Skip to main content

Advertisement

Log in

CXCL1 Derived from Mammary Fibroblasts Promotes Progression of Mammary Lesions to Invasive Carcinoma through CXCR2 Dependent Mechanisms

  • Published:
Journal of Mammary Gland Biology and Neoplasia Aims and scope Submit manuscript

Abstract

With improved screening methods, the numbers of abnormal breast lesions diagnosed in women have been increasing over time. However, it remains unclear whether these breast lesions will develop into invasive cancers. To more effectively predict the outcome of breast lesions and determine a more appropriate course of treatment, it is important to understand the underlying mechanisms that regulate progression of non-invasive lesions to invasive breast cancers. A hallmark of invasive breast cancers is the accumulation of fibroblasts. Fibroblast proliferation and activation in the mammary gland is in part regulated by the Transforming Growth Factor beta1 pathway (TGF-β). In animal models, TGF-β suppression of CCL2 and CXCL1 chemokine expression is associated with metastatic progression of mammary carcinomas. Here, we show that transgenic overexpression of the Polyoma middle T viral antigen in the mouse mammary gland of C57BL/6 mice results in slow growing non-invasive lesions that progress to invasive carcinomas in a stage dependent manner. Invasive carcinomas are associated with accumulation of fibroblasts that show decreased TGF-β expression and high levels of CXCL1, but not CCL2. Using co-transplant models, we show that decreased TGF-β signaling in fibroblasts contribute to mammary carcinoma progression through enhancement of CXCL1/CXCR2 dependent mechanisms. Using cell culture models, we show that CXCL1 mediated mammary carcinoma cell invasion through NF-κB, AKT, Stat3 and p42/44MAPK dependent mechanisms. These studies provide novel mechanistic insight into the progression of pre-invasive lesions and identify new stromal biomarkers, with important prognostic implications.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9

Similar content being viewed by others

References

  1. Hussain A, Gordon-Dixon A, Almusawy H, Sinha P, Desai A. The incidence and outcome of incidental breast lesions detected by computed tomography. Ann R Coll Surg Engl. 2010;92(2):124–6. https://doi.org/10.1308/003588410X12518836439083.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Falomo E, Strigel RM, Bruce R, Munoz Del Rio A, Adejumo C, Kelcz F. Incidence and outcomes of incidental breast lesions detected on cross-sectional imaging examinations. Breast J. 2018; https://doi.org/10.1111/tbj.13040.

  3. Menes TS, Rosenberg R, Balch S, Jaffer S, Kerlikowske K, Miglioretti DL. Upgrade of high-risk breast lesions detected on mammography in the breast Cancer surveillance consortium. Am J Surg. 2014;207(1):24–31. https://doi.org/10.1016/j.amjsurg.2013.05.014.

    Article  PubMed  Google Scholar 

  4. Schnitt SJ. Benign breast disease and breast cancer risk: morphology and beyond. Am J Surg Pathol. 2003;27(6):836–41.

    Article  PubMed  Google Scholar 

  5. Dyrstad SW, Yan Y, Fowler AM, Colditz GA. Breast cancer risk associated with benign breast disease: systematic review and meta-analysis. Breast Cancer Res Treat. 2015;149(3):569–75. https://doi.org/10.1007/s10549-014-3254-6.

    Article  CAS  PubMed  Google Scholar 

  6. Hartmann LC, Degnim AC, Santen RJ, Dupont WD, Ghosh K. Atypical hyperplasia of the breast--risk assessment and management options. N Engl J Med. 2015;372(1):78–89. https://doi.org/10.1056/NEJMsr1407164.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Tozbikian G, Brogi E, Vallejo CE, Giri D, Murray M, Catalano J, et al. Atypical ductal hyperplasia bordering on ductal carcinoma in situ. Int J Surg Pathol. 2017;25(2):100–7. https://doi.org/10.1177/1066896916662154.

    Article  PubMed  Google Scholar 

  8. Kim J, Han W, Go EY, Moon HG, Ahn SK, Shin HC, et al. Validation of a scoring system for predicting malignancy in patients diagnosed with atypical ductal hyperplasia using an ultrasound-guided core needle biopsy. J Breast Cancer. 2012;15(4):407–11. https://doi.org/10.4048/jbc.2012.15.4.407.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Wagoner MJ, Laronga C, Acs G. Extent and histologic pattern of atypical ductal hyperplasia present on core needle biopsy specimens of the breast can predict ductal carcinoma in situ in subsequent excision. Am J Clin Pathol. 2009;131(1):112–21. https://doi.org/10.1309/AJCPGHEJ2R8UYFGP.

