Mesenchymal stem cells in the tumor microenvironment (Review)

  • Authors:
    • Jian Guan
    • Jie Chen
  • View Affiliations

  • Published online on: May 10, 2013     https://doi.org/10.3892/br.2013.103
  • Pages: 517-521
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Mesenchymal stem cells (MSCs) are non‑hematopoietic, multipotent cells, which are able to differentiate to bone, adipose and cartilage tissue. MSCs have the characteristic of migration to injured areas or tumor microenvironment following induction by chemokines or inflammatory factors. An increasing number of studies have reported that MSCs recruited to the tumor microenvironment play various roles in tumor cell development and tumor progression. In this study, we reviewed the studies related to the tumor‑promoting roles of MSCs from several aspects, such as increasing stemness of tumor cells, mediating migration, promoting angiogenesis, suppressing immune response and inducing drug resistance.

Contents

Introduction

Homing to the tumor microenvironment

Differentiation of MSCs

Functions in the tumor microenvironment

Conclusion

Introduction

Mesenchymal stem cells (MSCs) are pluripotent cells that give rise to a variety of connective tissue cell types (1,2). They are considered to be non-hematopoietic, multipotent cells, characterized by the expression of stromal cell markers (CD73, CD105, CD44, CD29 and CD90) in the absence of hematopoietic markers (CD34, CD45 and CD14) and endothelial markers (CD34, CD31 and vWF) (3). MSCs have the potential to differentiate along osteogenic, adipogenic and chondrogenic lineages when placed in the appropriate environments (4). An increasing number of studies have demonstrated that MSCs play complicated roles in carcinogenesis and tumor development by differentiating into more mature mesenchymal cells to support tumor parts and by playing special roles as a resident component in the tumor microenvironment. Therefore, comprehensive knowledge on the mechanism of interaction between MSCs and cancers is vital.

MSCs may be extractable from the bone marrow but may also be isolated from other mature tissues, such as skeletal muscle, umbilical cord, amniotic fluid, fetal liver and adipose tissue (57). MSCs are characteristically recruited to injured areas or hypoxic tumor microenvironments. Following intravenous administration, MSCs were identified mainly in tumors and had been cleared from normal tissue (8). Therefore, MSCs may serve as a platform for the delivery of biological agents to tumors. During the progression of carcinogenesis, when induced by chemokines of tumor cells to migrate to the area surrounding the tumor, MSCs are involved in supporting the neoplastic properties of cancer cells.

Homing to the tumor microenvironment

During the progression of carcinogenesis, recruitment of MSCs to tumors is reportedly due to the presence of soluble factors secreted by tumor cells, as well as the inflammation background or the hypoxic condition in the tumor microenvironment. Tumor cells secrete growth factors, cytokines and chemokines, such as IL-6 (9), IL-1β (10), transforming growth factor-β1 (TGF-β1) and SDF-1α (11,12). Signaling events promote MSC migration and survival. Additionally, several tumors exhibit hypoxia, a state of reduced oxygen that often parallels inflammation. Hypoxia plays an important role in perpetuating the inflammatory process in tumors, resulting in the generation of chemokines that are involved in immune cell and likely MSC migration to tumors. It was reported that, under hypoxic conditions (1.5% O2), breast cancer cells secrete high levels of IL-6, which serve to activate and attract MSCs (9).

Differentiation of MSCs

When MSCs arrive at the area surrounding the tumor, they may differentiate into more mature mesenchymal cells, such as cancer-associated fibroblasts (CAF) (13), macrophages (14) or endothelial cells (15,16). It was reported that, when treated with tumor-conditioned media, MSCs exhibited upregulation of cancer-associated fibroblast-associated genes and expression of markers specific to the myofibroblast lineage, such as α-SMA, vimentin and fibroblast-specific protein 1 (FSP1/S100A4) (17). Previous studies on the origin of fibroblasts in the tumor microenvironment demonstrated that the bone marrow and the adipose tissue surrounding tumor areas are the most common sources (18,19). In a study conducted by Quante et al (11), bone marrow-MSCs were labeled with EGFP, following which >70% of CAFs detected in the tumor microenvironment were identified as EGFP-positive, indicating their bone marrow origin. The majority of FSP-positive and fibroblast activation protein-positive CAFs originate from MSCs located in the bone marrow, whereas adipose-derived MSCs tend to differentiate into vascular and fibrovascular stromal cells (20). In addition, α-SMA-positive myofibroblasts originate from MSCs in the bone marrow (11,13). Other molecular markers, such as platelet-derived growth factor (PDGFR)-β was found to be positive in CAFs in the area surrounding the tumor, originating from pericyte progenitors in the bone marrow, which are responsible for regulating vascular stability and survival (21).

