM-MSC can support tumor progression by modulating several processes elicited by tumor cells or the tumor-associated stroma. Nonetheless, reports have also proposed an antitumorigenic function for BM-MSC.

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517

F

O

R O

472 473

P

470 471

D

468 469

4.1.1. Role of BM-MSC-derived CAF in tumor progression In agreement with their role as tumor stroma-associated cells, BMMSC conditioned in vitro for several days with tumor derived medium can acquire a CAF-like phenotype and can start producing SDF-1, thereby sustaining tumor growth both in vitro and in vivo [35]. SDF-1 secretion is considered a critical feature of the CAF in several tumors and one of the factors which supports tumor progression [85]. On the other hand, non-conditioned BM-MSC expressed undetectable levels of the CAF markers and did not support tumor growth [35]. The importance of resident CAF in supporting tumor progression and the preparation of a metastatic niche is well acknowledged for prostate and breast cancers [86]. The CAF engage in a biunivocal interplay with cancer cells, described as the efferent and the afferent pathways. In the efferent branch of the loop, the cancer cells cause the activation of the bystander CAF, enhancing their reactivity in terms of contractility,

change of secreted ECM proteins and soluble factors [87]. The CAF undergo activation in response to cancer derived soluble factors such as IL-6, TGF-β or CCL2 [88]. To complete the diabolic circuitry through the afferent pathway, the activated CAF affect the malignancy of the cancer cells, mainly by eliciting EMT in the primary tumor cells, as well as by enhancing tumor growth and metastatic spread [88]. Of note, the signaling elicited by the resident CAF in cancer cells undergoing EMT is similar to the proinflammatory cues that involve cyclooxygenase-2, NF-κB and HIF-1 [89]. Once activated, the CAF become active players in the cancer malignancy in several other ways. First, they can cooperate in the recruitment and organization of endothelial precursor cells, thereby facilitating de novo vessel formation and tumor growth [90,91]. Second, they can participate in the orchestration of an inflammatory response, thereby enhancing the recruitment of monocytes to the primary tumor, as well as their polarization towards the M2 tumor-promoting phenotype [92]. Third, the CAF infiltrating aggressive cancers undergo expression/activation of carbonic anhydrase IX, leading to an extracellular pH decrease and favoring extracellular protease activation, thereby driving the EMT of the cancer cells [93]. Finally, the tumor-educated CAF are active modulators of tumor metabolism, behaving as tumor cell feeders and allowing the cancer cells to regain a respiratory phenotype. Indeed, upon contact with bystander CAF, the tumor cells induce a clear Warburg metabolism to their associated CAF, which enhances glucose uptake, aerobic glycolysis and lactate secretion through the increased expression of monocarboxylate transporter (MCT)-4. In turn, the cancer cells begin to exploit this lactate, uploaded through the specific import lactate carrier, MCT-1. The cancer cells use the CAF-derived lactate by both respiring it in their reactivated mitochondrial respiration, as well as by driving it into anabolic pathways to obtain proteins and biomass to facilitate tumor growth [94]. This reconverted metabolism has been called reverse Warburg metabolism [94,95]. The role of the CAF as feeders of cancer cells has been expanded to the secretion of ketone bodies, which are useful for maintaining a high lipid synthesis rate during glucose deprivation [96]. In addition, CAF have also been shown to undergo autophagy to fuel cancer cells and sustain their growth [97]. Of course, all these metabolic adaptations can foster the metabolic pathways of the tumor cells, thereby providing them with lactate and allowing the adaptation of the tumor cells to a low glucose environment [94]. Again, the role of CAF in supporting the metabolism of cancer cells is driven by oxidative stress and the activation of HIF-1, which is a key regulator of glucose addiction and catabolic/autophagic processes [94]. Hence, the metabolic reprogramming of cancer cells to a reverse Warburg phenotype allows the tumors that are highly infiltrated by a reactive stroma to allocate a Warburg metabolism to the stromal fibroblasts and to use their byproducts to survive and grow in a glucose-free ischemic milieu. The complexity of the mutual cooperation between the stromal fibroblasts and cancer cells is further underscored by some reports that again describe a mandatory interplay among these cells, but in the opposite metabolic direction. Indeed, immunohistochemistry data suggest that in some tumor types, the cancer cells express lactate dehydrogenase (LDH)-5 and MCT-4, which is essentially linked to anaerobic metabolism, while the stromal cells express high levels of LDH-1 and MCT-1, suggesting that they use aerobic metabolism [98,99]. This idea has also been confirmed in a breast cancer model, where BM-MSC are able to upload the lactate expelled from the tumor cells and use it as a source of energy [100]. Whatever the direction of the metabolic interplay, the stromal and cancer cells exchange lactate and energy-rich compounds in a mutually favorable relationship that favors cancer malignancy. Of course, these data have important pharmacological implications, as they include infiltrated CAF, either resident or recruited by bone marrow-derived cells, as key regulators of metabolic adaptive strategies engaged by the cancer cells to face the hostile tumor environment, thereby adding a further level of complexity to the system. In line with the role of engrafted BM-MSC in a tumor microenvironment, CXCL16 expression by prostatic carcinoma cells, besides

E

466 467

T

464 465

C

462 463

E

460 461

R

459

R

457 458

N C O

455 456

angiogenic role, is found upregulated in ovarian, breast and lung tumors [72–74]. Coffelt and collaborators described a role for LL-37 in recruiting BM-MSC to tumor sites [23]. Furthermore, LL-37-stimulated BM-MSC actively secrete proinflammatory cytokines, including IL-6, IL-10, CCL5, VEGF, as well as MMP-2. Thereby, LL-37 facilitates ovarian tumor progression through the support of the tumor engrafted BM-MSC, which direct the pro-angiogenic, metastatic and immunosuppressive responses [23]. Once the migrating BM-MSC have reached the tumor site, they can face a developing environment that is inhabited by inflammatory cells. Despite a lack of exploration of the influence of macrophages on BMMSC behavior, it has been suggested that BM-MSC respond to macrophage-derived IL-8, CCL2 and CCL5 and, in turn, acquire a motile phenotype, characterized by the secretion of IL-6, CCL5 and CXCL10 [75]. These cytokines participate in both the maintenance of a proinflammatory state and in tumor growth. Nevertheless, further investigations are necessary to clarify the relationship between BM-MSC and cancer-associated macrophages. Epithelial cells form a tissue sheet through the binding of adhesion molecules, most prominently epithelial cadherin (E-Cad). Thus, the disaggregation of both normal and tumor epithelial cells is characterized by a decreased expression of E-Cad. Moreover, E-Cad downregulation is also a key event in the activation of a post-transcriptional program termed epithelial-to-mesenchymal transition (EMT), in which the cells gradually lose their epithelial characteristics and acquire a more motile and invasive mesenchymal-like phenotype, which is associated with metastatic features [76,77]. Notably, the composition of the microenvironment, including oxygen tension, growth factors, and cytokines, has recently been suggested to modulate the EMT process in cancer cells [77,78]. Breast cancer cells, upon contact with BM-MSC, increase their expression of EMT specific markers, allowing a loss of the cellular adherens-junctions and of polarization, thereby suggesting that BMMSC can induce EMT [79,80]. Lysyl oxidase has been determined to be necessary for such an interaction. Indeed, BM-MSC contact induces de novo expression of lysyl oxidase and is a key event in enhancing the lung and bone metastasis of poorly metastatic cells [81]. BM-MSC have also elicited EMT in hepatocellular carcinoma cells, leading to their metastatic progression [82]. Emphasizing the role of a proinflammatory context to elicit BM-MSC pro-tumor actions, the prometastatic effect of BM-MSC could also be reproduced with the supernatant of BM-MSC pretreated with interferon (IFN)-γ and TNF-α. Moreover, treatment of hepatocellular carcinoma cells with this supernatant leads to EMT, mainly due to the secretion of TGF-β by BM-MSC stimulated with those proinflammatory cytokines. BM-MSC also enhance the metastatic ability of ovarian and colon carcinoma cells [20,83]. Of note, circulating BM-MSC are not only able to migrate towards the orthotopic primary colon carcinoma, but they are also recruited by liver metastatic colonies. In both primary and metastatic tumors, PDGF-BB signaling plays a critical role in the interaction between BM-MSC and tumor cells [84].

U

453 454

7

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583

607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645 646 647 648

668

Depending on the experimental setting, BM-MSC have also been acknowledged to lead anti-tumorigenic responses. Khakoo et al. described intrinsic BM-MSC antineoplastic properties through Akt inactivation in Kaposi's sarcoma cells [29]. In SCID mice bearing disseminated nonHodgkin's lymphoma xenografts, the BM-MSC exhibited antitumor activity and, although they increased the release of pro-angiogenic cytokines and induced the migration of endothelial cells, they also promoted endothelial cell apoptosis [109]. Even though studies underline BM-MSC assistance in engineering long-term functional vasculature, either by transdifferentiation into endothelial/pericyte phenotypes or by the stimulation of provasculogenesis paracrine factors [46,59–61], a putative antiangiogenic role for these cells has been proposed. This outcome can occur due to BM-MSC migration towards the capillaries and their intercalation between those endothelial cells. This would increase the production of ROS by the endothelial cells, thereby leading to their apoptosis and subsequent capillary degeneration [110]. BM-MSC inhibit the in vivo tumor growth and in vitro proliferation, migration, and invasion of lung adenocarcinoma cells through the secretion of oncostatin-M, a differentiation-promoting cytokine, that exerts antiproliferative effects against several types of cancers [78]. The inhibition of glioma cell line tumorigenesis has also been attributed to BM-MSC action because the co-injection of BM-MSC with the glioma cells resulted in a significant reduction in the tumor volume and vascular density. In particular, the effects observed in the co-culture model have to be related to impaired tumor angiogenesis by virtue of reduced secretion of PDGF-BB and IL-1β [111] (Fig. 3). To better understand the controversy concerning the role of BMMSC on tumor progression, it must be considered that the complex and variable crosstalk within the tumor microenvironment modulates the BM-MSC effects. It is likely that hypoxia, the composition of the extracellular matrix, the extracellular acidity as well as the inflammatory component of the stroma are all crucial players in determining the outcome of these BM-MSC effects. When BM-MSC have been engrafted into the primary tumor site, the balance between the pro- or anti-tumor effects is determined by the tumor microenvironmental changes. In a non-adverse milieu, BM-MSC can actually repair tumor “wounds” and can have antitumor effects. Conversely, in case of adverse and sustained inflammatory conditions, recruited BM-MSC behave contrasting healing of the tumor, and concurring to sustain malignancy of the neoplasia.