    Article  PubMed  Google Scholar 

  10. Nizri E, Schneebaum S, Klausner JM, Menes TS. Current management practice of breast borderline lesions--need for further research and guidelines. Am J Surg. 2012;203(6):721–5. https://doi.org/10.1016/j.amjsurg.2011.06.052.

    Article  PubMed  Google Scholar 

  11. Georgian-Smith D, Lawton TJ. Variations in physician recommendations for surgery after diagnosis of a high-risk lesion on breast core needle biopsy. AJR Am J Roentgenol. 2012;198(2):256–63. https://doi.org/10.2214/AJR.11.7717.

    Article  PubMed  Google Scholar 

  12. Ahn S, Cho J, Sung J, Lee JE, Nam SJ, Kim KM, et al. The prognostic significance of tumor-associated stroma in invasive breast carcinoma. Tumour Biol. 2012;33(5):1573–80. https://doi.org/10.1007/s13277-012-0411-6.

    Article  PubMed  Google Scholar 

  13. Cid S, Eiro N, Fernandez B, Sanchez R, Andicoechea A, Fernandez-Muniz PI, et al. Prognostic influence of tumor stroma on breast Cancer subtypes. Clin Breast Cancer. 2018;18(1):e123–e33. https://doi.org/10.1016/j.clbc.2017.08.008.

    Article  CAS  PubMed  Google Scholar 

  14. Bussard KM, Mutkus L, Stumpf K, Gomez-Manzano C, Marini FC. Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res. 2016;18(1):84. https://doi.org/10.1186/s13058-016-0740-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Komohara Y, Takeya M. CAFs and TAMs: maestros of the tumour microenvironment. J Pathol. 2017;241(3):313–5. https://doi.org/10.1002/path.4824.

    Article  CAS  PubMed  Google Scholar 

  16. Zhang Y, Alexander PB, Wang XF. TGF-beta Family Signaling in the Control of Cell Proliferation and Survival. Cold Spring Harb Perspect Biol. 2017;9(4) https://doi.org/10.1101/cshperspect.a022145.

  17. Kahata K, Dadras MS, Moustakas A. TGF-beta family signaling in epithelial differentiation and epithelial-mesenchymal transition. Cold Spring Harb Perspect Biol. 2017;10 https://doi.org/10.1101/cshperspect.a022194.

  18. Bhowmick N, Chytiil A, Plieth D, Gorska A, Dumont N, Shappel S, et al. TGF-ß signaling in fibroblasts modulates the oncogenic potential of adjacent epithelia. Science. 2004;303:847–51.

    Article  Google Scholar 

  19. Cheng N, Bhowmick NA, Chytil A, Gorksa AE, Brown KA, Muraoka R, et al. Loss of TGF-beta type II receptor in fibroblasts promotes mammary carcinoma growth and invasion through upregulation of TGF-alpha-, MSP- and HGF-mediated signaling networks. Oncogene. 2005;24(32):5053–68. https://doi.org/10.1038/sj.onc.1208685.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hembruff SL, Jokar I, Yang L, Cheng N. Loss of transforming growth factor-beta signaling in mammary fibroblasts enhances CCL2 secretion to promote mammary tumor progression through macrophage-dependent and -independent mechanisms. Neoplasia. 2010;12(5):425–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fang WB, Mafuvadze B, Yao M, Zou A, Portsche M, Cheng N. TGF-beta negatively regulates CXCL1 chemokine expression in mammary fibroblasts through enhancement of Smad2/3 and suppression of HGF/c-met signaling mechanisms. PLoS One. 2015;10(8):e0135063. https://doi.org/10.1371/journal.pone.0135063.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Rees PA, Greaves NS, Baguneid M, Bayat A. Chemokines in wound healing and as potential therapeutic targets for reducing cutaneous scarring. Adv Wound Care. 2015;4(11):687–703. https://doi.org/10.1089/wound.2014.0568.