Following differentiation, MSCs exhibited a series of morphological changes, including elongated phenotype, reduced adhesion, cytoskeletal stiffening and increased migration (22). TGF-β participates in the process of differentiation from MSCs to CAFs, and coordinates the increase of α-SMA and the decrease of gelsolin to promote MSC differentiation (23). Under the stimulation of TGF-β, MSCs are hypomethylated and exhibit alterations in their gene expression profiles towards myofibroblast signature-expressing markers, such as α-SMA, tenascin-C and fibroblast surface protein (FSP), as well as an increased expression and secretion of growth-stimulating factors such as CCL5 and stromal cell-derived factor-1 (SDF-1) (11,17). The pathway involved is reportedly dependent on TGF-β signaling via Smad3 in MSCs (24). Adipose-derived MSCs may differentiate into α-SMA-expressing CAFs by expression of periostin, an extracellular matrix (ECM) protein, induced by A549 human lung adenocarcinoma cells (25). Under tumor conditions, MSCs may differentiate into hematopoietic cells, more precisely into macrophages, through reduced Ca2+ influx (14), or PDGFR-positive CAFs (26). In addition, it was reported that the exosome secreted by tumor cells may induce MSCs to express α-SMA and tumor-promoting factors SDF-1, VEGF, CCL5 and TGF-β (27).

Functions in the tumor microenvironment

In the tumor microenvironment, MSCs promote tumor cell development and progression. The tumor-MSCs crosstalk is complex. When stimulated by tumor cells, MSCs exhibit certain changes at the gene level. Data from a recent study demonstrated that following co-culture with MSCs and analysis by transcriptomic methods, tumor cells exhibited altered biological function of certain gene clusters, such as those related to the increase of metastatic ability, proliferation and chemoresistance (28). Therefore, upon interaction with MSCs, tumor cell functions are significantly supported on multiple levels.

Stemness

MSCs have multilineage potentials and also provide a beneficial microenvironment for tumor cells and enhance the stemness of tumor cells through multiple pathways. For example, in breast cancer, co-culture of MSCs and tumor cells promoted mammosphere formation, which is the three-dimensional culture morphology of cancer-initiating cells (29,30). MSCs regulate breast cancer stem cell self-renewal through cytokine loops involving IL-6 and CXCL7 (31). Carcinoma-associated MSCs (CA-MSCs) promoted tumor growth by increasing the number of cancer stem cells through bone morphogenetic protein signaling (32). Other pathways include the WNT, TGF-β (33) and the IL-6/JAK2/STAT3 signaling pathways (34,35). In addition, a pathway may be induced by tumor cells, which results in the enhancement of the stemness of tumor cells. Tumor cells may induce MSCs to express greatly elevated levels of PGE2 which, in combination with the cytokines that are also induced in the MSCs, contribute to the increase of the ALDH-positive fraction of cancer cells (36).