669 670

O

F

4.2. Anti-tumor progression evidence

R O

605 606

P

603 604

649 650

D

601 602

the tumor neovasculature, subsequently acquiring a pericyte-like phenotype and acting as pericytes in the tumor stroma [60]. A decreased oxygen concentration, a common state in growing solid tumors, acquires an important role in promoting angiogenesis because hypoxia can augment the expression of several angiogenic factors, including the VEGFs, angiopoietin-2, PDGF-BB, placenta growth factor, TGF-β, IL-8, and HGF [105,106]. In keeping with this, BM-MSC also respond to hypoxia, increasing their secretion of VEGF-AA, thereby supporting the idea for a key role for BM-MSC during de novo angiogenesis [107]. Moreover, hypoxia was also important in stimulating TGF-β secretion from BM-MSC, which is important in breast tumor vascularization [19]. Inflammation also interacts with the BM-MSC induced vascularization as colon cancer growth was accelerated when the BM-MSC were pre-stimulated with the inflammatory cytokines IFN-γ and TNF-α. The BM-MSC pre-stimulated by both inflammatory factors expressed higher levels of VEGF via the HIF-1 signaling pathway and then had enhanced tumor angiogenesis, finally leading to colon cancer growth in mice [108].

T

599 600

C

597 598

E

595 596

R

593 594

R

591 592

O

590

C

588 589

N

586 587

attracting BM-MSC, is also able to induce their transdifferentiation into CAF-like cells, which secrete high levels of SDF-1. Thus, the SDF-1 produced by these CAF-like cells, originated from BM-MSC, is able to induce EMT in prostate tumor cells, consequently promoting metastasis [25]. In addition to prostate and breast cancers, similar cross-talk between CAF and cancer cells has been reported in other tumor histotypes. Skov3 ovarian cancer cells secrete IL-6, VEGF-AA and TGF-β1 which affect the recruitment and differentiation of BM-MSC. The BM-MSC establish a reciprocal connection with the Skov-3 cells and upregulate the secretion of these cytokines by the tumor cells. Indeed, the tumor-engrafted BM-MSC subsequently differentiate into CAF-like cells and contribute to tumor microvascularization, stromal networks, and the production of tumor-stimulating paracrine factors [21]. Aligned with the importance of the inflammatory component of BMMSC homing to tumor sites, CAF that originated from BM-MSC are recruited to inflammation-induced gastric cancers in a TGF-β and SDF1α-dependent manner [34]. Consequently, the BM-MSC-derived CAF show DNA-hypomethylation, which is a feature of CAF, and they express IL-6 and other factors that contribute to a stem cell niche, such as Wnt5α and BMP4, ultimately promoting tumor growth [34]. In a primary hepatocellular carcinoma, the engrafted BM-MSC show increased expression of FSP, a CAF marker. The BM-MSC-derived CAF increase tumor proliferation and invasion, mainly through the FSPdependent up-regulation of miR155 expression. The upregulation of miR155 has been correlated with the negative regulation of suppressor of cytokine signaling 1, thereby activating STAT3 signaling. In turn, STAT3 promotes MMP-9 expression, thus increasing tumor invasiveness [101]. In colorectal cancers, Neuregulin1 protein is secreted by BMMSC, which activates the human epidermal growth factor receptor (HER) 2/3, a family of proteins implicated in tumor progression [102]. The sustained HER2/3 activation leads to PI3K/Akt signaling, consequently increasing the survival, invasion and metastatic spread of colorectal cancer [70]. Similarly to colorectal cancer cells, cancerassociated MSC demonstrate higher expression of transmembrane Neuregulin1, a feature significantly associated with advanced cancer stages, invasion depth and a decreased 5-year progression-free survival [70]. MMPs secreted by MSC-derived CAF play a pivotal role in inducing tumor pro-invasive characteristics. In a murine model of skin carcinoma, the BM-MSC engrafted at tumor sites have turned into pericytelike cells or α-SMA expressing CAF, and the latter were exclusively responsible for producing MMPs in the tumor stroma, thereby affecting stromal reactivity [103]. Although several factors that are present in the tumor stroma can putatively drive the BM-MSC transdifferentiation towards CAF, a main role for TGF-β1 was proposed. Indeed, the inhibition of the TGF-β1/ SMAD pathway blocks the BM-MSC differentiation into CAF-like cells, and the inhibition of this signaling suppressed BM-MSC pro-tumor effect [104]. 4.1.2. BM-MSC-induced tumor neovascularization The pro-tumor behavior of BM-MSC may be due to their combined effect on tumor proliferation, tumor dissemination, and vessel distribution [21,23,66]. In certain studies, the BM-MSC-elicited tumor progression was not primarily due to the promotion of tumor cell growth or an increased number of BM-MSC, but rather to the secretion of angiogenic factors [17]. Several pro-angiogenic factors, including VEGF-AA, IL-6, TGF-β, IL-8, leukemia inhibitory factor, macrophage-colony stimulating factor and macrophage inflammatory protein-2 are produced by BM-MSC, mainly in response to treatment with tumor cell conditioned medium [17,21]. Aside from producing factors that trigger and stimulate angiogenesis, the transdifferentiation into endothelial and/or pericytes-like cells endows BM-MSC with a structural role in tumor neo-angiogenesis. In experimental glioma models, BM-MSC can efficiently integrate into

U

584 585

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

E

8

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667

671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

9

5. BM-MSC and evasion from tumor sites

5.2. Adaptive immunity modulation by BM-MSC

774

713 714

The growth of a tumor mass and the subsequent tumor cell evasion from the primary neoplastic site into the bloodstream relies on the impairment of the immune surveillance of the tumor microenvironment. The concept that BM-MSC are potent suppressors of many immune cells has been widely spread in the last decade. More recently, several authors have suggested that the BM-MSC could perform an intricate cross-talk with many effectors of the immune system, rather than a simple immune-suppression. BM-MSC are considered to be immune-privileged cells because they express MHC class I molecules, whereas they have low expression of MHC class II and co-stimulatory molecules on their cell surface. However, they still have a fascinating relationship with immune cells through direct cell contact or the secretion of soluble factors [112].

Several studies have investigated the interaction between BMMSC and T cells, the main effectors of adaptive immunity. Briefly, BM-MSC have been classified as T cell suppressors for both CD4+ T helper cells and CD8+ cytotoxic T lymphocytes. It has been described that BM-MSC can induce T cell anergy, which renders them functionally inactive [122,123], inhibits their proliferation [124] and elicits their apoptosis [125]. Moreover, BM-MSC inhibit proinflammatory cytokine production, including that of IFN-γ and TNF-α and suppress naïve and memory T cell responses to their cognate antigens [123,126]. In addition, BM-MSC also impair proliferation and partially affect the cytokine production and cytotoxic activity of natural killer T cells and γδ lymphocytes, two subsets of T cells that link innate and adaptive immunity [127]. Interestingly, BM-MSC can induce a pro-Th2 response in disorders that are associated with the dominance of Th1 cells [128] or modulate the Th1 response in Th2prevalent scenarios [129], consequently restoring the balance between these two T helper subtypes. Therefore, it has been proposed that BM-MSC sense the immunological environment and respond to it accordingly [112]. Moreover, in accordance with the complex crosstalk between BM-MSC and inflammatory and immune cells, the full T cell-suppressive capacity of human BM-MSC requires monocytes that produce IL-1β. The IL-1β, in turn, enhances the BM-MSC-mediated secretion of TGF-β, which is involved in lymphocyte suppression [130]. BM-MSC immunomodulatory effects are also supported by the upregulation of regulatory T (TReg) cells, an immune-suppressive population that is able to inhibit the production of the proinflammatory cytokines IL-17 and IL-22 by Th17 cells and to reprogram differentiated Th17 cells into IL-10+ FOXP3+ TReg cells [131]. The recent literature emphasizes the significance of the proinflammatory microenvironment on shaping the immunomodulatory repertoire of human BM-MSC. Their plasticity endows them with the capacity to adapt to external conditions by switching suppressive mechanisms [120]. Some studies have also investigated the relationship between BMMSC and dendritic cells (DC), the main type of antigen-presenting cells that modulate the adaptive immunity. BM-MSC impair DC differentiation and maturation because DC matured with TNF-α and cultured in the presence of BM-MSC failed to upregulate the maturation marker CD86 and did not increase the cell surface expression of MHC class II. Hence, BM-MSC promote a phenotype that leads to T cell anergy and TReg cell induction [132]. The immunosuppressive role of BM-MSC also extends to their effects on B-cells because when co-cultured with BM-MSC, B-cell proliferation and differentiation are impaired, resulting in decreased immunoglobulin production and impaired chemotaxis [133]. Recently, in a clear parallel with macrophage polarization, it has been proposed that BM-MSC could polarize into two different types of cells with distinct phenotypes, depending on the triggered TLR signaling. Waterman and collaborators suggested this model based on observations that TLR4-primed BM-MSC mostly secrete proinflammatory mediators, while TLR3-primed BM-MSC usually express immunosuppressive ones. Thus we can identify proinflammatory and immunocompetent MSC1 and immunosuppressive MSC2 phenotypes [134]. Accordingly, the in vitro co-culture of MSC1 with several cancer cell lines diminished tumor growth, whereas an MSC2 co-culture had opposite effects. Likewise, the treatment of tumors established in immune competent models with MSC1 resulted in attenuated tumor growth and metastasis while an MSC2-treatment led to tumor growth and spread promotion [135]. These data spotlight the importance of the activation state/polarization of the BM-MSC for their role in the microenvironment community and the further development of pro- or antitumor actions (Fig. 4). It is widely accepted that BM-MSC need to be activated by neighboring cells/factors to exert their immunomodulatory properties. In this

775 776

724 725

O

722 723

5.1. BM-MSC modulation of innate immunity

727 728

After migrating into the tissues, or in response to an inflammatory burst, monocytes can be differentiated into macrophages, which are key players in inflammatory responses and tissue regeneration. Macrophages are phenotypically differentiated into classically activated type macrophages (M1), which initiate and sustain proinflammatory actions, or alternatively polarized macrophages (M2), which are further classified into subtypes M2a, 2b or 2c according to their stimulation and subsequent cytokine and surface molecule expression. M2 macrophages are recognized to have immunoregulatory functions that are involved in tissue repair and ongoing chronic inflammation [113]. Thus, M1 macrophages assist in the Th1 responses guided by cytokines such as IL-12 and IL-23, whereas M2 macrophages express low levels of IL-12 but high levels of IL-10. In the tumor microenvironment, cancerassociated macrophages (CAM) represent the major inflammatory component, and clinical observations indicate that the CAM mainly show an M2 phenotype polarization [114]. Thus, M2-CAM influence multiple steps in tumor development including growth, invasion and metastasis of tumor cells. The CAM participate in a reciprocal interplay with CAF and the tumor cells that eventually increases tumor malignancy and favors tumor metastasis [92]. The chemokines produced by BM-MSC, including CCL3, CCL12 and CXCL12, attract and mobilize monocytes and macrophages into inflammatory sites [115]. Macrophages co-cultivated with BM-MSC show high levels of IL-10 production and phagocytic activity, and low levels of TNF-α and IFN-γ production as well as MHC class II expression, leading to polarization towards an M2-like macrophage phenotype [116,117]. The inflammatory component plays an important role in the modulation of the BM-MSC/macrophage crosstalk because BM-MSC stimulation by proinflammatory cytokines, such as IFN-γ, TNF-α, lipopolysaccharide (LPS) [118,119], or activated T cells [120] supports macrophage polarization towards an M2-like phenotype, through the expression of indoleamine 2,3-dioxygenase enzymes in BM-MSC. Neutrophils, another leukocyte cell type, are also recruited to inflamed tissues containing BM-MSC, as IFN-γ-activated BM-MSC stimulated with a TLR4 agonist secrete the neutrophil chemoattractants IL-1, IL-6 and CCL5 [121]. Taken as a whole, it is postulated that the BM-MSC/macrophage interactions in homeostatic conditions usually direct wound healing effects by the promotion of a regulatory M2-like phenotype. This crosstalk is regulated in feedback loops to prevent excessive inflammatory responses and to support tissue regeneration [112]. In a tumor microenvironment situation, where a constant and sustained proinflammatory scenario is common, this modulation is impaired and the BM-MSC-induced macrophage polarization into an M2type could allow tumor progression. This tumor progression could be due to an increase in the pro-angiogenic and immunosuppressive factors produced by M2-macrophages.