    Article  Google Scholar 

  23. Ridiandries A, Tan JT, Bursill CA. The Role of CC-Chemokines in the Regulation of Angiogenesis. Int J Mol Sci. 2016;17(11) https://doi.org/10.3390/ijms17111856.

  24. Strieter RM, Belperio JA, Burdick MD, Keane MP. CXC chemokines in angiogenesis relevant to chronic fibroproliferation. Curr Drug Targets Inflamm Allergy. 2005;4(1):23–6.

    Article  CAS  PubMed  Google Scholar 

  25. Stillie R, Farooq SM, Gordon JR, Stadnyk AW. The functional significance behind expressing two IL-8 receptor types on PMN. J Leukoc Biol. 2009;86(3):529–43. https://doi.org/10.1189/jlb.0208125.

    Article  CAS  PubMed  Google Scholar 

  26. Balkwill FR. The chemokine system and cancer. J Pathol. 2012;226(2):148–57. https://doi.org/10.1002/path.3029.

    Article  CAS  PubMed  Google Scholar 

  27. White GE, Iqbal AJ, Greaves DR. CC chemokine receptors and chronic inflammation--therapeutic opportunities and pharmacological challenges. Pharmacol Rev. 2013;65(1):47–89. https://doi.org/10.1124/pr.111.005074.

    Article  CAS  PubMed  Google Scholar 

  28. Greaves NS, Ashcroft KJ, Baguneid M, Bayat A. Current understanding of molecular and cellular mechanisms in fibroplasia and angiogenesis during acute wound healing. J Dermatol Sci. 2013;72(3):206–17. https://doi.org/10.1016/j.jdermsci.2013.07.008.

    Article  CAS  PubMed  Google Scholar 

  29. Lowman HB, Slagle PH, DeForge LE, Wirth CM, Gillece-Castro BL, Bourell JH, et al. Exchanging interleukin-8 and melanoma growth-stimulating activity receptor binding specificities. J Biol Chem. 1996;271(24):14344–52.

    Article  CAS  PubMed  Google Scholar 

  30. Kurihara T, Bravo R. Cloning and functional expression of mCCR2, a murine receptor for the C-C chemokines JE and FIC. J Biol Chem. 1996;271(20):11603–7.

    Article  CAS  PubMed  Google Scholar 

  31. Singh S, Varney M, Singh RK. Host CXCR2-dependent regulation of melanoma growth, angiogenesis, and experimental lung metastasis. Cancer Res. 2009;69(2):411–5. https://doi.org/10.1158/0008-5472.CAN-08-3378.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sharma B, Nannuru KC, Varney ML, Singh RK. Host Cxcr2-dependent regulation of mammary tumor growth and metastasis. Clin Exp Metastasis. 2015;32(1):65–72. https://doi.org/10.1007/s10585-014-9691-0.

    Article  CAS  PubMed  Google Scholar 

  33. Acharyya S, Oskarsson T, Vanharanta S, Malladi S, Kim J, Morris PG, et al. A CXCL1 paracrine network links cancer chemoresistance and metastasis. Cell. 2012;150(1):165–78. https://doi.org/10.1016/j.cell.2012.04.042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dong YL, Kabir SM, Lee ES, Son DS. CXCR2-driven ovarian cancer progression involves upregulation of proinflammatory chemokines by potentiating NF-kappaB activation via EGFR-transactivated Akt signaling. PLoS One. 2013;8(12):e83789. https://doi.org/10.1371/journal.pone.0083789.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Saintigny P, Massarelli E, Lin S, Ahn YH, Chen Y, Goswami S, et al. CXCR2 expression in tumor cells is a poor prognostic factor and promotes invasion and metastasis in lung adenocarcinoma. Cancer Res. 2013;73(2):571–82. https://doi.org/10.1158/0008-5472.CAN-12-0263.

    Article  CAS  PubMed  Google Scholar 

  36. Wang B, Khachigian LM, Esau L, Birrer MJ, Zhao X, Parker MI, et al. A key role for early growth response-1 and nuclear factor-kappaB in mediating and maintaining GRO/CXCR2 proliferative signaling in esophageal cancer. Mol Cancer Res. 2009;7(5):755–64. https://doi.org/10.1158/1541-7786.MCR-08-0472.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wang B, Hendricks DT, Wamunyokoli F, Parker MI. A growth-related oncogene/CXC chemokine receptor 2 autocrine loop contributes to cellular proliferation in esophageal cancer. Cancer Res. 2006;66(6):3071–7. https://doi.org/10.1158/0008-5472.CAN-05-2871.