Migration

Epithelial-mesenchymal transition (EMT) is the early event of tumor cell migration. EMT renders podocytes motile, leading to their detachment from the basement membrane. Increased expression of the zinc finger proteins Snail and Slug, fibroblast-specific protein 1 (FSP1/S100A4), fibronectin and vimentin are correlated with mesenchymal phenotype and prometastatic behavior (37,38). MSCs actively recruited to the tumor stromal microenvironment may induce EMT in numerous tumors and actively increase the metastatic potential of tumor cells. When co-cultured with MSCs, breast cancer cells exhibited an enhanced expression of vimentin, Snail and Slug and a decreased expression of E-cadherin and β-catenin (30). MSCs promoted the production of lysyl oxidase from breast cancer cells, which in turn stimulated the Twist transcription factor, which mediates the MSCs-triggered EMT of carcinoma cells. Thus, MSCs enhance metastasis of cancer cells to the lungs and bones (39). It was also reported that paracrine transforming growth factor-β1 (TGF-β1) secreted by MSCs regulated the establishment of EMT in MCF7 cells by targeting the ZEB/miR-200 regulatory loop (40). The same phenomenon may be observed in hepatic carcinoma cells. In addition, when co-cultured with tumor cells, MSCs acquired the CAF markers tenascin-c and SDF-1 (41,42). During a study on pancreatic cancer, MSCs were found to regulate EMT and sphere formation in pancreatic cancer cells through a Notch-dependent mechanism (43).

In addition to EMT, MSCs may affect the migration of tumor cells through a variety of mechanisms, such as via the CXCL1/CXCL5-CXCR2 pathway (44), the ER and CXCR4 pathways (45) or the regulation of RANTES and IL-6 (46) in breast tumors. Breast cancer cells may also induce CCL5 expression in MSCs, which in turn enhances motility and the invasive potential of cancer cells (47). A previous study demonstrated that MSCs promote prostate cancer invasion and migration by upregulating MMP2/9 expression (48).

Angiogenesis

One of the effects of MSCs on tumor cells is angiogenesis. A previous study reported that the conditioned media from MSCs may increase sprouting of human umbilical vein endothelial cells due to VEGF production by MSCs (49). In vivo, co-injected MSCs directly supported the tumor vasculature by localizing close to the vascular walls and expressing CD31 (50). In vitro, when co-cultured with tumor cells, MSCs exhibited a distinct tendency to organize in clusters and form capillary-like structures (51). The soluble molecular factors secreted by MSCs, such as LIF, M-CSF, MIP-2, VEGF (50), IFN-γ and TNFα (52) promote angiogenesis. A previous study demonstrated that exosomes, which are 40–100 nm diameter vesicles secreted by MSCs, may enhance angiogenesis of tumors by VEGF and CXCR4 expression in tumor cells through the ERK1/2 and p38 MAPK pathways (53).

Immunosuppression

MSCs may affect the proliferation and maturation of T cells (54), B cells (55), dendritic cells (DCs) (56,57) and natural killer (NK) cells (58,59). Recent studies have focused on MSC-based immune therapy in allogeneic cell and organ transplantation (6062). Cytokines secreted by MSCs, such as IL-10 (60), TGF-β (63), nitric oxide (64), indoleamine 2,3-dioxygenase (65) and prostaglandin E2 (66) are involved in immunosuppression.

Furthermore, MSCs-mediated immunosuppression has been variously demonstrated in tumor development and progression. MSCs may protect breast cancer cells by increasing Tregs and reducing the activity of natural killer cells and cytotoxic T lymphocytes (67). In melanoma, the immunosuppressive function of MSCs was elicited by IFN-γ and TNF-α. These cytokine combinations led to the expression of inducible nitric oxide synthase by MSCs (68). The inflammatory background in prostate cancer elicited the upregulation of TGF-β in MSCs, which enabled prostate cancer cells to escape from immune surveillance (69).

Chemotherapy resistance

The mechanisms involved in classic chemotherapy resistance include enhanced activity of positive regulators of cell proliferation, such as oncogenes, loss of tumor suppressor gene function, inactivation of cell death or enhancement of survival functions and activation of telomerase. Besides the classically defined causes of drug resistance, environment mediated-drug resistance (EMDR) arises from an adaptive, reciprocal signaling dialogue between tumor cells and the surrounding microenvironment. EMDR may be subdivided into two categories: soluble factor-mediated drug resistance, which is induced by cytokines, chemokines and growth factors secreted by fibroblast-like tumor stroma; and cell adhesion-mediated drug resistance, which is mediated by the adhesion of tumor cell integrins to stromal fibroblasts or to components of the ECM, such as fibronectin, laminin and collagen (70). MSCs are associated with EMDR by secreting soluble molecular factors such as SDF-1, IL-6, NO, IL-3, G-CSF, M-CSF and GM-CSF and by activating proliferation pathways in tumor cells and producing ECM to protect tumor cells against chemotherapy drugs. MSCs utilize autophagy to recycle macromolecules and synthesize antiapoptotic factors to facilitate the survival and growth of surrounding tumor cells (71). In colorectal carcinoma, MSCs stimulated survival of tumor cells through the release of soluble NRG1, activating the HER2/HER3-dependent PI3K/AKT signaling cascade (72). Platinum-based chemotherapy in breast cancer may induce MSCs to secrete two unique fatty acids that confer resistance to chemotherapy (73).

