Mesenchymal stem cell transition to tumor-associated fibroblasts contributes to fibrovascular network expansion and tumor progression

Erika L Spaeth, Jennifer L Dembinski, A Kate Sasser, Keri Watson, Ann Klopp, Brett Hall, Michael Andreeff, Frank Marini, Erika L Spaeth, Jennifer L Dembinski, A Kate Sasser, Keri Watson, Ann Klopp, Brett Hall, Michael Andreeff, Frank Marini

Abstract

Background: Tumor associated fibroblasts (TAF), are essential for tumor progression providing both a functional and structural supportive environment. TAF, known as activated fibroblasts, have an established biological impact on tumorigenesis as matrix synthesizing or matrix degrading cells, contractile cells, and even blood vessel associated cells. The production of growth factors, cytokines, chemokines, matrix-degrading enzymes, and immunomodulatory mechanisms by these cells augment tumor progression by providing a suitable environment. There are several suggested origins of the TAF including tissue-resident, circulating, and epithelial-to-mesenchymal-transitioned cells.

Methodology/principal findings: We provide evidence that TAF are derived from mesenchymal stem cells (MSC) that acquire a TAF phenotype following exposure to or systemic recruitment into adenocarcinoma xenograft models including breast, pancreatic, and ovarian. We define the MSC derived TAF in a xenograft ovarian carcinoma model by the immunohistochemical presence of 1) fibroblast specific protein and fibroblast activated protein; 2) markers phenotypically associated with aggressiveness, including tenascin-c, thrombospondin-1, and stromelysin-1; 3) production of pro-tumorigenic growth factors including hepatocyte growth factor, epidermal growth factor, and interleukin-6; and 4) factors indicative of vascularization, including alpha-smooth muscle actin, desmin, and vascular endothelial growth factor. We demonstrate that under long-term tumor conditioning in vitro, MSC express TAF-like proteins. Additionally, human MSC but not murine MSC stimulated tumor growth primarily through the paracrine production of secreted IL6.

Conclusions/significance: Our results suggest the dependence of in vitro Skov-3 tumor cell proliferation is due to the presence of tumor-stimulated MSC secreted IL6. The subsequent TAF phenotype arises from the MSC which ultimately promotes tumor growth through the contribution of microvascularization, stromal networks, and the production of tumor-stimulating paracrine factors.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. In admixtures of tumor and…
Figure 1. In admixtures of tumor and MSC, MSC exhibit four attributes of TAF.
(A) Initially, MSC do not express all TAF markers. As shown by western, huMSC, prior to Skov-3 tumor exposure, express low levels of α-SMA, FAP and desmin and are negative for the expression of, FSP, TSP-1, and Tn-C. Following 16 days exposure to Skov-3-conditioned medium, the MSC express Tn-C, TSP-1, FSP and increase expression of α-SMA, FAP and desmsin. (B) The TAF is a fibroblastic cell that has four defining characteristics, which together, distinguish it from the normal fibroblast: The fibroblastic nature of the cell (red; Fibroblast activation protein and fibroblast specific protein); the aggressive/invasive nature of the cell defined by the secreted proteins (green; TSP-1, Tn-C and SL1); the vascularization potential of the cell (blue; α-SMA, desmin and VEGF); and the growth factors secreted by the cell to aid in tumor growth and development (yellow; EGF, HGF, IL-6 and bFGF).
Figure 2. Admixed Skov-3/MSC xenografts express fibroblast…
Figure 2. Admixed Skov-3/MSC xenografts express fibroblast markers and invasive markers indicative of TAF.
IHC staining for markers that define fibroblast presence are visible in the Skov-3/MSC (1∶1) admixed xenografts but not in Skov-3-only xenografts. Fibroblast markers FAP (A) and FSP (B) are both present throughout the admixed tumors. The three tissue remodeling proteins Tn-C (C), TSP-1 (D) and SL-1 (E) are detectable within the admixed tumors. These two categories cover the first two characteristics of a TAF, and are observed only in tumors exposed to MSC. Human-specific, mouse non-cross-reactive antibodies allow identification of the human MSC-contributory components within the tumor microenvironment.
Figure 3. Admixed Skov-3/MSC xenografts express provascularization…
Figure 3. Admixed Skov-3/MSC xenografts express provascularization markers.
One characteristic of the TAF is their ability to form fibrovascular networks, which include vessels and the architectural foundation in which they lay. IHC staining reveals structural proteins α-SMA (A) and desmin (B) as well as factors suggestive of tumor vascularization, including VEGF (C) in the presence of admixed MSC but not in tumors that received no MSC (far right panel). HGF (D) is found within the stromal compartment of the tumor. A strong delineation separating the tumor and stromal compartments is formed when the admixed xenograft is stained for HGF. Likewise, EGF (E) is highly expressed in the stromal regions of the admixed Skov-3 tumor. (F) IL-6 staining is expressed in the Skov-3 alone tumors, however the expression is increased in the admixed tumors.
Figure 4. huMSC secreted growth factors promote…
Figure 4. huMSC secreted growth factors promote growth of Skov-3 tumors.
The TAF phenotype includes secretion of pro-tumorigenic growth factors. (A) Supported by the secretion of growth factors by the huMSC. In vivo growth of xenograft Skov-3 tumors steadily progress but when mixed at a 1∶1 ratio with MSC rapid growth ensued after day 65. At day 91, the Skov-3/MSC 50/50 tumors were significantly larger than the Skov-3 alone (P<0.05). (B) Skov-3 tumor cell growth is species dependent. The Skov-3 ovarian tumor cells were growth in co-culture with huMSC, and several muMSC including cells isolated from balb/c and C57 mice, and a fibroblast cell line, 3T3. Growth is graphed as fold change relative to normal Skov-3 proliferation. The only cell line that produced adequate factors to induce cell growth is the huMSC (red circles). The huMSC induced Skov-3 growth significantly (P<0.01) better than the other cell lines. (C) Naïve MSC produce basal levels of IL-6, TGF-β, VEGF and HGF. All secreted factors with the exception of HGF are significantly increased upon stimulation with Skov-3 CM (P<0.001).
Figure 5. Growth factors are critical to…
Figure 5. Growth factors are critical to Skov-3 tumor progression.
(A) An in vitro 3D tumor growth assay (TGA) at day 8. Briefly, RFP-labeled Skov-3 tumor cells were mixed with huMSC cells, huMSC conditioned media (CM) or various recombinant cytokines (FGF, HGF, TGF-β, VEGF, EGF, IL-6) in a 3D assays (as described in the materials and methods). EGF (P<0.05) and IL6 (P<0.01) significantly increased the proliferation of Skov-3 cells compared to the Skov-3 only, but less than the MSC (P<0.01) induced Skov-3 proliferation. (B) Immunoprecipitation of IL-6 from the Skov-3/MSC co-culture medium over an 8 day period reveals significantly reduced growth of the Skov-3 (P<0.01) cells as compared with the Skov-3/MSCco-culture. Cell growth is graphed as fold change relative to normal Skov-3 proliferation.