    Article  CAS  PubMed  Google Scholar 

  38. Dhawan P, Richmond A. A novel NF-kappa B-inducing kinase-MAPK signaling pathway up-regulates NF-kappa B activity in melanoma cells. J Biol Chem. 2002;277(10):7920–8.

    Article  CAS  PubMed  Google Scholar 

  39. Lim SY, Yuzhalin AE, Gordon-Weeks AN, Muschel RJ. Targeting the CCL2-CCR2 signaling axis in cancer metastasis. Oncotarget. 2016;7(19):28697–710. https://doi.org/10.18632/oncotarget.7376.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Borsig L, Wolf MJ, Roblek M, Lorentzen A, Heikenwalder M. Inflammatory chemokines and metastasis--tracing the accessory. Oncogene. 2014;33(25):3217–24. https://doi.org/10.1038/onc.2013.272.

    Article  CAS  PubMed  Google Scholar 

  41. Granot Z, Henke E, Comen EA, King TA, Norton L, Benezra R. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell. 2011;20(3):300–14. https://doi.org/10.1016/j.ccr.2011.08.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lu Y, Chen Q, Corey E, Xie W, Fan J, Mizokami A, et al. Activation of MCP-1/CCR2 axis promotes prostate cancer growth in bone. Clin Exp Metastasis. 2009;26(2):161–9. https://doi.org/10.1007/s10585-008-9226-7.

    Article  CAS  PubMed  Google Scholar 

  43. Fang WB, Jokar I, Zou A, Lambert D, Dendukuri P, Cheng N. CCL2/CCR2 chemokine signaling coordinates survival and motility of breast cancer cells through Smad3 protein- and p42/44 mitogen-activated protein kinase (MAPK)-dependent mechanisms. J Biol Chem. 2012;287(43):36593–608. https://doi.org/10.1074/jbc.M112.365999.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zou A, Lambert D, Yeh H, Yasukawa K, Behbod F, Fan F, et al. Elevated CXCL1 expression in breast cancer stroma predicts poor prognosis and is inversely associated with expression of TGF-beta signaling proteins. BMC Cancer. 2014;14:781. https://doi.org/10.1186/1471-2407-14-781.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yao M, Yu E, Staggs V, Fan F, Cheng N. Elevated expression of chemokine C-C ligand 2 in stroma is associated with recurrent basal-like breast cancers. Mod Pathol. 2016;29:810–23. https://doi.org/10.1038/modpathol.2016.78.

    Article  CAS  PubMed  Google Scholar 

  46. Bierie B, Chung CH, Parker JS, Stover DG, Cheng N, Chytil A, et al. Abrogation of TGF-beta signaling enhances chemokine production and correlates with prognosis in human breast cancer. J Clin Invest. 2009;119(6):1571–82. https://doi.org/10.1172/JCI37480.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bugge TH, Lund LR, Kombrinck KK, Nielsen BS, Holmback K, Drew AF, et al. Reduced metastasis of Polyoma virus middle T antigen-induced mammary cancer in plasminogen-deficient mice. Oncogene. 1998;16(24):3097–104. https://doi.org/10.1038/sj.onc.1201869.

    Article  CAS  PubMed  Google Scholar 

  48. Langley RR, Ramirez KM, Tsan RZ, Van Arsdall M, Nilsson MB, Fidler IJ. Tissue-specific microvascular endothelial cell lines from H-2K(b)-tsA58 mice for studies of angiogenesis and metastasis. Cancer Res. 2003;63(11):2971–6.

    CAS  PubMed  Google Scholar 

  49. Lambert D, Cheng N. Mammary transplantation of stromal cells and carcinoma cells in C57BL/6 mice. J Vis Exp. 2011;54

  50. Salomonis N, Schlieve CR, Pereira L, Wahlquist C, Colas A, Zambon AC, et al. Alternative splicing regulates mouse embryonic stem cell pluripotency and differentiation. Proc Natl Acad Sci U S A. 2010;107(23):10514–9. https://doi.org/10.1073/pnas.0912260107.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Borowicz S, Van Scoyk M, Avasarala S, Karuppusamy Rathinam MK, Tauler J, Bikkavilli RK, et al. The soft agar colony formation assay. J Vis Exp: JoVE. 2014;92:e51998. https://doi.org/10.3791/51998.