Conclusion

MSCs exert multiple effects on tumor development and progression, by increasing stemness of tumor cells, mediating migration, promoting angiogenesis, suppressing immune response and inducing drug resistance. In-depth understanding of these actions may provide information regarding the biological behavior of tumors and set the basis for the design of an MSCs-based therapeutic method for carcinomas and immunological diseases. The tumor-promoting molecules secreted by MSCs or the pathway activated by MSCs in tumor cells may enrich the list of potential targets for molecular therapy.

Acknowledgements

The study was supported by The National Nature Science Foundation of China (grant no. 81172334).

References

1. 

Jiang Y, Jahagirdar BN, Reinhardt RL, et al: Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 418:41–49. 2002. View Article : Google Scholar : PubMed/NCBI

2. 

Bianco P, Robey PG and Simmons PJ: Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell. 2:313–319. 2008. View Article : Google Scholar : PubMed/NCBI

3. 

Dominici M, Le Blanc K, Mueller I, et al: Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 8:315–317. 2006. View Article : Google Scholar

4. 

Tropel P, Noel D, Platet N, Legrand P, Benabid AL and Berger F: Isolation and characterisation of mesenchymal stem cells from adult mouse bone marrow. Exp Cell Res. 295:395–406. 2004. View Article : Google Scholar : PubMed/NCBI

5. 

Zuk PA, Zhu M, Mizuno H, et al: Multilineage cells from human adipose tissue: implications for cell-based therapies. Tissue Eng. 7:211–228. 2001. View Article : Google Scholar : PubMed/NCBI

6. 

Jones EA, English A, Henshaw K, et al: Enumeration and phenotypic characterization of synovial fluid multipotential mesenchymal progenitor cells in inflammatory and degenerative arthritis. Arthritis Rheum. 50:817–827. 2004. View Article : Google Scholar

7. 

Williams JT, Southerland SS, Souza J, Calcutt AF and Cartledge RG: Cells isolated from adult human skeletal muscle capable of differentiating into multiple mesodermal phenotypes. Am Surg. 65:22–26. 1999.PubMed/NCBI

8. 

Studeny M, Marini FC, Champlin RE, Zompetta C, Fidler IJ and Andreeff M: Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res. 62:3603–3608. 2002.PubMed/NCBI

9. 

Rattigan Y, Hsu JM, Mishra PJ, Glod J and Banerjee D: Interleukin 6 mediated recruitment of mesenchymal stem cells to the hypoxic tumor milieu. Exp Cell Res. 316:3417–3424. 2010. View Article : Google Scholar : PubMed/NCBI

10. 

Tu S, Bhagat G, Cui G, et al: Overexpression of interleukin-1beta induces gastric inflammation and cancer and mobilizes myeloid-derived suppressor cells in mice. Cancer Cell. 14:408–419. 2008. View Article : Google Scholar : PubMed/NCBI

11. 

Quante M, Tu SP, Tomita H, et al: Bone marrow-derived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth. Cancer Cell. 19:257–272. 2011. View Article : Google Scholar : PubMed/NCBI

12. 

Gao H, Priebe W, Glod J and Banerjee D: Activation of signal transducers and activators of transcription 3 and focal adhesion kinase by stromal cell-derived factor 1 is required for migration of human mesenchymal stem cells in response to tumor cell-conditioned medium. Stem Cells. 27:857–865. 2009. View Article : Google Scholar

13. 