741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773

C

739 740

E

737 738

R

735 736

R

733 734

N C O

731 732

U

729 730

T

726

R O

720 721

P

718 719

D

717

E

715 716

F

712

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

R O

O

F

10

863

6. BM-MSC and the metastatic niche

864

Tumor metastasis is a multistep process. First, it requires adjacent stromal invasion and subsequent intravasation into the bloodstream by the tumor cells. Next, the cells must survive in the circulation, adhere to the endothelium, extravasate, invade the matrix and grow in the target organ. In all these stages, the tumor cells must escape from recognition and avoid destruction by the immune system. Finally, tumor cells have to adapt to several other circumstances such as oxidative stress, surviving anoikis, and the interaction with different ECM components at the metastatic site [138]. As already discussed, BM-MSC and their engrafted differentiated cells with distinct phenotypes have been involved in promoting metastasis in several tumor models. In addition to affecting invasion, tumor

853 854 855 856 857 858 859 860

865 866 867 868 869 870 871 872 873 874 875

C

E

R

851 852

R

849 850

O

847 848

C

846

N

844 845

U

842 843

growth and evasion from a primary neoplastic site, the biological responses elicited by BM-MSC may influence in several ways the further steps of the metastatic progression, particularly the formation of the metastatic niche. According to Paget's hypothesis, the sites where metastases develop are not randomly chosen; on the contrary, the tumor cell (“the seed”) has a specific affinity for the microenvironment of certain organs (“the soil”). Therefore, tumor growth in the metastatic niche is dependent on the proper growth signals from the environment where the malignant cells anchor [139]. Interestingly, clinical and experimental observations often indicate that metastases take place at sites of injury [140]. It is hypothesized that signals indicating a loss of differentiated cells can induce the self-renewal of cancer stem cells (CSC), a small subpopulation of tumor cells with an infinite proliferative potential. CSC are proposed to give rise to new tumors and, therefore, be the cause of relapse and metastasis formation [141]. CSC can also start a regenerative wound-healing process, which is thought to activate the inflammatory processes that culminate in delivering signals required to initiate metastatic tumor growth [142]. Consequently, as the BM-MSC preferentially migrate to tumors and sites of tissue injury, it is likely that they may prepare the sites for subsequent colonization, analogously to those CSC effects. This perspective could explain the evidence that BM-MSC behave in a pro-metastatic manner [140]. The role of BM-MSC in metastatic niche formation can also be extended to the regulation of CSC features. When co-cultured with breast cancer cells, the BM-MSC promoted an increase in the CSC aldehyde dehydrogenase 1-positive population [67]. This co-culture also enhanced mammosphere-forming capacities, which is consistent with the acquisition of CSC traits [81]. In addition, tumor-secreted IL-1 induces prostaglandin E(2) secretion by BM-MSC. Prostaglandin E(2), in an autocrine manner, provokes the expression of a group of cytokines which, in turn, activate β-catenin signaling in the tumor cell, triggering EMT and creating a CSC niche [143]. Moreover, Giannoni and collaborators observed that the CAF-mediated EMT in prostate carcinoma cells can increase CSC features [88]. This might suggest a role for BM-MSCderived CAF in the CSC niche formation. Bone marrow is a target organ in many metastases. BM-MSC have a role in bone marrow metastatic niche formation, as they can interact with disseminated tumor cells into the marrow space, and modify

T

861 862

scenario, an inflammatory environment seems to be necessary to promote their effect and some inflammation-related molecules such as TNF-α, IFN-γ and IL-1β might be implicated [136]. Thus, proinflammatory mediators activate, or employing a definition used by immunologists, “license” BM-MSC to be immunosuppressive or to improve their suppressive actions. The proinflammatory background is usually found at tumor sites as a result of cancer-related inflammation exerted by stromal and immune cells. The immunomodulation exerted by BMMSC would be a result of the context produced by immune cellderived signals and not a stereotypical immunosuppressive response [108,112,121]. Experimental evidence indicates that the BM-MSC immuneregulatory properties support the development of tumors as evaluated in allogeneic immunocompetent mice after the co-injection of BMMSC and B16 melanoma cells locally or at distant sites [65]. Further data suggests that the BM-MSC in the tumor inflammatory microenvironment may be endowed with immunosuppressive functions, which will help tumor cells to escape from the immune surveillance [137]. In conclusion, BM-MSC, through a complex crosstalk with several elements, inhibit many effector functions of immune cells, thereby promoting an immunosuppressive state in the tumor microenvironment. This scenario allows tumor cells and their associated stroma to overcome the immune surveillance, ultimately leading to tumor evasion from primary neoplastic sites and enhanced metastasis formation.

840 841

E

839

D

P

Fig. 4. Principal aspects of BM-MSC polarization towards MSC1 or MSC2 phenotypes. The suggested MSC1 and MSC2 polarization phenotypes and their main characteristics are listed above.

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951

F

O

R O

934 935

In the past decade, several groups have investigated the role of BMMSC on tumor progression. The initial studies demonstrated a remarkable homing to tumor sites and a putative immune-privilege from BM-MSC, which have promoted enthusiasm because these cells can be used as suitable carriers to deliver anti-tumor agents to a tumor microenvironment. Because of their features BM-MSC have the potential to serve as useful vehicles for many anti-tumor agents, especially for those having a short systemic half-life and excessive toxicity. In addition, therapeutic BM-MSC may be able to reach poorly accessible body sites. Based on gene modification techniques, researchers are transferring the killing activity of immune cells to BM-MSC, creating the socalled mesenkillers [149]. Nonetheless, subsequent data have identified BM-MSC as pro-active tumor stroma-associated cells; hence, they have been implicated in promoting almost all the features recognized as hallmarks of cancer [64], including cell survival, angiogenesis, invasion, evasion of immune system and metastasis. Moreover, significant cell plasticity has been identified, which allows engrafted-BM-MSC to acquire a range of phenotypes that is determined by the complex tumor microenvironment. Aside from the classic bone, cartilage and fat transdifferentiation ability, certain scenarios can induce myofibroblast, endothelial, pericyte or macrophage-like cell types. The cancer-related inflammatory

P

932 933

964

D

930 931

7. Conclusions

E

928 929

T

926 927

C

924 925

E

922 923

R

921

952 953

R

919 920

EMT induction [79–82,143], despite recent work suggesting that these cells can also inhibit EMT or even promote MET. Conditioned medium from BM-MSC treated with bFGF and EGF increases HGF secretion and inhibits TGF-β-induced EMT in renal tubular epithelial cells [148]. In lung adenocarcinoma cells, the treatment with conditioned medium from BM-MSC elicits a clear MET, mainly by the enhancement of epithelial markers and suppression of EMT markers and the Nanog stemness factor [78]. Therefore, depending on the context, the induction of MET by BM-MSC may be a double-edge sword because it could diminish metastatic progression, if it takes place in the primary site; however, it could also favor the metastatic exit from dormancy and colony growth, if it occurs within the metastatic niche (Fig. 5).

N C O

917 918

their proliferative and morphogenetic organization patterns [144]. Moreover, BM-MSC can also facilitate breast cancer cell migration across bone marrow endothelial cells, assisting tumor cell entry into the bone marrow. Of note, low invasive breast cancer cells seem to have a preferential tropism to bone marrow compared with highly invasive cells [145]. Therefore, it is likely that cancer subpopulations with a special tropism towards bone marrow at an initial tumorigenesis could be responsible for cancer recurrence and the resistance to therapy [145]. Interestingly, metastatic cells can bring with them certain stromal components, including activated CAF, during their circulation. This heterotypic association between the metastatic tumors and the CAF provides higher viability and growth advantages to the metastatic cells [146]. Indeed the CAF induce EMT, an epigenetic motogenic program that is correlated with resistance to anoikis, which is useful when traveling cancer cells lack adhesion in the bloodstream, and to the acquisition of stem-like traits, that are effective in regenerating tumors in the metastatic niche. As BM-MSC can engraft at tumor sites and transdifferentiate into CAF, it is tempting to assume a role for the CAF-like BM-MSC in promoting metastatic progression. Furthermore, a recent report shows that BM-MSC can spontaneously form hybrids with breast cancer cells. These hybrids showed predominantly mesenchymal morphological characteristics, mixed gene expression profiles, and increased DNA ploidy, which gave them an increased metastatic ability. Both in culture and in xenografts, the hybrids subsequently underwent DNA ploidy reduction and a morphological reversal to breast carcinoma-like morphological characteristics, while maintaining a mixed breast cancer–mesenchymal expression profile [147]. As has been argued before, EMT is considered to be critical to endowing carcinomas with metastatic features because it enhances cell motility by changing cell–cell interactions and adhesive characteristics and augments cell matrix degradation, resulting in a more invasive cell type. However, to establish and stabilize distant metastases, it is suggested that cancer cells revert to an epithelial phenotype and incorporate into remote tissues through the execution of mesenchymal-toepithelial transition (MET), the post-transcriptional reprogram that is the opposite of EMT. Several studies have noted a role for BM-MSC in

U

915 916

11

Fig. 5. BM-MSC association with tumor cells during circulation and metastatic niche formation. As it has been acknowledged for CAF, so we suggest that tumor educated-BM-MSC can (1) be transported with tumor cells during their circulation, (2) help tumor transendothelial migration, and (3) contribute to the formation of metastasis in the appropriate niche. Recently, it has been described that BM-MSC can form a putative hybrid with tumor cells [147]. Although this needs to be supported by more experimental data, we speculate that (4) the hybrid fusion possibly supports tumor survival in the bloodstream, inhibiting anoikis, assisting (5) transendothelial migration and (3) the development of a metastatic niche, where a reversion to carcinoma-like morphological characteristics through MET takes place.