References

    1. Wels J, Kaplan RN, Rafii S, Lyden D. Migratory neighbors and distant invaders: Tumor-associated niche cells. Genes Dev. 2008;22:559–574.
    1. Udagawa T, Puder M, Wood M, Schaefer BC, D'Amato RJ. Analysis of tumor-associated stromal cells using SCID GFP transgenic mice: contribution of local and bone marrow-derived host cells. FASEB J. 2006;20:95–102.
    1. Koyama H, Kobayashi N, Harada M, Takeoka M, Kawai Y, et al. Significance of tumor-associated stroma in promotion of intratumoral lymphangiogenesis: pivotal role of a hyaluronan-rich tumor microenvironment. Am J Pathol. 2008;172:179–193.
    1. Dong J, Grunstein J, Tejada M, Peale F, Frantz G, et al. VEGF-null cells require PDGFR alpha signaling-mediated stromal fibroblast recruitment for tumorigenesis. EMBO J. 2004;23:2800–2810.
    1. Jodele S, Chantrain CF, Blavier L, Lutzko C, Crooks GM, et al. The contribution of bone marrow-derived cells to the tumor vasculature in neuroblastoma is matrix metalloproteinase-9 dependent. Cancer Res. 2005;65:3200–3208.
    1. Ogawa M, LaRue AC, Drake CJ. Hematopoietic origin of fibroblasts/myofibroblasts: Its pathophysiologic implications. Blood. 2006;108:2893–2896.
    1. Tsujino T, Seshimo I, Yamamoto H, Ngan CY, Ezumi K, et al. Stromal myofibroblasts predict disease recurrence for colorectal cancer. Clin Cancer Res. 2007;13:2082–2090.
    1. Yazhou C, Wenlv S, Weidong Z, Licun W. Clinicopathological significance of stromal myofibroblasts in invasive ductal carcinoma of the breast. Tumour Biol. 2004;25:290–295.
    1. Silzle T, Randolph GJ, Kreutz M, Kunz-Schughart LA. The fibroblast: sentinel cell and local immune modulator in tumor tissue. Int J Cancer. 2004;108:173–180.
    1. Kunz-Schughart LA, Knuechel R. Tumor-associated fibroblasts (part I): Active stromal participants in tumor development and progression? Histol Histopathol. 2002;17:599–621.
    1. Studeny M, Marini FC, Champlin RE, Zompetta C, Fidler IJ, et al. Bone marrow-derived mesenchymal stem cells as vehicles for interferon-beta delivery into tumors. Cancer Res. 2002;62:3603–3608.
    1. Studeny M, Marini FC, Dembinski JL, Zompetta C, Cabreira-Hansen M, et al. Mesenchymal stem cells: potential precursors for tumor stroma and targeted-delivery vehicles for anticancer agents. J Natl Cancer Inst. 2004;96:1593–1603.
    1. Spaeth E, Klopp A, Dembinski J, Andreeff M, Marini F. Inflammation and tumor microenvironments: defining the migratory itinerary of mesenchymal stem cells. Gene Ther 2008
    1. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315:1650–1659.
    1. Haddow A. Molecular repair, wound healing, and carcinogenesis: tumor production a possible overhealing? Adv Cancer Res. 1972;16:181–234.
    1. Nakamizo A, Marini F, Amano T, Khan A, Studeny M, et al. Human bone marrow-derived mesenchymal stem cells in the treatment of gliomas. Cancer Res. 2005;65:3307–3318.
    1. Kanehira M, Xin H, Hoshino K, Maemondo M, Mizuguchi H, et al. Targeted delivery of NK4 to multiple lung tumors by bone marrow-derived mesenchymal stem cells. Cancer Gene Ther. 2007;14:894–903.
    1. Xin H, Kanehira M, Mizuguchi H, Hayakawa T, Kikuchi T, et al. Targeted-Delivery of CX3CL1 to Multiple Lung Tumors by Mesenchymal Stem Cells. Stem Cells 2007
    1. Sonabend AM, Ulasov IV, Tyler MA, Rivera AA, Mathis JM, et al. Mesenchymal Stem Cells Effectively Deliver an Oncolytic Adenovirus to Intracranial Glioma. Stem Cells 2008