Direkze NC, Hodivala-Dilke K, Jeffery R, et al: Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts. Cancer Res. 64:8492–8495. 2004. View Article : Google Scholar : PubMed/NCBI

14. 

Zhang L, Tang A, Zhou Y, et al: Tumor-conditioned mesenchymal stem cells display hematopoietic differentiation and diminished influx of Ca2+. Stem Cells Dev. 21:1418–1428. 2012. View Article : Google Scholar : PubMed/NCBI

15. 

Zhou Y, Guan X, Wang H, et al: Hypoxia induces osteogenic/angiogenic responses of bone marrow-derived mesenchymal stromal cells seeded on bone-derived scaffolds via ERK1/2 and p38 pathways. Biotechnol Bioeng. Jan 7–2013.(Epub ahead of print).

16. 

Janeczek Portalska K, Leferink A, Groen N, et al: Endothelial differentiation of mesenchymal stromal cells. PLoS One. 7:e468422012.PubMed/NCBI

17. 

Mishra PJ, Mishra PJ, Humeniuk R, Medina DJ, et al: Carcinoma-associated fibroblast-like differentiation of human mesenchymal stem cells. Cancer Res. 68:4331–4339. 2008. View Article : Google Scholar : PubMed/NCBI

18. 

Zuk PA, Zhu M, Ashjian P, et al: Human adipose tissue is a source of multipotent stem cells. Mol Biol Cell. 13:4279–4295. 2002.PubMed/NCBI

19. 

Mosna F, Sensebé L and Krampera M: Human bone marrow and adipose tissue mesenchymal stem cells: a user’s guide. Stem Cells Dev. 19:1449–1470. 2010.

20. 

Kidd S, Spaeth E, Watson K, et al: Origins of the tumor microenvironment: quantitative assessment of adipose-derived and bone marrow-derived stroma. PLoS One. 7:e305632012. View Article : Google Scholar : PubMed/NCBI

21. 

Song S, Ewald AJ, Stallcup W, Werb Z and Bergers G: PDGFRbeta+ perivascular progenitor cells in tumours regulate pericyte differentiation and vascular survival. Nat Cell Biol. 7:870–879. 2005.

22. 

McGrail DJ, Ghosh D, Quach ND and Dawson MR: Differential mechanical response of mesenchymal stem cells and fibroblasts to tumor-secreted soluble factors. PLoS One. 7:e332482012. View Article : Google Scholar : PubMed/NCBI

23. 

Wang D, Park JS, Chu JS, et al: Proteomic profiling of bone marrow mesenchymal stem cells upon transforming growth factor beta1 stimulation. J Biol Chem. 279:43725–43734. 2004. View Article : Google Scholar : PubMed/NCBI

24. 

Jotzu C, Alt E, Welte G, et al: Adipose tissue-derived stem cells differentiate into carcinoma-associated fibroblast-like cells under the influence of tumor-derived factors. Anal Cell Pathol (Amst). 33:61–79. 2010. View Article : Google Scholar

25. 

Heo SC, Lee KO, Shin SH, et al: Periostin mediates human adipose tissue-derived mesenchymal stem cell-stimulated tumor growth in a xenograft lung adenocarcinoma model. Biochim Biophys Acta. 1813:2061–2070. 2011. View Article : Google Scholar : PubMed/NCBI

26. 

Shinagawa K, Kitadai Y, Tanaka M, et al: 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. 132:813–823. 2012. View Article : Google Scholar

27. 

Cho JA, Park H, Lim EH and Lee KW: Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int J Oncol. 40:130–138. 2012.PubMed/NCBI

28. 

Lis R, Touboul C, Raynaud CM, et al: Mesenchymal cell interaction with ovarian cancer cells triggers pro-metastatic properties. PLoS One. 7:e383402012. View Article : Google Scholar : PubMed/NCBI

29. 

Yan XL, Fu CJ, Chen L, et al: Mesenchymal stem cells from primary breast cancer tissue promote cancer proliferation and enhance mammosphere formation partially via EGF/EGFR/Akt pathway. Breast Cancer Res Treat. 132:153–164. 2012. View Article : Google Scholar : PubMed/NCBI

30. 