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

954 955 956 957 958 959 960 961 962 963

965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986

12

987 988

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

999 1001 1000

Abbreviations bFGF Basic Fibroblast Growth Factor

1002

BM-MSC CAF CAM CSC DAMP DC E-Cad ECM EGF EMT FAP FSP HGF HIF HMGB IFN IL LDH LPS MCP MCT MET MHC miR MMP MSC NG PDGF NF-κB SDF SMA TGF TNF TLR TReg UCB uPA VEGF vWF

1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040

O

R O

P

D

1013 1014

E

1011 1012

1041

1042 1043

Acknowledgements

The authors would like to thank the Conselho Nacional de Pesquisa e Desenvolvimento (CNPq — Ministry of Education, Brazil), the Italian Association for Cancer Research (AIRC), and the Istituto Toscano Tumori 1046 and FESR-PorCreo 2012 for financial support. We would also like to 1047 express our gratitude to Dr. Andrea Morandi for kindly revising this 1048 manuscript. 1044 1045

[1] Y. Fukuchi, H. Nakajima, D. Sugiyama, I. Hirose, T. Kitamura, K. Tsuji, Human placenta-derived cells have mesenchymal stem/progenitor cell potential, Stem Cells 22 (2004) 649–658. [2] S. Ilancheran, Y. Moodley, U. Manuelpillai, Human fetal membranes: a source of stem cells for tissue regeneration and repair? Placenta 30 (2009) 2–10. [3] F. Mosna, L. Sensebe, M. Krampera, Human bone marrow and adipose tissue mesenchymal stem cells: a user's guide, Stem Cells Dev. 19 (2010) 1449–1470. [4] S. Vellasamy, P. Sandrasaigaran, S. Vidyadaran, E. George, R. Ramasamy, Isolation and characterisation of mesenchymal stem cells derived from human placenta tissue, World J. Stem Cells 4 (2012) 53–61. [5] A.K. Batsali, M.C. Kastrinaki, H.A. Papadaki, C. Pontikoglou, Mesenchymal stem cells derived from Wharton's Jelly of the umbilical cord: biological properties and emerging clinical applications, Curr. Stem Cell Res. Ther. 8 (2013) 144–155. [6] M. Dominici, K. Le Blanc, I. Mueller, I. Slaper-Cortenbach, F. Marini, D. Krause, R. Deans, A. Keating, D. Prockop, E. Horwitz, Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement, Cytotherapy 8 (2006) 315–317. [7] Z. Miao, J. Jin, L. Chen, J. Zhu, W. Huang, J. Zhao, H. Qian, X. Zhang, Isolation of mesenchymal stem cells from human placenta: comparison with human bone marrow mesenchymal stem cells, Cell Biol. Int. 30 (2006) 681–687. [8] M. Li, S. Ikehara, Bone-marrow-derived mesenchymal stem cells for organ repair, Stem Cells Int. 2013 (2013) 132642. [9] N.C. Direkze, S.J. Forbes, M. Brittan, T. Hunt, R. Jeffery, S.L. Preston, R. Poulsom, K. Hodivala-Dilke, M.R. Alison, N.A. Wright, Multiple organ engraftment by bonemarrow-derived myofibroblasts and fibroblasts in bone-marrow-transplanted mice, Stem Cells 21 (2003) 514–520. [10] C.H. Wang, W.J. Cherng, N.I. Yang, L.T. Kuo, C.M. Hsu, H.I. Yeh, Y.J. Lan, C.H. Yeh, W.L. Stanford, Late-outgrowth endothelial cells attenuate intimal hyperplasia contributed by mesenchymal stem cells after vascular injury, Arterioscler. Thromb. Vasc. Biol. 28 (2008) 54–60. [11] M. Schafer, S. Werner, Cancer as an overhealing wound: an old hypothesis revisited, Nat. Rev. Mol. Cell Biol. 9 (2008) 628–638. [12] A.E. Karnoub, A.B. Dash, A.P. Vo, A. Sullivan, M.W. Brooks, G.W. Bell, A.L. Richardson, K. Polyak, R. Tubo, R.A. Weinberg, Mesenchymal stem cells within tumour stroma promote breast cancer metastasis, Nature 449 (2007) 557–563. [13] R.M. Dwyer, S.M. Potter-Beirne, K.A. Harrington, A.J. Lowery, E. Hennessy, J.M. Murphy, F.P. Barry, T. O'Brien, M.J. Kerin, Monocyte chemotactic protein-1 secreted by primary breast tumors stimulates migration of mesenchymal stem cells, Clin. Cancer Res. 13 (2007) 5020–5027. [14] Y. Rattigan, J.M. Hsu, P.J. Mishra, J. Glod, D. Banerjee, Interleukin 6 mediated recruitment of mesenchymal stem cells to the hypoxic tumor milieu, Exp. Cell Res. 316 (2010) 3417–3424. [15] Y. Zhang, P. Yang, T. Sun, D. Li, X. Xu, Y. Rui, C. Li, M. Chong, T. Ibrahim, L. Mercatali, D. Amadori, X. Lu, D. Xie, Q.J. Li, X.F. Wang, miR-126 and miR-126* repress recruitment of mesenchymal stem cells and inflammatory monocytes to inhibit breast cancer metastasis, Nat. Cell Biol. 15 (2013) 284–294. [16] M. Gutova, J. Najbauer, R.T. Frank, S.E. Kendall, A. Gevorgyan, M.Z. Metz, M. Guevorkian, M. Edmiston, D. Zhao, C.A. Glackin, S.U. Kim, K.S. Aboody, Urokinase plasminogen activator and urokinase plasminogen activator receptor mediate human stem cell tropism to malignant solid tumors, Stem Cells 26 (2008) 1406–1413. [17] K. Suzuki, R. Sun, M. Origuchi, M. Kanehira, T. Takahata, J. Itoh, A. Umezawa, H. Kijima, S. Fukuda, Y. Saijo, Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization, Mol. Med. 17 (2011) 579–587. [18] B.M. Beckermann, G. Kallifatidis, A. Groth, D. Frommhold, A. Apel, J. Mattern, A.V. Salnikov, G. Moldenhauer, W. Wagner, A. Diehlmann, R. Saffrich, M. Schubert, A.D. Ho, N. Giese, M.W. Buchler, H. Friess, P. Buchler, I. Herr, VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma, Br. J. Cancer 99 (2008) 622–631. [19] S.C. Hung, W.P. Deng, W.K. Yang, R.S. Liu, C.C. Lee, T.C. Su, R.J. Lin, D.M. Yang, C.W. Chang, W.H. Chen, H.J. Wei, J.G. Gelovani, Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging, Clin. Cancer Res. 11 (2005) 7749–7756. [20] K. Shinagawa, Y. Kitadai, M. Tanaka, T. Sumida, M. Kodama, Y. Higashi, S. Tanaka, W. Yasui, K. Chayama, Mesenchymal stem cells enhance growth and metastasis of colon cancer, Int. J. Cancer 127 (2010) 2323–2333. [21] E.L. Spaeth, J.L. Dembinski, A.K. Sasser, K. Watson, A. Klopp, B. Hall, M. Andreeff, F. Marini, Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression, PloS one 4 (2009) e4992. [22] S. Komarova, J. Roth, R. Alvarez, D.T. Curiel, L. Pereboeva, Targeting of mesenchymal stem cells to ovarian tumors via an artificial receptor, J. Ovarian Res. 3 (2010) 12. [23] S.B. Coffelt, F.C. Marini, K. Watson, K.J. Zwezdaryk, J.L. Dembinski, H.L. LaMarca, S.L. Tomchuck, K. Honer zu Bentrup, E.S. Danka, S.L. Henkle, A.B. Scandurro, The pro-inflammatory peptide LL-37 promotes ovarian tumor progression through recruitment of multipotent mesenchymal stromal cells, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 3806–3811. [24] H. Ye, J. Cheng, Y. Tang, Z. Liu, C. Xu, Y. Liu, Y. Sun, Human bone marrow-derived mesenchymal stem cells produced TGFbeta contributes to progression and metastasis of prostate cancer, Cancer Invest. 30 (2012) 513–518. [25] Y. Jung, J.K. Kim, Y. Shiozawa, J. Wang, A. Mishra, J. Joseph, J.E. Berry, S. McGee, E. Lee, H. Sun, J. Wang, T. Jin, H. Zhang, J. Dai, P.H. Krebsbach, E.T. Keller, K.J. Pienta,

T

1009 1010

C

1007 1008

E

1005 1006

bone marrow-derived MSC cancer-associated fibroblasts cancer-associated macrophages cancer stem cells damage-associated molecular patterns dendritic cells epithelial cadherin extracellular matrix epidermal growth factor epithelial-to-mesenchymal transition fibroblast activation protein fibroblast specific protein hepatocyte growth factor hypoxia-inducible factors high-mobility group box interferon Interleukin lactate dehydrogenase lipopolysaccharide Monocyte Chemoattractant Protein monocarboxylate transporter mesenchymal-to-epithelial transition major histocompatibility complex microRNA Metalloproteinases mesenchymal stem cells neuron/glial antigen Platelet-Derived Growth Factor Nuclear Factor-κB Stromal-Derived Factor smooth muscle actin transforming growth factor tumor necrosis factor Toll-like receptors Regulatory T cells Umbilical Cord Blood urokinase plasminogen activator vascular endothelial growth factor von Willebrand Factor.

R

1003 1004

R

996 997

O

994 995

C

993

N

991 992

U

989 990

1049

References

F

998

component plays a major role in directing BM-MSC actions in the tumor stroma, not only in the recruitment and induction of subsequent transdifferentiation but also in the favoring of MSC polarization towards a pro- or anti-tumor supportive type (MSC1/2). Likewise, inflammation has been proposed to promote BM-MSC licensing to exert immune-suppressive effects, impairing immune surveillance in the tumor microenvironment and enhancing tumor progression. To date, the understanding of BM-MSC biology is far from complete and strongly needs further work that will lead to the development of more rational and selective therapies in advance of their safe use in patients.