    1. Emura M, Ochiai A, Horino M, Arndt W, Kamino K, et al. Development of myofibroblasts from human bone marrow mesenchymal stem cells cocultured with human colon carcinoma cells and TGF beta 1 [3]. In Vitro Cell Dev Biol Anim. 2000;36:77–80.
    1. Hall B, Andreeff M, Marini F. The participation of mesenchymal stem cells in tumor stroma formation and their application as targeted-gene delivery vehicles. Handb Exp Pharmacol. 2007;(180):263–283.
    1. Hall B, Dembinski J, Sasser AK, Studeny M, Andreeff M, et al. Mesenchymal stem cells in cancer: tumor-associated fibroblasts and cell-based delivery vehicles. Int J Hematol. 2007;86:8–16.
    1. Ruiter D, Bogenrieder T, Elder D, Herlyn M. Melanoma-stroma interactions: Structural and functional aspects. Lancet Oncol. 2002;3:35–43.
    1. Ball SG, Shuttleworth AC, Kielty CM. Direct cell contact influences bone marrow mesenchymal stem cell fate. Int J Biochem Cell Biol. 2004;36:714–727.
    1. Kinner B, Zaleskas JM, Spector M. Regulation of smooth muscle actin expression and contraction in adult human mesenchymal stem cells. Exp Cell Res. 2002;278:72–83.
    1. Bexell D, Gunnarsson S, Tormin A, Darabi A, Gisselsson D, et al. Bone Marrow Multipotent Mesenchymal Stroma Cells Act as Pericyte-like Migratory Vehicles in Experimental Gliomas. Mol Ther 2008
    1. Jeon ES, Moon HJ, Lee MJ, Song HY, Kim YM, et al. Cancer-derived lysophosphatidic acid stimulates differentiation of human mesenchymal stem cells to myofibroblast-like cells. Stem Cells. 2008;26:789–797.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini FC, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–317.
    1. Javazon EH, Beggs KJ, Flake AW. Mesenchymal stem cells: Paradoxes of passaging. Exp Hematol. 2004;32:414–425.
    1. Studeny M, Marini FC, Zompetta C, Champlin RE, Filder IJ, et al. Bone marrow derived mesenchymal stem cells serve as precursors for stromal fibroblasts in malignant tumors and show potential for cancer therapy. Blood. 2001;98
    1. Hung SC, Deng WP, Yang WK, Liu RS, Lee CC, et al. Mesenchymal stem cell targeting of microscopic tumors and tumor stroma development monitored by noninvasive in vivo positron emission tomography imaging. Clin Cancer Res. 2005;11:7749–7756.
    1. Annabi B, Naud E, Lee Y-, Eliopoulos N, Galipeau J. Vascular progenitors derived from murine bone marrow stromal cells are regulated by fibroblast growth factor and are avidly recruited by vascularizing tumors. J Cell Biochem. 2004;91:1146–1158.
    1. Karnoub AE, Dash AB, Vo AP, Sullivan A, Brooks MW, et al. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature. 2007;449:557–563.
    1. Nakamura K, Ito Y, Kawano Y, Kurozumi K, Kobune M, et al. Antitumor effect of genetically engineered mesenchymal stem cells in a rat glioma model. Gene Ther. 2004;11:1155–1164.
    1. Jiang H, Ren J. Inoculation of murine bone marrow mesenchymal stem cells induces tumor necrosis in mouse with orthotopic hepatocellular carcinoma. Beijing Da Xue Xue Bao. 2008;40:453–458.
    1. Khakoo AY, Pati S, Anderson SA, Reid W, Elshal MF, et al. Human mesenchymal stem cells exert potent antitumorigenic effects in a model of Kaposi's sarcoma. J Exp Med. 2006;203:1235–1247.
    1. Ohlsson LB, Varas L, Kjellman C, Edvardsen K, Lindvall M. Mesenchymal progenitor cell-mediated inhibition of tumor growth in vivo and in vitro in gelatin matrix. Exp Mol Pathol. 2003;75:248–255.