    Article  CAS  Google Scholar 

  52. Paddison PJ, Cleary M, Silva JM, Chang K, Sheth N, Sachidanandam R, et al. Cloning of short hairpin RNAs for gene knockdown in mammalian cells. Nat Methods. 2004;1(2):163–7. https://doi.org/10.1038/nmeth1104-163.

    Article  CAS  PubMed  Google Scholar 

  53. Fang WB, Yao M, Brummer G, Acevedo D, Alhakamy N, Berkland C et al. Targeted gene silencing of CCL2 inhibits triple negative breast cancer progression by blocking cancer stem cell renewal and M2 macrophage recruitment. Oncotarget. 2016. https://doi.org/10.18632/oncotarget.9885.

  54. Cheng N, Chytil A, Shyr Y, Joly A, Moses HL. Transforming growth factor-beta signaling-deficient fibroblasts enhance hepatocyte growth factor signaling in mammary carcinoma cells to promote scattering and invasion. Mol Cancer Res. 2008;6(10):1521–33. https://doi.org/10.1158/1541-7786.MCR-07-2203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lin E, Jones J, Zhu L, Whitney K, Muller W, Pollard J. Progression to malignancy in the polyoma middle T oncoprotein mouse breast cancer model provides a reliable model for human diseases. Am J of Path. 2003;163(5):2113–26.

    Article  Google Scholar 

  56. Maglione JE, Moghanaki D, Young LJ, Manner CK, Ellies LG, Joseph SO, et al. Transgenic Polyoma middle-T mice model premalignant mammary disease. Cancer Res. 2001;61(22):8298–305.

    CAS  PubMed  Google Scholar 

  57. Guy C, Cardiff R, Muller W. Induction of mammary tumors by expression a polyomavirus middle T oncogene: a transgenic mouse model for metastatic disease. Mol Cell Biol. 1992;12:954–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Davie SA, Maglione JE, Manner CK, Young D, Cardiff RD, MacLeod CL, et al. Effects of FVB/NJ and C57Bl/6J strain backgrounds on mammary tumor phenotype in inducible nitric oxide synthase deficient mice. Transgenic Res. 2007;16(2):193–201. https://doi.org/10.1007/s11248-006-9056-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Qiu TH, Chandramouli GV, Hunter KW, Alkharouf NW, Green JE, Liu ET. Global expression profiling identifies signatures of tumor virulence in MMTV-PyMT-transgenic mice: correlation to human disease. Cancer Res. 2004;64(17):5973–81. https://doi.org/10.1158/0008-5472.CAN-04-0242.

    Article  CAS  PubMed  Google Scholar 

  60. Tse GM, Tan PH, Lui PC, Gilks CB, Poon CS, Ma TK, et al. The role of immunohistochemistry for smooth-muscle actin, p63, CD10 and cytokeratin 14 in the differential diagnosis of papillary lesions of the breast. J Clin Pathol. 2007;60(3):315–20. https://doi.org/10.1136/jcp.2006.036830.

    Article  CAS  PubMed  Google Scholar 

  61. Reisenbichler ES, Balmer NN, Adams AL, Pfeifer JD, Hameed O. Luminal cytokeratin expression profiles of breast papillomas and papillary carcinomas and the utility of a cytokeratin 5/p63/cytokeratin 8/18 antibody cocktail in their distinction. Mod Pathol. 2011;24(2):185–93. https://doi.org/10.1038/modpathol.2010.197.

    Article  CAS  PubMed  Google Scholar 

  62. Fouad TM, Kogawa T, Liu DD, Shen Y, Masuda H, El-Zein R, et al. Overall survival differences between patients with inflammatory and noninflammatory breast cancer presenting with distant metastasis at diagnosis. Breast Cancer Res Treat. 2015;152(2):407–16. https://doi.org/10.1007/s10549-015-3436-x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Weidner N. Current pathologic methods for measuring intratumoral microvessel density within breast carcinoma and other solid tumors. Breast Cancer Res Treat. 1995;36(2):169–80.