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

1050 1051 1052 1053 1054 1055 1056 1057 1058 1059 1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075 1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091 1092 1093 1094 1095 1096 1097 1098 1099 1100 1101 1102 1103 1104 1105 1106 1107 1108 1109 1110 1111 1112 1113 1114 1115 1116 1117 1118 1119 1120 1121 1122 1123 1124 1125 1126 1127 1128 1129 1130 1131 1132 1133

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

[31] [32]

[33]

[34]

[35]

[36]

[41]

[42]

[43]

[44]

[45] [46]

[47]

[48]

[49] [50]

E

R

[40]

R

[39]

N C O

[38]

C

F

T

[37]

O

[30]

R O

[29]

P

[28]

D

[27]

[51] R. Lotfi, J. Eisenbacher, G. Solgi, K. Fuchs, T. Yildiz, C. Nienhaus, M.T. Rojewski, H. Schrezenmeier, Human mesenchymal stem cells respond to native but not oxidized damage associated molecular pattern molecules from necrotic (tumor) material, Eur. J. Immunol. 41 (2011) 2021–2028. [52] V. Pistoia, L. Raffaghello, Damage-associated molecular patterns (DAMPs) and mesenchymal stem cells: a matter of attraction and excitement, Eur. J. Immunol. 41 (2011) 1828–1831. [53] L. Micallef, N. Vedrenne, F. Billet, B. Coulomb, I.A. Darby, A. Desmouliere, The myofibroblast, multiple origins for major roles in normal and pathological tissue repair, Fibrogenesis Tissue Repair 5 (Suppl. 1) (2012) S5. [54] L. Zhang, A. Tang, Y. Zhou, J. Tang, Z. Luo, C. Jiang, X. Li, J. Xiang, G. Li, Tumor-conditioned mesenchymal stem cells display hematopoietic differentiation and diminished influx of Ca2+, Stem Cells Dev. 21 (2012) 1418–1428. [55] Y. Cao, Y. Meng, Z. Sun, L.M. Liao, Q. Han, J. Li, Y.N. Liu, C.H. Zhao, [Potential of human adipose tissue derived adult stem cells differentiate into endothelial cells], Zhongguo yi xue ke xue yuan xue bao, Acta Acad. Med. Sin. 27 (2005) 678–682. [56] L.J. Fischer, S. McIlhenny, T. Tulenko, N. Golesorkhi, P. Zhang, R. Larson, J. Lombardi, I. Shapiro, P.J. DiMuzio, Endothelial differentiation of adipose-derived stem cells: effects of endothelial cell growth supplement and shear force, J. Surg. Res. 152 (2009) 157–166. [57] J. Oswald, S. Boxberger, B. Jorgensen, S. Feldmann, G. Ehninger, M. Bornhauser, C. Werner, Mesenchymal stem cells can be differentiated into endothelial cells in vitro, Stem Cells 22 (2004) 377–384. [58] K. Janeczek Portalska, A. Leferink, N. Groen, H. Fernandes, L. Moroni, C. van Blitterswijk, J. de Boer, Endothelial differentiation of mesenchymal stromal cells, PloS one 7 (2012) e46842. [59] P. Au, J. Tam, D. Fukumura, R.K. Jain, Bone marrow-derived mesenchymal stem cells facilitate engineering of long-lasting functional vasculature, Blood 111 (2008) 4551–4558. [60] D. Bexell, S. Gunnarsson, A. Tormin, A. Darabi, D. Gisselsson, L. Roybon, S. Scheding, J. Bengzon, Bone marrow multipotent mesenchymal stroma cells act as pericyte-like migratory vehicles in experimental gliomas, Mol. Ther. 17 (2009) 183–190. [61] T. Birnbaum, J. Hildebrandt, G. Nuebling, P. Sostak, A. Straube, Glioblastomadependent differentiation and angiogenic potential of human mesenchymal stem cells in vitro, J. Neuro-Oncol. 105 (2011) 57–65. [62] P. Dore-Duffy, Pericytes: pluripotent cells of the blood brain barrier, Curr. Pharm. Des. 14 (2008) 1581–1593. [63] E. Freisinger, C. Cramer, X. Xia, S.N. Murthy, D.P. Slakey, E. Chiu, E.R. Newsome, E.U. Alt, R. Izadpanah, Characterization of hematopoietic potential of mesenchymal stem cells, J. Cell. Physiol. 225 (2010) 888–897. [64] D. Hanahan, R.A. Weinberg, Hallmarks of cancer: the next generation, Cell 144 (2011) 646–674. [65] F. Djouad, P. Plence, C. Bony, P. Tropel, F. Apparailly, J. Sany, D. Noel, C. Jorgensen, Immunosuppressive effect of mesenchymal stem cells favors tumor growth in allogeneic animals, Blood 102 (2003) 3837–3844. [66] W. Zhu, W. Xu, R. Jiang, H. Qian, M. Chen, J. Hu, W. Cao, C. Han, Y. Chen, Mesenchymal stem cells derived from bone marrow favor tumor cell growth in vivo, Exp. Mol. Pathol. 80 (2006) 267–274. [67] S. Liu, C. Ginestier, S.J. Ou, S.G. Clouthier, S.H. Patel, F. Monville, H. Korkaya, A. Heath, J. Dutcher, C.G. Kleer, Y. Jung, G. Dontu, R. Taichman, M.S. Wicha, Breast cancer stem cells are regulated by mesenchymal stem cells through cytokine networks, Cancer Res. 71 (2011) 614–624. [68] Z.Y. Bian, Q.M. Fan, G. Li, W.T. Xu, T.T. Tang, Human mesenchymal stem cells promote growth of osteosarcoma: involvement of interleukin-6 in the interaction between human mesenchymal stem cells and Saos-2, Cancer Sci. 101 (2010) 2554–2560. [69] K.S. Tsai, S.H. Yang, Y.P. Lei, C.C. Tsai, H.W. Chen, C.Y. Hsu, L.L. Chen, H.W. Wang, S.A. Miller, S.H. Chiou, M.C. Hung, S.C. Hung, Mesenchymal stem cells promote formation of colorectal tumors in mice, Gastroenterology 141 (2011) 1046–1056. [70] A. De Boeck, P. Pauwels, K. Hensen, J.L. Rummens, W. Westbroek, A. Hendrix, D. Maynard, H. Denys, K. Lambein, G. Braems, C. Gespach, M. Bracke, O. De Wever, Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression through paracrine neuregulin 1/HER3 signalling, Gut 62 (2013) 550–560. [71] B. Tu, L. Du, Q.M. Fan, Z. Tang, T.T. Tang, STAT3 activation by IL-6 from mesenchymal stem cells promotes the proliferation and metastasis of osteosarcoma, Cancer Lett. 325 (2012) 80–88. [72] J.D. Heilborn, M.F. Nilsson, C.I. Jimenez, B. Sandstedt, N. Borregaard, E. Tham, O.E. Sorensen, G. Weber, M. Stahle, Antimicrobial protein hCAP18/LL-37 is highly expressed in breast cancer and is a putative growth factor for epithelial cells, Int. J. Cancer 114 (2005) 713–719. [73] S.B. Coffelt, R.S. Waterman, L. Florez, K. Honer zu Bentrup, K.J. Zwezdaryk, S.L. Tomchuck, H.L. LaMarca, E.S. Danka, C.A. Morris, A.B. Scandurro, Ovarian cancers overexpress the antimicrobial protein hCAP-18 and its derivative LL-37 increases ovarian cancer cell proliferation and invasion, Int. J. Cancer 122 (2008) 1030–1039. [74] J. von Haussen, R. Koczulla, R. Shaykhiev, C. Herr, O. Pinkenburg, D. Reimer, R. Wiewrodt, S. Biesterfeld, A. Aigner, F. Czubayko, R. Bals, The host defence peptide LL-37/hCAP-18 is a growth factor for lung cancer cells, Lung Cancer 59 (2008) 12–23. [75] K. Anton, D. Banerjee, J. Glod, Macrophage-associated mesenchymal stem cells assume an activated, migratory, pro-inflammatory phenotype with increased IL-6 and CXCL10 secretion, PloS one 7 (2012) e35036. [76] T.T. Onder, P.B. Gupta, S.A. Mani, J. Yang, E.S. Lander, R.A. Weinberg, Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways, Cancer Res. 68 (2008) 3645–3654.

E

[26]