    1. Sasser AK, Mundy BL, Smith KM, Studebaker AW, Axel AE, et al. Human bone marrow stromal cells enhance breast cancer cell growth rates in a cell line-dependent manner when evaluated in 3D tumor environments. Cancer Lett. 2007;254:255–264.
    1. Sasser AK, Sullivan NJ, Studebaker AW, Hendey LF, Axel AE, et al. Interleukin-6 is a potent growth factor for ER-α-positive human breast cancer. FASEB J. 2007;21:3763–3770.
    1. Bae S, Park CW, Son HK, Ju HK, Paik D, et al. Fibroblast activation protein α identifies mesenchymal stromal cells from human bone marrow. Br J Haematol. 2008;142:827–830.
    1. Littlepage LE, Egeblad M, Werb Z. Coevolution of cancer and stromal cellular responses. Cancer Cell. 2005;7:499–500.
    1. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401.
    1. Lazarides E, Granger BL, Gard DL. Desmin- and vimentin-containing filaments and their role in the assembly of the Z disk in muscle cells. COLD SPRING HARBOR SYMP QUANT BIOL. 1982;46:351–378.
    1. Oswald J, Boxberger S, Jørgensen B, Feldmann S, Ehninger G, et al. Mesenchymal stem cells can be differentiated into endothelial cells in vitro. Stem Cells. 2004;22:377–384.
    1. Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LMG, et al. Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am J Pathol. 1999;155:739–752.
    1. Ilan N, Madri JA. PECAM-1: Old friend, new partners. Curr Opin Cell Biol. 2003;15:515–524.
    1. Harrison P, Bradley L, Bomford A. Mechanism of regulation of HGF/SF gene expression in fibroblasts by TGF-β1. Biochem Biophys Res Commun. 2000;271:203–211.
    1. Untergasser G, Gander R, Lilg C, Lepperdinger G, Plas E, et al. Profiling molecular targets of TGF-β1 in prostate fibroblast-to- myofibroblast transdifferentiation. Mech Ageing Dev. 2005;126:59–69.
    1. Corso S, Migliore C, Ghiso E, De Rosa G, Comoglio PM, et al. Silencing the MET oncogene leads to regression of experimental tumors and metastases. Oncogene. 2008;27:684–693.
    1. Labelle M, Schnittler HJ, Aust DE, Friedrich K, Etton G, et al. Vascular endothelial cadherin promotes breast cancer progression via transforming growth factor β signaling. Cancer Res. 2008;68:1388–1397.
    1. Yang F, Strand DW, Rowley DR. Fibroblast growth factor-2 mediates transforming growth factor-Β action in prostate cancer reactive stroma. Oncogene. 2008;27:450–459.
    1. Matsumoto Y, Motoki T, Kubota S, Takigawa M, Tsubouchi H, et al. Inhibition of tumor-stromal interaction through HGF/Met signaling by valproic acid. Biochem Biophys Res Commun. 2008;366:110–116.
    1. Abounader R, Laterra J. Scatter factor/hepatocyte growth factor in brain tumor growth and angiogenesis. Neuro-Oncology. 2005;7:436–451.
    1. Spix JK, Chay EY, Block ER, Klarlund JK. Hepatocyte growth factor induces epithelial cell motility through transactivation of the epidermal growth factor receptor. Exp Cell Res. 2007;313:3319–3325.
    1. Peister A, Mellad JA, Larson BL, Hall BM, Gibson LF, et al. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood. 2004;103:1662–1668.
    1. Klopp AH, Spaeth EL, Dembinski JL, Woodward WA, Munshi A, et al. Tumor irradiation increases the recruitment of circulating mesenchymal stem cells into the tumor microenvironment. Cancer Res. 2007;67:11687–11695.
    1. Miyoshi H, Blomer U, Takahashi M, Gage FH, Verma IM. Development of a self-inactivating lentivirus vector. J Virol. 1998;72:8150–8157.

Source: PubMed

3
Prenumerera