    Article  CAS  PubMed  Google Scholar 

  64. Sacks GP, Clover LM, Bainbridge DR, Redman CW, Sargent IL. Flow cytometric measurement of intracellular Th1 and Th2 cytokine production by human villous and extravillous cytotrophoblast. Placenta. 2001;22(6):550–9. https://doi.org/10.1053/plac.2001.0686.

    Article  CAS  PubMed  Google Scholar 

  65. Mori M, Sadahira Y, Kawasaki S, Hayashi T, Awai M. Macrophage heterogeneity in bone marrow culture in vitro. J Cell Sci. 1990;95(Pt 3):481–5.

    PubMed  Google Scholar 

  66. Cooper KD, Duraiswamy N, Hammerberg C, Allen E, Kimbrough-Green C, Dillon W, et al. Neutrophils, differentiated macrophages, and monocyte/macrophage antigen presenting cells infiltrate murine epidermis after UV injury. J Invest Dermatol. 1993;101(2):155–63.

    Article  CAS  PubMed  Google Scholar 

  67. Sharon Y, Alon L, Glanz S, Servais C, Erez N. Isolation of normal and cancer-associated fibroblasts from fresh tissues by fluorescence activated cell sorting (FACS). J Vis Exp: JoVE. 2013;71:e4425. https://doi.org/10.3791/4425.

    Article  CAS  Google Scholar 

  68. Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification of fibroblast heterogeneity in the tumor microenvironment. Cancer Biol Ther. 2006;5(12):1640–6.

    Article  CAS  PubMed  Google Scholar 

  69. Matsuyoshi N, Imamura S. Multiple cadherins are expressed in human fibroblasts. Biochem Biophys Res Commun. 1997;235(2):355–8. https://doi.org/10.1006/bbrc.1997.6707.

    Article  CAS  PubMed  Google Scholar 

  70. Bhowmick NA, Ghiassi M, Bakin A, Aakre M, Lundquist CA, Engel ME, et al. Transforming growth factor-beta1 mediates epithelial to mesenchymal transdifferentiation through a RhoA-dependent mechanism. Mol Biol Cell. 2001;12(1):27–36. https://doi.org/10.1091/mbc.12.1.27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Li Q, Zhang D, Wang Y, Sun P, Hou X, Larner J, et al. MiR-21/Smad 7 signaling determines TGF-beta1-induced CAF formation. Sci Rep. 2013;3:2038. https://doi.org/10.1038/srep02038.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Lenicek T, Szerda F, Demirovic A, Mijic A, Kruslin B, Tomas D. Pleomorphic ductal carcinoma of the breast with predominant micropapillary features. Pathol Int. 2007;57(10):694–7. https://doi.org/10.1111/j.1440-1827.2007.02159.x.

    Article  PubMed  Google Scholar 

  73. Aslakson CJ, Miller FR. Selective events in the metastatic process defined by analysis of the sequential dissemination of subpopulations of a mouse mammary tumor. Cancer Res. 1992;52(6):1399–405.

    CAS  PubMed  Google Scholar 

  74. Owens RB. Glandular epithelial cells from mice: a method for selective cultivation. J Natl Cancer Inst. 1974;52(4):1375–8.

    Article  CAS  PubMed  Google Scholar 

  75. Bandyopadhyay A, Cibull ML, Sun LZ. Isolation and characterization of a spontaneously transformed malignant mouse mammary epithelial cell line in culture. Carcinogenesis. 1998;19(11):1907–11.

    Article  CAS  PubMed  Google Scholar 

  76. Lazard D, Sastre X, Frid MG, Glukhova MA, Thiery JP, Koteliansky VE. Expression of smooth muscle-specific proteins in myoepithelium and stromal myofibroblasts of normal and malignant human breast tissue. Proc Natl Acad Sci U S A. 1993;90(3):999–1003.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Duncia JV, Santella JB 3rd, Higley CA, Pitts WJ, Wityak J, Frietze WE, et al. MEK inhibitors: the chemistry and biological activity of U0126, its analogs, and cyclization products. Bioorg Med Chem Lett. 1998;8(20):2839–44.

    Article  CAS  PubMed  Google Scholar 

  78. Pierce JW, Schoenleber R, Jesmok G, Best J, Moore SA, Collins T, et al. Novel inhibitors of cytokine-induced IkappaBalpha phosphorylation and endothelial cell adhesion molecule expression show anti-inflammatory effects in vivo. J Biol Chem. 1997;272(34):21096–103.