R.S. Taichman, Recruitment of mesenchymal stem cells into prostate tumours promotes metastasis, Nat. Commun. 4 (2013) 1795. M. Studeny, F.C. Marini, R.E. Champlin, C. Zompetta, I.J. Fidler, M. Andreeff, Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors, Cancer Res. 62 (2002) 3603–3608. A. Nakamizo, F. Marini, T. Amano, A. Khan, M. Studeny, J. Gumin, J. Chen, S. Hentschel, G. Vecil, J. Dembinski, M. Andreeff, F.F. Lang, Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas, Cancer Res. 65 (2005) 3307–3318. C. Schichor, T. Birnbaum, N. Etminan, O. Schnell, S. Grau, S. Miebach, K. Aboody, C. Padovan, A. Straube, J.C. Tonn, R. Goldbrunner, Vascular endothelial growth factor A contributes to glioma-induced migration of human marrow stromal cells (hMSC), Exp. Neurol. 199 (2006) 301–310. A.Y. Khakoo, S. Pati, S.A. Anderson, W. Reid, M.F. Elshal, I.I. Rovira, A.T. Nguyen, D. Malide, C.A. Combs, G. Hall, J. Zhang, M. Raffeld, T.B. Rogers, W. Stetler-Stevenson, J.A. Frank, M. Reitz, T. Finkel, Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma, J. Exp. Med. 203 (2006) 1235–1247. W.T. Xu, Z.Y. Bian, Q.M. Fan, G. Li, T.T. Tang, Human mesenchymal stem cells (hMSCs) target osteosarcoma and promote its growth and pulmonary metastasis, Cancer Lett. 281 (2009) 32–41. S.A. Bergfeld, Y.A. DeClerck, Bone marrow-derived mesenchymal stem cells and the tumor microenvironment, Cancer Metastasis Rev. 29 (2010) 249–261. G. Ishii, T. Sangai, T. Oda, Y. Aoyagi, T. Hasebe, N. Kanomata, Y. Endoh, C. Okumura, Y. Okuhara, J. Magae, M. Emura, T. Ochiya, A. Ochiai, Bone-marrow-derived myofibroblasts contribute to the cancer-induced stromal reaction, Biochem. Biophys. Res. Commun. 309 (2003) 232–240. N.C. Direkze, K. Hodivala-Dilke, R. Jeffery, T. Hunt, R. Poulsom, D. Oukrif, M.R. Alison, N.A. Wright, Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts, Cancer Res. 64 (2004) 8492–8495. M. Quante, S.P. Tu, H. Tomita, T. Gonda, S.S. Wang, S. Takashi, G.H. Baik, W. Shibata, B. Diprete, K.S. Betz, R. Friedman, A. Varro, B. Tycko, T.C. Wang, Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth, Cancer Cell 19 (2011) 257–272. P.J. Mishra, P.J. Mishra, R. Humeniuk, D.J. Medina, G. Alexe, J.P. Mesirov, S. Ganesan, J.W. Glod, D. Banerjee, Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells, Cancer Res. 68 (2008) 4331–4339. S. Kidd, E. Spaeth, K. Watson, J. Burks, H. Lu, A. Klopp, M. Andreeff, F.C. Marini, Origins of the tumor microenvironment: quantitative assessment of adipose-derived and bone marrow-derived stroma, PloS one 7 (2012) e30563. V. Sordi, M.L. Malosio, F. Marchesi, A. Mercalli, R. Melzi, T. Giordano, N. Belmonte, G. Ferrari, B.E. Leone, F. Bertuzzi, G. Zerbini, P. Allavena, E. Bonifacio, L. Piemonti, Bone marrow mesenchymal stem cells express a restricted set of functionally active chemokine receptors capable of promoting migration to pancreatic islets, Blood 106 (2005) 419–427. I. Von Luttichau, M. Notohamiprodjo, A. Wechselberger, C. Peters, A. Henger, C. Seliger, R. Djafarzadeh, R. Huss, P.J. Nelson, Human adult CD34-progenitor cells functionally express the chemokine receptors CCR1, CCR4, CCR7, CXCR5, and CCR10 but not CXCR4, Stem Cells Dev. 14 (2005) 329–336. M. Honczarenko, Y. Le, M. Swierkowski, I. Ghiran, A.M. Glodek, L.E. Silberstein, Human bone marrow stromal cells express a distinct set of biologically functional chemokine receptors, Stem Cells 24 (2006) 1030–1041. J. Ringe, S. Strassburg, K. Neumann, M. Endres, M. Notter, G.R. Burmester, C. Kaps, M. Sittinger, Towards in situ tissue repair: human mesenchymal stem cells express chemokine receptors CXCR1, CXCR2 and CCR2, and migrate upon stimulation with CXCL8 but not CCL2, J. Cell. Biochem. 101 (2007) 135–146. E. Spaeth, A. Klopp, J. Dembinski, M. Andreeff, F. Marini, Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells, Gene Ther. 15 (2008) 730–738. B. Ruster, S. Gottig, R.J. Ludwig, R. Bistrian, S. Muller, E. Seifried, J. Gille, R. Henschler, Mesenchymal stem cells display coordinated rolling and adhesion behavior on endothelial cells, Blood 108 (2006) 3938–3944. C. Ries, V. Egea, M. Karow, H. Kolb, M. Jochum, P. Neth, MMP-2, MT1-MMP, and TIMP-2 are essential for the invasive capacity of human mesenchymal stem cells: differential regulation by inflammatory cytokines, Blood 109 (2007) 4055–4063. I.A. Ho, K.Y. Chan, W.H. Ng, C.M. Guo, K.M. Hui, P. Cheang, P.Y. Lam, Matrix metalloproteinase 1 is necessary for the migration of human bone marrow-derived mesenchymal stem cells toward human glioma, Stem Cells 27 (2009) 1366–1375. S.I. Grivennikov, F.R. Greten, M. Karin, Immunity, inflammation, and cancer, Cell 140 (2010) 883–899. B. Annabi, Y.T. Lee, S. Turcotte, E. Naud, R.R. Desrosiers, M. Champagne, N. Eliopoulos, J. Galipeau, R. Beliveau, Hypoxia promotes murine bone-marrowderived stromal cell migration and tube formation, Stem Cells 21 (2003) 337–347. S. Proulx-Bonneau, A. Guezguez, B. Annabi, A concerted HIF-1alpha/MT1-MMP signalling axis regulates the expression of the 3BP2 adaptor protein in hypoxic mesenchymal stromal cells, PloS one 6 (2011) e21511. I. Rosova, M. Dao, B. Capoccia, D. Link, J.A. Nolta, Hypoxic preconditioning results in increased motility and improved therapeutic potential of human mesenchymal stem cells, Stem Cells 26 (2008) 2173–2182. H.K. Eltzschig, P. Carmeliet, Hypoxia and inflammation, N. Engl. J. Med. 364 (2011) 656–665. E. Meng, Z. Guo, H. Wang, J. Jin, J. Wang, H. Wang, C. Wu, L. Wang, High mobility group box 1 protein inhibits the proliferation of human mesenchymal stem cells and promotes their migration and differentiation along osteoblastic pathway, Stem Cells Dev. 17 (2008) 805–813.

U

1134 1135 1136 1137 1138 1139 1140 1141 1142 1143 1144 1145 1146 1147 Q3 1148 1149 1150 1151 1152 1153 1154 1155 1156 1157 1158 1159 1160 1161 1162 1163 1164 1165 1166 1167 1168 1169 1170 1171 1172 1173 1174 1175 1176 1177 1178 1179 1180 1181 1182 1183 1184 1185 1186 1187 1188 1189 1190 1191 1192 1193 1194 1195 1196 1197 1198 1199 1200 1201 1202 1203 1204 1205 1206 1207 1208 1209 1210 1211 1212 1213 1214 1215 1216 1217 1218 1219

13

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

1220 1221 1222 1223 1224 1225 1226 1227 1228 1229 1230 1231 1232 1233 1234 1235 1236 1237 1238 1239 1240 1241 1242 1243 1244 1245 1246 1247 1248 1249 1250 1251 1252 1253 1254 1255 1256 1257 1258 1259 1260 1261 1262 1263 1264 1265 1266 1267 1268 1269 1270 1271 1272 1273 1274 1275 1276 1277 1278 1279 1280 1281 1282 1283 1284 1285 1286 1287 1288 1289 1290 1291 1292 1293 1294 1295 1296 1297 1298 1299 1300 1301 1302 1303 1304 1305

D

P

R O

O

F

[100] Y.I. Rattigan, B.B. Patel, E. Ackerstaff, G. Sukenick, J.A. Koutcher, J.W. Glod, D. Banerjee, Lactate is a mediator of metabolic cooperation between stromal carcinoma associated fibroblasts and glycolytic tumor cells in the tumor microenvironment, Exp. Cell Res. 318 (2012) 326–335. [101] X.L. Yan, Y.L. Jia, L. Chen, Q. Zeng, J.N. Zhou, C.J. Fu, H.X. Chen, H.F. Yuan, Z.W. Li, L. Shi, Y.C. Xu, J.X. Wang, X.M. Zhang, L.J. He, C. Zhai, W. Yue, X.T. Pei, Hepatocellular carcinoma-associated mesenchymal stem cells promote hepatocarcinoma progression: role of the S100A4-miR155-SOCS1-MMP9 axis, Hepatology 57 (2013) 2274–2286. [102] N.V. Sergina, M.M. Moasser, The HER family and cancer: emerging molecular mechanisms and therapeutic targets, Trends Mol. Med. 13 (2007) 527–534. [103] J. Lecomte, A. Masset, S. Blacher, L. Maertens, A. Gothot, M. Delgaudine, F. Bruyere, O. Carnet, J. Paupert, M. Illemann, J.M. Foidart, I.K. Lund, G. Hoyer-Hansen, A. Noel, Bone marrow-derived myofibroblasts are the providers of pro-invasive matrix metalloproteinase 13 in primary tumor, Neoplasia 14 (2012) 943–951. [104] L. Shangguan, X. Ti, U. Krause, B. Hai, Y. Zhao, Z. Yang, F. Liu, Inhibition of TGF-beta/Smad signaling by BAMBI blocks differentiation of human mesenchymal stem cells to carcinoma-associated fibroblasts and abolishes their protumor effects, Stem Cells 30 (2012) 2810–2819. [105] P.L. Marjon, E.V. Bobrovnikova-Marjon, S.F. Abcouwer, Expression of the pro-angiogenic factors vascular endothelial growth factor and interleukin-8/ CXCL8 by human breast carcinomas is responsive to nutrient deprivation and endoplasmic reticulum stress, Mol. Cancer 3 (2004) 4. [106] D. Fukumura, R.K. Jain, Tumor microvasculature and microenvironment: targets for anti-angiogenesis and normalization, Microvasc. Res. 74 (2007) 72–84. [107] E. Potier, E. Ferreira, R. Andriamanalijaona, J.P. Pujol, K. Oudina, D. LogeartAvramoglou, H. Petite, Hypoxia affects mesenchymal stromal cell osteogenic differentiation and angiogenic factor expression, Bone 40 (2007) 1078–1087. [108] Y. Liu, Z.P. Han, S.S. Zhang, Y.Y. Jing, X.X. Bu, C.Y. Wang, K. Sun, G.C. Jiang, X. Zhao, R. Li, L. Gao, Q.D. Zhao, M.C. Wu, L.X. Wei, Effects of inflammatory factors on mesenchymal stem cells and their role in the promotion of tumor angiogenesis in colon cancer, J. Biol. Chem. 286 (2011) 25007–25015. [109] P. Secchiero, S. Zorzet, C. Tripodo, F. Corallini, E. Melloni, L. Caruso, R. Bosco, S. Ingrao, B. Zavan, G. Zauli, Human bone marrow mesenchymal stem cells display anti-cancer activity in SCID mice bearing disseminated non-Hodgkin's lymphoma xenografts, PloS one 5 (2010) e11140. [110] K. Otsu, S. Das, S.D. Houser, S.K. Quadri, S. Bhattacharya, J. Bhattacharya, Concentration-dependent inhibition of angiogenesis by mesenchymal stem cells, Blood 113 (2009) 4197–4205. [111] I.A. Ho, H.C. Toh, W.H. Ng, Y.L. Teo, C.M. Guo, K.M. Hui, P.Y. Lam, Human bone marrow-derived mesenchymal stem cells suppress human glioma growth through inhibition of angiogenesis, Stem Cells 31 (2013) 146–155. [112] K. Le Blanc, D. Mougiakakos, Multipotent mesenchymal stromal cells and the innate immune system, Nature reviews, Immunology 12 (2012) 383–396. [113] A. Sica, A. Mantovani, Macrophage plasticity and polarization: in vivo veritas, J. Clin. Invest. 122 (2012) 787–795. [114] A. Mantovani, P. Allavena, A. Sica, F. Balkwill, Cancer-related inflammation, Nature 454 (2008) 436–444. [115] L. Chen, E.E. Tredget, P.Y. Wu, Y. Wu, Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing, PloS one 3 (2008) e1886. [116] J. Kim, P. Hematti, Mesenchymal stem cell-educated macrophages: a novel type of alternatively activated macrophages, Exp. Hematol. 37 (2009) 1445–1453. [117] J. Maggini, G. Mirkin, I. Bognanni, J. Holmberg, I.M. Piazzon, I. Nepomnaschy, H. Costa, C. Canones, S. Raiden, M. Vermeulen, J.R. Geffner, Mouse bone marrowderived mesenchymal stromal cells turn activated macrophages into a regulatory-like profile, PloS one 5 (2010) e9252. [118] M. Francois, R. Romieu-Mourez, M. Li, J. Galipeau, Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation, Mol. Ther. 20 (2012) 187–195. [119] J. Croitoru-Lamoury, F.M. Lamoury, M. Caristo, K. Suzuki, D. Walker, O. Takikawa, R. Taylor, B.J. Brew, Interferon-gamma regulates the proliferation and differentiation of mesenchymal stem cells via activation of indoleamine 2,3 dioxygenase (IDO), PloS one 6 (2011) e14698. [120] D. Mougiakakos, R. Jitschin, C.C. Johansson, R. Okita, R. Kiessling, K. Le Blanc, The impact of inflammatory licensing on heme oxygenase-1-mediated induction of regulatory T cells by human mesenchymal stem cells, Blood 117 (2011) 4826–4835. [121] R. Romieu-Mourez, M. Francois, M.N. Boivin, M. Bouchentouf, D.E. Spaner, J. Galipeau, Cytokine modulation of TLR expression and activation in mesenchymal stromal cells leads to a proinflammatory phenotype, J. Immunol. 182 (2009) 7963–7973. [122] M. Di Nicola, C. Carlo-Stella, M. Magni, M. Milanesi, P.D. Longoni, P. Matteucci, S. Grisanti, A.M. Gianni, Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli, Blood 99 (2002) 3838–3843. [123] E. Zappia, S. Casazza, E. Pedemonte, F. Benvenuto, I. Bonanni, E. Gerdoni, D. Giunti, A. Ceravolo, F. Cazzanti, F. Frassoni, G. Mancardi, A. Uccelli, Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy, Blood 106 (2005) 1755–1761. [124] M. Najar, G. Raicevic, H.I. Boufker, H. Fayyad Kazan, C. De Bruyn, N. Meuleman, D. Bron, M. Toungouz, L. Lagneaux, Mesenchymal stromal cells use PGE2 to modulate activation and proliferation of lymphocyte subsets: combined comparison of adipose tissue, Wharton's Jelly and bone marrow sources, Cell. Immunol. 264 (2010) 171–179.