Klopp AH, Lacerda L, Gupta A, et al: Mesenchymal stem cells promote mammosphere formation and decrease E-cadherin in normal and malignant breast cells. PLoS One. 5:e121802010. View Article : Google Scholar : PubMed/NCBI

31. 

Liu S, Ginestier C, Ou SJ, et al: Breast cancer stem cells are regulated by mesenchymal stem cells through cytokine networks. Cancer Res. 71:614–624. 2011. View Article : Google Scholar : PubMed/NCBI

32. 

McLean K, Gong Y, Choi Y, et al: Human ovarian carcinoma-associated mesenchymal stem cells regulate cancer stem cells and tumorigenesis via altered BMP production. J Clin Invest. 121:3206–3219. 2011. View Article : Google Scholar : PubMed/NCBI

33. 

Nishimura K, Semba S, Aoyagi K, Sasaki H and Yokozaki H: Mesenchymal stem cells provide an advantageous tumor microenvironment for the restoration of cancer stem cells. Pathobiology. 79:290–306. 2012. View Article : Google Scholar : PubMed/NCBI

34. 

Hsu HS, Lin JH, Hsu TW, et al: Mesenchymal stem cells enhance lung cancer initiation through activation of IL-6/JAK2/STAT3 pathway. Lung Cancer. 75:167–177. 2012. View Article : Google Scholar : PubMed/NCBI

35. 

Tsai KS, Yang SH, Lei YP, et al: Mesenchymal stem cells promote formation of colorectal tumors in mice. Gastroenterology. 141:1046–1056. 2011. View Article : Google Scholar : PubMed/NCBI

36. 

Li HJ, Reinhardt F, Herschman HR and Weinberg RA: Cancer-stimulated mesenchymal stem cells create a carcinoma stem-cell niche via prostaglandin E2 signaling. Cancer Discov. 2:840–855. 2012. View Article : Google Scholar : PubMed/NCBI

37. 

Thiery JP: Epithelial-mesenchymal transitions in development and pathologies. Curr Opin Cell Biol. 15:740–746. 2003. View Article : Google Scholar : PubMed/NCBI

38. 

Kalluri R and Weinberg RA: The basics of epithelial-mesenchymal transition. J Clin Invest. 119:1420–1428. 2009. View Article : Google Scholar : PubMed/NCBI

39. 

El-Haibi CP, Bell GW, Zhang J, et al: Critical role for lysyl oxidase in mesenchymal stem cell-driven breast cancer malignancy. Proc Natl Acad Sci USA. 109:17460–17465. 2012. View Article : Google Scholar : PubMed/NCBI

40. 

Xu Q, Wang L, Li H, et al: 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-β. Int J Oncol. 41:959–968. 2012.PubMed/NCBI

41. 

Bhattacharya SD, Mi Z, Talbot LJ, Guo H and Kuo PC: Human mesenchymal stem cell and epithelial hepatic carcinoma cell lines in admixture: concurrent stimulation of cancer-associated fibroblasts and epithelial-to-mesenchymal transition markers. Surgery. 152:449–454. 2012. View Article : Google Scholar

42. 

Jing Y, Han Z, Liu Y, et al: Mesenchymal stem cells in inflammation microenvironment accelerates hepatocellular carcinoma metastasis by inducing epithelial-mesenchymal transition. PLoS One. 7:e432722012. View Article : Google Scholar

43. 

Kabashima-Niibe A, Higuchi H, Takaishi H, et al: Mesenchymal stem cells regulate epithelial-to-mesenchymal transition and tumor progression of pancreatic cancer cells. Cancer Sci. 104:157–164. 2012. View Article : Google Scholar

44. 

Halpern JL, Kilbarger A and Lynch CC: Mesenchymal stem cells promote mammary cancer cell migration in vitro via the CXCR2 receptor. Cancer Lett. 308:91–99. 2011. View Article : Google Scholar : PubMed/NCBI

45. 

Rhodes LV, Antoon JW, Muir SE, Elliott S, Beckman BS and Burow ME: Effects of human mesenchymal stem cells on ER-positive human breast carcinoma cells mediated through ER-SDF-1/CXCR4 crosstalk. Mol Cancer. 9:2952010. View Article : Google Scholar : PubMed/NCBI

46. 