    Article  CAS  PubMed  Google Scholar 

  79. Jeon M, Han J, Nam SJ, Lee JE, Kim S. Elevated IL-1beta expression induces invasiveness of triple negative breast cancer cells and is suppressed by zerumbone. Chem Biol Interact. 2016;258:126–33. https://doi.org/10.1016/j.cbi.2016.08.021.

    Article  CAS  PubMed  Google Scholar 

  80. Sangai T, Akcakanat A, Chen H, Tarco E, Wu Y, Do KA, et al. Biomarkers of response to Akt inhibitor MK-2206 in breast cancer. Clin Cancer Res. 2012;18(20):5816–28. https://doi.org/10.1158/1078-0432.CCR-12-1141.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Turkson J, Ryan D, Kim JS, Zhang Y, Chen Z, Haura E, et al. Phosphotyrosyl peptides block Stat3-mediated DNA binding activity, gene regulation, and cell transformation. J Biol Chem. 2001;276(48):45443–55. https://doi.org/10.1074/jbc.M107527200.

    Article  CAS  PubMed  Google Scholar 

  82. Zhao W, Jaganathan S, Turkson J. A cell-permeable Stat3 SH2 domain mimetic inhibits Stat3 activation and induces antitumor cell effects in vitro. J Biol Chem. 2010;285(46):35855–65. https://doi.org/10.1074/jbc.M110.154088.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Bortner DM, Rosenberg MP. Induction of mammary gland hyperplasia and carcinomas in transgenic mice expressing human cyclin E. Mol Cell Biol. 1997;17(1):453–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Andrechek ER, Cardiff RD, Chang JT, Gatza ML, Acharya CR, Potti A, et al. Genetic heterogeneity of Myc-induced mammary tumors reflecting diverse phenotypes including metastatic potential. Proc Natl Acad Sci U S A. 2009;106(38):16387–92. https://doi.org/10.1073/pnas.0901250106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Rosner A, Miyoshi K, Landesman-Bollag E, Xu X, Seldin DC, Moser AR, et al. Pathway pathology: histological differences between ErbB/Ras and Wnt pathway transgenic mammary tumors. Am J Pathol. 2002;161(3):1087–97. https://doi.org/10.1016/S0002-9440(10)64269-1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Nassar H, Qureshi H, Adsay NV, Visscher D. Clinicopathologic analysis of solid papillary carcinoma of the breast and associated invasive carcinomas. Am J Surg Pathol. 2006;30(4):501–7.

    Article  PubMed  Google Scholar 

  87. Collins LC, Schnitt SJ. Papillary lesions of the breast: selected diagnostic and management issues. Histopathology. 2008;52(1):20–9. https://doi.org/10.1111/j.1365-2559.2007.02898.x.

    Article  CAS  PubMed  Google Scholar 

  88. Tan PH, Schnitt SJ, van de Vijver MJ, Ellis IO, Lakhani SR. Papillary and neuroendocrine breast lesions: the WHO stance. Histopathology. 2015;66(6):761–70. https://doi.org/10.1111/his.12463.

    Article  PubMed  Google Scholar 

  89. Yamashita M, Ogawa T, Zhang X, Hanamura N, Kashikura Y, Takamura M, et al. Role of stromal myofibroblasts in invasive breast cancer: stromal expression of alpha-smooth muscle actin correlates with worse clinical outcome. Breast Cancer. 2012;19(2):170–6. https://doi.org/10.1007/s12282-010-0234-5.

    Article  PubMed  Google Scholar 

  90. Cardone A, Tolino A, Zarcone R, Borruto Caracciolo G, Tartaglia E. Prognostic value of desmoplastic reaction and lymphocytic infiltration in the management of breast cancer. Panminerva Med. 1997;39(3):174–7.

    CAS  PubMed  Google Scholar 

  91. Webster M, Hutchinson JN, Rauh MJ, Muthuswamy SK, Anton A, Tortorice CG, et al. Requirement of both Shc and Phosphatidyl 3′ kinase signaling pathways in polyomavirus middle T-mediated mammar tumorigenesis. Mol Cell Biol. 1998;18(4):2344–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Schwab LP, Peacock DL, Majumdar D, Ingels JF, Jensen LC, Smith KD, et al. Hypoxia-inducible factor 1alpha promotes primary tumor growth and tumor-initiating cell activity in breast cancer. Breast Cancer Res. 2012;14(1):R6. https://doi.org/10.1186/bcr3087.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, et al. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48. https://doi.org/10.1016/j.cell.2005.02.034.