N

C

O

R

R

E

C

T

[77] E. Giannoni, M. Parri, P. Chiarugi, EMT and oxidative stress: a bidirectional interplay affecting tumor malignancy, Antioxid. Redox Signal. 16 (2012) 1248–1263. [78] M.L. Wang, C.M. Pan, S.H. Chiou, W.H. Chen, H.Y. Chang, O.K. Lee, H.S. Hsu, C.W. Wu, Oncostatin m modulates the mesenchymal-epithelial transition of lung adenocarcinoma cells by a mesenchymal stem cell-mediated paracrine effect, Cancer Res. 72 (2012) 6051–6064. [79] F.T. Martin, R.M. Dwyer, J. Kelly, S. Khan, J.M. Murphy, C. Curran, N. Miller, E. Hennessy, P. Dockery, F.P. Barry, T. O'Brien, M.J. Kerin, Potential role of mesenchymal stem cells (MSCs) in the breast tumour microenvironment: stimulation of epithelial to mesenchymal transition (EMT), Breast Cancer Res. Treat. 124 (2010) 317–326. [80] Q. Xu, L. Wang, H. Li, Q. Han, J. Li, X. Qu, S. Huang, R.C. Zhao, Mesenchymal stem cells play a potential role in regulating the establishment and maintenance of epithelial-mesenchymal transition in MCF7 human breast cancer cells by paracrine and induced autocrine TGF-beta, Int. J. Oncol. 41 (2012) 959–968. [81] C.P. El-Haibi, G.W. Bell, J. Zhang, A.Y. Collmann, D. Wood, C.M. Scherber, E. Csizmadia, O. Mariani, C. Zhu, A. Campagne, M. Toner, S.N. Bhatia, D. Irimia, A. Vincent-Salomon, A.E. Karnoub, Critical role for lysyl oxidase in mesenchymal stem cell-driven breast cancer malignancy, Proc. Natl. Acad. Sci. U. S. A. 109 (2012) 17460–17465. [82] Y. Jing, Z. Han, Y. Liu, K. Sun, S. Zhang, G. Jiang, R. Li, L. Gao, X. Zhao, D. Wu, X. Cai, M. Wu, L. Wei, Mesenchymal stem cells in inflammation microenvironment accelerates hepatocellular carcinoma metastasis by inducing epithelial–mesenchymal transition, PloS one 7 (2012) e43272. [83] R. Lis, C. Touboul, C.M. Raynaud, J.A. Malek, K. Suhre, M. Mirshahi, A. Rafii, Mesenchymal cell interaction with ovarian cancer cells triggers pro-metastatic properties, PloS one 7 (2012) e38340. [84] K. Shinagawa, Y. Kitadai, M. Tanaka, T. Sumida, M. Onoyama, M. Ohnishi, E. Ohara, Y. Higashi, S. Tanaka, W. Yasui, K. Chayama, Stroma-directed imatinib therapy impairs the tumor-promoting effect of bone marrow-derived mesenchymal stem cells in an orthotopic transplantation model of colon cancer, Int. J. Cancer 132 (2013) 813–823. [85] A. Orimo, P.B. Gupta, D.C. Sgroi, F. Arenzana-Seisdedos, T. Delaunay, R. Naeem, V.J. Carey, A.L. Richardson, R.A. Weinberg, Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion, Cell 121 (2005) 335–348. [86] P. Cirri, P. Chiarugi, Cancer-associated-fibroblasts and tumour cells: a diabolic liaison driving cancer progression, Cancer Metastasis Rev. 31 (2012) 195–208. [87] R. Kalluri, EMT: when epithelial cells decide to become mesenchymal-like cells, J. Clin. Invest. 119 (2009) 1417–1419. [88] E. Giannoni, F. Bianchini, L. Masieri, S. Serni, E. Torre, L. Calorini, P. Chiarugi, Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial–mesenchymal transition and cancer stemness, Cancer Res. 70 (2010) 6945–6956. [89] E. Giannoni, F. Bianchini, L. Calorini, P. Chiarugi, Cancer associated fibroblasts exploit reactive oxygen species through a proinflammatory signature leading to epithelial mesenchymal transition and stemness, Antioxid. Redox Signal. 14 (2011) 2361–2371. [90] A. Orimo, R.A. Weinberg, Stromal fibroblasts in cancer: a novel tumor-promoting cell type, Cell Cycle 5 (2006) 1597–1601. [91] E. Giannoni, M.L. Taddei, M. Parri, F. Bianchini, M. Santosuosso, R. Grifantini, G. Fibbi, B. Mazzanti, L. Calorini, P. Chiarugi, EphA2-mediated mesenchymalamoeboid transition induced by endothelial progenitor cells enhances metastatic spread due to cancer-associated fibroblasts, J. Mol. Med. 91 (2013) 103–115. [92] G. Comito, E. Giannoni, C.P. Segura, P. Barcellos-de-Souza, M.R. Raspollini, G. Baroni, M. Lanciotti, S. Serni, P. Chiarugi, Cancer-associated fibroblasts and M2-polarized macrophages synergize during prostate carcinoma progression, Oncogene (2013). [93] T. Fiaschi, E. Giannoni, M.L. Taddei, P. Cirri, A. Marini, G. Pintus, C. Nativi, B. Richichi, A. Scozzafava, F. Carta, E. Torre, C.T. Supuran, P. Chiarugi, Carbonic anhydrase IX from cancer-associated fibroblasts drives epithelial–mesenchymal transition in prostate carcinoma cells, Cell Cycle 12 (2013) 1791–1801. [94] T. Fiaschi, A. Marini, E. Giannoni, M.L. Taddei, P. Gandellini, A. De Donatis, M. Lanciotti, S. Serni, P. Cirri, P. Chiarugi, Reciprocal metabolic reprogramming through lactate shuttle coordinately influences tumor-stroma interplay, Cancer Res. 72 (2012) 5130–5140. [95] S. Pavlides, D. Whitaker-Menezes, R. Castello-Cros, N. Flomenberg, A.K. Witkiewicz, P.G. Frank, M.C. Casimiro, C. Wang, P. Fortina, S. Addya, R.G. Pestell, U.E. Martinez-Outschoorn, F. Sotgia, M.P. Lisanti, The reverse Warburg effect: aerobic glycolysis in cancer associated fibroblasts and the tumor stroma, Cell Cycle 8 (2009) 3984–4001. [96] G. Bonuccelli, A. Tsirigos, D. Whitaker-Menezes, S. Pavlides, R.G. Pestell, B. Chiavarina, P.G. Frank, N. Flomenberg, A. Howell, U.E. Martinez-Outschoorn, F. Sotgia, M.P. Lisanti, Ketones and lactate “fuel” tumor growth and metastasis: evidence that epithelial cancer cells use oxidative mitochondrial metabolism, Cell Cycle 9 (2010) 3506–3514. [97] U.E. Martinez-Outschoorn, S. Pavlides, A. Howell, R.G. Pestell, H.B. Tanowitz, F. Sotgia, M.P. Lisanti, Stromal–epithelial metabolic coupling in cancer: integrating autophagy and metabolism in the tumor microenvironment, Int. J. Biochem. Cell Biol. 43 (2011) 1045–1051. [98] A. Giatromanolaki, M.I. Koukourakis, A. Koutsopoulos, S. Mendrinos, E. Sivridis, The metabolic interactions between tumor cells and tumor-associated stroma (TAS) in prostatic cancer, Cancer Biol. Ther. 13 (2012) 1284–1289. [99] M.I. Koukourakis, A. Giatromanolaki, A.L. Harris, E. Sivridis, Comparison of metabolic pathways between cancer cells and stromal cells in colorectal carcinomas: a metabolic survival role for tumor-associated stroma, Cancer Res. 66 (2006) 632–637.