Gallo M, De Luca A, Lamura L and Normanno N: Zoledronic acid blocks the interaction between mesenchymal stem cells and breast cancer cells: implications for adjuvant therapy of breast cancer. Ann Oncol. 23:597–604. 2012. View Article : Google Scholar : PubMed/NCBI

47. 

Karnoub AE, Dash AB, Vo AP, et al: Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 449:557–563. 2007. View Article : Google Scholar : PubMed/NCBI

48. 

Ye H, Cheng J, Tang Y, et al: Human bone marrow-derived mesenchymal stem cells produced TGFbeta contributes to progression and metastasis of prostate cancer. Cancer Invest. 30:513–518. 2012. View Article : Google Scholar : PubMed/NCBI

49. 

Beckermann BM, Kallifatidis G, Groth A, et al: VEGF expression by mesenchymal stem cells contributes to angiogenesis in pancreatic carcinoma. Br J Cancer. 99:622–631. 2008. View Article : Google Scholar : PubMed/NCBI

50. 

Suzuki K, Sun R, Origuchi M, et al: Mesenchymal stromal cells promote tumor growth through the enhancement of neovascularization. Mol Med. 17:579–587. 2011. View Article : Google Scholar : PubMed/NCBI

51. 

Comsa S, Ciuculescu F and Raica M: Mesenchymal stem cell-tumor cell cooperation in breast cancer vasculogenesis. Mol Med Rep. 5:1175–1180. 2012.PubMed/NCBI

52. 

Liu Y, Han ZP, Zhang SS, et al: Effects of inflammatory factors on mesenchymal stem cells and their role in the promotion of tumor angiogenesis in colon cancer. J Biol Chem. 286:25007–25015. 2011. View Article : Google Scholar : PubMed/NCBI

53. 

Zhu W, Huang L, Li Y, et al: Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo. Cancer Lett. 315:28–37. 2012. View Article : Google Scholar : PubMed/NCBI

54. 

Krampera M, Glennie S, Dyson J, et al: Bone marrow mesenchymal stem cells inhibit the response of naive and memory antigen-specific T cells to their cognate peptide. Blood. 101:3722–3729. 2003. View Article : Google Scholar : PubMed/NCBI

55. 

Tabera S, Perez-Simon JA, Diez-Campelo M, et al: The effect of mesenchymal stem cells on the viability, proliferation and differentiation of B-lymphocytes. Haematologica. 93:1301–1309. 2008. View Article : Google Scholar : PubMed/NCBI

56. 

Jiang XX, Zhang Y, Liu B, et al: Human mesenchymal stem cells inhibit differentiation and function of monocyte-derived dendritic cells. Blood. 105:4120–4126. 2005. View Article : Google Scholar : PubMed/NCBI

57. 

Nauta AJ, Kruisselbrink AB, Lurvink E, Willemze R and Fibbe WE: Mesenchymal stem cells inhibit generation and function of both CD34+-derived and monocyte-derived dendritic cells. J Immunol. 177:2080–2087. 2006. View Article : Google Scholar : PubMed/NCBI

58. 

Sotiropoulou PA, Perez SA, Gritzapis AD, Baxevanis CN and Papamichail M: Interactions between human mesenchymal stem cells and natural killer cells. Stem Cells. 24:74–85. 2006. View Article : Google Scholar : PubMed/NCBI

59. 

Spaggiari GM and Moretta L: Cellular and molecular interactions of mesenchymal stem cells in innate immunity. Immunol Cell Biol. 91:27–31. 2013. View Article : Google Scholar : PubMed/NCBI

60. 

Batten P, Sarathchandra P, Antoniw JW, et al: Human mesenchymal stem cells induce T cell anergy and downregulate T cell allo-responses via the TH2 pathway: relevance to tissue engineering human heart valves. Tissue Eng. 12:2263–2273. 2006. View Article : Google Scholar : PubMed/NCBI

61. 

Casiraghi F, Perico N and Remuzzi G: Mesenchymal stromal cells to promote solid organ transplantation tolerance. Curr Opin Organ Transplant. 18:51–58. 2013. View Article : Google Scholar : PubMed/NCBI

62. 