U

1306 1307 1308 1309 1310 1311 1312 1313 1314 1315 1316 1317 1318 1319 1320 1321 1322 1323 1324 1325 1326 1327 1328 1329 1330 1331 1332 1333 1334 1335 1336 1337 1338 1339 1340 1341 1342 1343 1344 1345 1346 1347 1348 1349 1350 1351 1352 1353 1354 1355 1356 1357 1358 1359 1360 1361 1362 Q4 1363 1364 1365 1366 1367 1368 1369 1370 1371 1372 1373 1374 1375 1376 1377 1378 1379 1380 1381 1382 1383 1384 1385 1386 1387 1388 1389 1390 1391

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

E

14

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

1392 1393 1394 1395 1396 1397 1398 1399 1400 1401 1402 1403 1404 1405 1406 1407 1408 1409 1410 1411 1412 1413 1414 1415 1416 1417 1418 1419 1420 1421 1422 1423 1424 1425 1426 1427 1428 1429 1430 1431 1432 1433 1434 1435 1436 1437 1438 1439 1440 1441 1442 1443 1444 1445 1446 1447 1448 1449 1450 1451 1452 1453 1454 1455 1456 1457 1458 1459 1460 1461 1462 1463 1464 1465 1466 1467 1468 1469 1470 1471 1472 1473 1474 1475 1476 1477

P. Barcellos-de-Souza et al. / Biochimica et Biophysica Acta xxx (2013) xxx–xxx

E

D

P

R O

O

F

[144] F.A. Fierro, W.D. Sierralta, M.J. Epunan, J.J. Minguell, Marrow-derived mesenchymal stem cells: role in epithelial tumor cell determination, Clin. Exp. Metastasis 21 (2004) 313–319. [145] K.E. Corcoran, K.A. Trzaska, H. Fernandes, M. Bryan, M. Taborga, V. Srinivas, K. Packman, P.S. Patel, P. Rameshwar, Mesenchymal stem cells in early entry of breast cancer into bone marrow, PloS one 3 (2008) e2563. [146] D.G. Duda, A.M. Duyverman, M. Kohno, M. Snuderl, E.J. Steller, D. Fukumura, R.K. Jain, Malignant cells facilitate lung metastasis by bringing their own soil, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 21677–21682. [147] G. Rappa, J. Mercapide, A. Lorico, Spontaneous formation of tumorigenic hybrids between breast cancer and multipotent stromal cells is a source of tumor heterogeneity, Am. J. Pathol. 180 (2012) 2504–2515. [148] J.W. Chang, H.L. Tsai, C.W. Chen, H.W. Yang, A.H. Yang, L.Y. Yang, P.S. Wang, Y.Y. Ng, T.L. Lin, O.K. Lee, Conditioned mesenchymal stem cells attenuate progression of chronic kidney disease through inhibition of epithelial-to-mesenchymal transition and immune modulation, J. Cell Mol. Med. 16 (2012) 2935–2949. [149] G. Grisendi, R. Bussolari, E. Veronesi, S. Piccinno, J.S. Burns, G. De Santis, P. Loschi, M. Pignatti, F. Di Benedetto, R. Ballarin, C. Di Gregorio, V. Guarneri, L. Piccinini, E.M. Horwitz, P. Paolucci, P. Conte, M. Dominici, Understanding tumor-stroma interplays for targeted therapies by armed mesenchymal stromal progenitors: the Mesenkillers, Am. J. Cancer Res. 1 (2011) 787–805. [150] A.B. Adesida, A. Mulet-Sierra, N.M. Jomha, Hypoxia mediated isolation and expansion enhances the chondrogenic capacity of bone marrow mesenchymal stromal cells, Stem Cell Res. Ther. 3 (2012) 9. [151] A. Malgieri, E. Kantzari, M.P. Patrizi, S. Gambardella, Bone marrow and umbilical cord blood human mesenchymal stem cells: state of the art, Int. J. Clin. Exp. Med. 3 (2010) 248–269. [152] E.D. O'Cearbhaill, M. Murphy, F. Barry, P.E. McHugh, V. Barron, Behavior of human mesenchymal stem cells in fibrin-based vascular tissue engineering constructs, Ann. Biomed. Eng. 38 (2010) 649–657. [153] A. Tocci, L. Forte, Mesenchymal stem cell: use and perspectives, Hematol. J. 4 (2003) 92–96. [154] R. Abdi, P. Fiorina, C.N. Adra, M. Atkinson, M.H. Sayegh, Immunomodulation by mesenchymal stem cells: a potential therapeutic strategy for type 1 diabetes, Diabetes 57 (2008) 1759–1767. [155] W. Vogel, F. Grunebach, C.A. Messam, L. Kanz, W. Brugger, H.J. Buhring, Heterogeneity among human bone marrow-derived mesenchymal stem cells and neural progenitor cells, Haematologica 88 (2003) 126–133. [156] V. Rasini, M. Dominici, T. Kluba, G. Siegel, G. Lusenti, H. Northoff, E.M. Horwitz, R. Schafer, Mesenchymal stromal/stem cells markers in the human bone marrow, Cytotherapy 15 (2013) 292–306. [157] M. Al-Nbaheen, R. Vishnubalaji, D. Ali, A. Bouslimi, F. Al-Jassir, M. Megges, A. Prigione, J. Adjaye, M. Kassem, A. Aldahmash, Human stromal (mesenchymal) stem cells from bone marrow, adipose tissue and skin exhibit differences in molecular phenotype and differentiation potential, Stem Cell Rev. 9 (2013) 32–43. [158] L. Song, N.E. Webb, Y. Song, R.S. Tuan, Identification and functional analysis of candidate genes regulating mesenchymal stem cell self-renewal and multipotency, Stem Cells 24 (2006) 1707–1718. [159] J.T. Watson, T. Foo, J. Wu, B.R. Moed, M. Thorpe, L. Schon, Z. Zhang, CD271 as a marker for mesenchymal stem cells in bone marrow versus umbilical cord blood, Cells Tissues Organs (2013). [160] A. Nasef, Y.Z. Zhang, C. Mazurier, S. Bouchet, M. Bensidhoum, S. Francois, N.C. Gorin, M. Lopez, D. Thierry, L. Fouillard, A. Chapel, Selected Stro-1-enriched bone marrow stromal cells display a major suppressive effect on lymphocyte proliferation, Int. J. Lab. Hematol. 31 (2009) 9–19.

N C O

R

R

E

C

T

[125] J. Plumas, L. Chaperot, M.J. Richard, J.P. Molens, J.C. Bensa, M.C. Favrot, Mesenchymal stem cells induce apoptosis of activated T cells, Leukemia 19 (2005) 1597–1604. [126] M. Krampera, S. Glennie, J. Dyson, D. Scott, R. Laylor, E. Simpson, F. Dazzi, Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide, Blood 101 (2003) 3722–3729. [127] I. Prigione, F. Benvenuto, P. Bocca, L. Battistini, A. Uccelli, V. Pistoia, Reciprocal interactions between human mesenchymal stem cells and gammadelta T cells or invariant natural killer T cells, Stem Cells 27 (2009) 693–702. [128] C. Bouffi, C. Bony, G. Courties, C. Jorgensen, D. Noel, IL-6-dependent PGE2 secretion by mesenchymal stem cells inhibits local inflammation in experimental arthritis, PloS one 5 (2010) e14247. [129] K. Nemeth, A. Keane-Myers, J.M. Brown, D.D. Metcalfe, J.D. Gorham, V.G. Bundoc, M.G. Hodges, I. Jelinek, S. Madala, S. Karpati, E. Mezey, Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma, Proc. Natl. Acad. Sci. U. S. A. 107 (2010) 5652–5657. [130] M.E. Groh, B. Maitra, E. Szekely, O.N. Koc, Human mesenchymal stem cells require monocyte-mediated activation to suppress alloreactive T cells, Exp. Hematol. 33 (2005) 928–934. [131] S. Ghannam, J. Pene, G. Torcy-Moquet, C. Jorgensen, H. Yssel, Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype, J. Immunol. 185 (2010) 302–312. [132] K. English, F.P. Barry, B.P. Mahon, Murine mesenchymal stem cells suppress dendritic cell migration, maturation and antigen presentation, Immunol. Lett. 115 (2008) 50–58. [133] M.P. De Miguel, S. Fuentes-Julian, A. Blazquez-Martinez, C.Y. Pascual, M.A. Aller, J. Arias, F. Arnalich-Montiel, Immunosuppressive properties of mesenchymal stem cells: advances and applications, Curr. Mol. Med. 12 (2012) 574–591. [134] R.S. Waterman, S.L. Tomchuck, S.L. Henkle, A.M. Betancourt, A new mesenchymal stem cell (MSC) paradigm: polarization into a pro-inflammatory MSC1 or an Immunosuppressive MSC2 phenotype, PloS one 5 (2010) e10088. [135] R.S. Waterman, S.L. Henkle, A.M. Betancourt, Mesenchymal stem cell 1 (MSC1)based therapy attenuates tumor growth whereas MSC2-treatment promotes tumor growth and metastasis, PloS one 7 (2012) e45590. [136] K. English, Mechanisms of mesenchymal stromal cell immunomodulation, Immunol. Cell Biol. 91 (2013) 19–26. [137] Z. Han, Z. Tian, G. Lv, L. Zhang, G. Jiang, K. Sun, C. Wang, X. Bu, R. Li, Y. Shi, M. Wu, L. Wei, Immunosuppressive effect of bone marrow-derived mesenchymal stem cells in inflammatory microenvironment favours the growth of B16 melanoma cells, J. Cell Mol. Med. 15 (2011) 2343–2352. [138] J.E. Talmadge, I.J. Fidler, AACR centennial series: the biology of cancer metastasis: historical perspective, Cancer Res. 70 (2010) 5649–5669. [139] A. van de Stolpe, On the origin and destination of cancer stem cells: a conceptual evaluation, Am. J. Cancer Res. 3 (2013) 107–116. [140] D.R. Hurst, D.R. Welch, A MSC-ing link in metastasis? Nat. Med. 13 (2007) 1289–1291. [141] P. Munoz, M.S. Iliou, M. Esteller, Epigenetic alterations involved in cancer stem cell reprogramming, Mol. Oncol. 6 (2012) 620–636. [142] Z. Yu, T.G. Pestell, M.P. Lisanti, R.G. Pestell, Cancer stem cells, Int. J. Biochem. Cell Biol. 44 (2012) 2144–2151. [143] H.J. Li, F. Reinhardt, H.R. Herschman, R.A. Weinberg, Cancer-stimulated mesenchymal stem cells create a carcinoma stem cell niche via prostaglandin E2 signaling, Cancer Discov. 2 (2012) 840–855.

U

1478 1479 1480 1481 1482 1483 1484 1485 1486 1487 1488 1489 1490 1491 1492 1493 1494 1495 1496 1497 1498 1499 1500 1501 1502 1503 1504 1505 1506 1507 1508 1509 1510 1511 1512 1513 1514 1515 1516 1517 1518 1519 1520 1521 1522 1523 1524 1525 1526 1527 1528 1529 1530 1531 1532 1588

15

Please cite this article as: P. Barcellos-de-Souza, et al., Tumor microenvironment: Bone marrow-mesenchymal stem cells as key players, Biochim. Biophys. Acta (2013), http://dx.doi.org/10.1016/j.bbcan.2013.10.004

1533 1534 1535 1536 1537 1538 1539 1540 1541 1542 1543 1544 1545 1546 1547 1548 1549 1550 1551 1552 1553 1554 1555 1556 1557 1558 1559 1560 1561 1562 1563 1564 1565 1566 1567 1568 1569 1570 1571 1572 1573 1574 1575 1576 1577 1578 1579 1580 1581 1582 1583 Q5 1584 1585 1586 1587

Tumor microenvironment: bone marrow-mesenchymal stem cells as key players.

Tumor progression is a multistep phenomenon in which tumor-associated stromal cells perform an intricate cross-talk with tumor cells, supplying approp...
2MB Sizes 0 Downloads 0 Views