Reinders ME, de Fijter JW, Roelofs H, et al: Autologous bone marrow-derived mesenchymal stromal cells for the treatment of allograft rejection after renal transplantation: results of a phase I study. Stem Cells Transl Med. 2:107–111. 2013. View Article : Google Scholar

63. 

Groh ME, Maitra B, Szekely E and Koc ON: Human mesenchymal stem cells require monocyte-mediated activation to suppress alloreactive T cells. Exp Hematol. 33:928–934. 2005. View Article : Google Scholar : PubMed/NCBI

64. 

Sato K, Ozaki K, Oh I, et al: Nitric oxide plays a critical role in suppression of T-cell proliferation by mesenchymal stem cells. Blood. 109:228–234. 2007. View Article : Google Scholar : PubMed/NCBI

65. 

Meisel R, Zibert A, Laryea M, Gobel U, Daubener W and Dilloo D: Human bone marrow stromal cells inhibit allogeneic T-cell responses by indoleamine 2,3-dioxygenase-mediated tryptophan degradation. Blood. 103:4619–4621. 2004. View Article : Google Scholar : PubMed/NCBI

66. 

Aggarwal S and Pittenger MF: Human mesenchymal stem cells modulate allogeneic immune cell responses. Blood. 105:1815–1822. 2005. View Article : Google Scholar : PubMed/NCBI

67. 

Patel SA, Meyer JR, Greco SJ, Corcoran KE, Bryan M and Rameshwar P: Mesenchymal stem cells protect breast cancer cells through regulatory T cells: role of mesenchymal stem cell-derived TGF-beta. J Immunol. 184:5885–5894. 2010. View Article : Google Scholar : PubMed/NCBI

68. 

Han Z, Tian Z, Lv G, et al: Immunosuppressive effect of bone marrow-derived mesenchymal stem cells in inflammatory microenvironment favours the growth of B16 melanoma cells. J Cell Mol Med. 15:2343–2352. 2011. View Article : Google Scholar : PubMed/NCBI

69. 

Cheng J, Li L, Liu Y, Wang Z, Zhu X and Bai X: Interleukin-1α induces immunosuppression by mesenchymal stem cells promoting the growth of prostate cancer cells. Mol Med Rep. 6:955–960. 2012.

70. 

Meads MB, Gatenby RA and Dalton WS: Environment-mediated drug resistance: a major contributor to minimal residual disease. Nat Rev Cancer. 9:665–674. 2009. View Article : Google Scholar : PubMed/NCBI

71. 

Sanchez CG, Penfornis P, Oskowitz AZ, et al: Activation of autophagy in mesenchymal stem cells provides tumor stromal support. Carcinogenesis. 32:964–972. 2011. View Article : Google Scholar : PubMed/NCBI

72. 

De Boeck A, Pauwels P, Hensen K, et al: Bone marrow-derived mesenchymal stem cells promote colorectal cancer progression through paracrine neuregulin 1/HER3 signalling. Gut. 62:550–560. 2013.PubMed/NCBI

73. 

Roodhart JM, Daenen LG, Stigter EC, et al: Mesenchymal stem cells induce resistance to chemotherapy through the release of platinum-induced fatty acids. Cancer Cell. 20:370–383. 2011. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-August 2013
Volume 1 Issue 4

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guan J and Guan J: Mesenchymal stem cells in the tumor microenvironment (Review). Biomed Rep 1: 517-521, 2013
APA
Guan, J., & Guan, J. (2013). Mesenchymal stem cells in the tumor microenvironment (Review). Biomedical Reports, 1, 517-521. https://doi.org/10.3892/br.2013.103
MLA
Guan, J., Chen, J."Mesenchymal stem cells in the tumor microenvironment (Review)". Biomedical Reports 1.4 (2013): 517-521.
Chicago
Guan, J., Chen, J."Mesenchymal stem cells in the tumor microenvironment (Review)". Biomedical Reports 1, no. 4 (2013): 517-521. https://doi.org/10.3892/br.2013.103