    Article  CAS  PubMed  Google Scholar 

  94. Gache C, Berthois Y, Martin PM, Saez S. Positive regulation of normal and tumoral mammary epithelial cell proliferation by fibroblasts in coculture. In Vitro Cell Dev Biol Anim. 1998;34(4):347–51. https://doi.org/10.1007/s11626-998-0012-2.

    Article  CAS  PubMed  Google Scholar 

  95. Studebaker AW, Storci G, Werbeck JL, Sansone P, Sasser AK, Tavolari S, et al. Fibroblasts isolated from common sites of breast cancer metastasis enhance cancer cell growth rates and invasiveness in an interleukin-6-dependent manner. Cancer Res. 2008;68(21):9087–95. https://doi.org/10.1158/0008-5472.CAN-08-0400.

    Article  CAS  PubMed  Google Scholar 

  96. Horgan K, Jones DL, Mansel RE. Mitogenicity of human fibroblasts in vivo for human breast cancer cells. Br J Surg. 1987;74(3):227–9.

    Article  CAS  PubMed  Google Scholar 

  97. Ray P, Stacer AC, Fenner J, Cavnar SP, Meguiar K, Brown M, et al. CXCL12-gamma in primary tumors drives breast cancer metastasis. Oncogene. 2015;34(16):2043–51. https://doi.org/10.1038/onc.2014.157.

    Article  CAS  PubMed  Google Scholar 

  98. Gabrilovich DI. Myeloid-derived suppressor cells. Cancer Immunol Res. 2017;5(1):3–8. https://doi.org/10.1158/2326-6066.CIR-16-0297.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Jorch SK, Kubes P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med. 2017;23(3):279–87. https://doi.org/10.1038/nm.4294.

    Article  CAS  PubMed  Google Scholar 

  100. Tabaries S, Ouellet V, Hsu BE, Annis MG, Rose AA, Meunier L, et al. Granulocytic immune infiltrates are essential for the efficient formation of breast cancer liver metastases. Breast Cancer Res. 2015;17:45. https://doi.org/10.1186/s13058-015-0558-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Matsui A, Yokoo H, Negishi Y, Endo-Takahashi Y, Chun NA, Kadouchi I, et al. CXCL17 expression by tumor cells recruits CD11b+Gr1 high F4/80- cells and promotes tumor progression. PLoS One. 2012;7(8):e44080. https://doi.org/10.1371/journal.pone.0044080.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Morales JK, Kmieciak M, Knutson KL, Bear HD, Manjili MH. GM-CSF is one of the main breast tumor-derived soluble factors involved in the differentiation of CD11b-Gr1- bone marrow progenitor cells into myeloid-derived suppressor cells. Breast Cancer Res Treat. 2010;123(1):39–49. https://doi.org/10.1007/s10549-009-0622-8.

    Article  CAS  PubMed  Google Scholar 

  103. Lin D, Lei L, Zhang Y, Hu B, Bao G, Liu Y, et al. Secreted IL-1alpha promotes T-cell activation and expansion of CD11b(+) Gr1(+) cells in carbon tetrachloride-induced liver injury in mice. Eur J Immunol. 2015;45(7):2084–98. https://doi.org/10.1002/eji.201445195.

    Article  CAS  PubMed  Google Scholar 

  104. Sharma M, Beck AH, Webster JA, Espinosa I, Montgomery K, Varma S, et al. Analysis of stromal signatures in the tumor microenvironment of ductal carcinoma in situ. Breast Cancer Res Treat. 2010;123(2):397–404. https://doi.org/10.1007/s10549-009-0654-0.

    Article  CAS  PubMed  Google Scholar 

  105. Sobolik T, Su YJ, Wells S, Ayers GD, Cook RS, Richmond A. CXCR4 drives the metastatic phenotype in breast cancer through induction of CXCR2 and activation of MEK and PI3K pathways. Mol Biol Cell. 2014;25(5):566–82. https://doi.org/10.1091/mbc.E13-07-0360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Singh JK, Farnie G, Bundred NJ, Simoes BM, Shergill A, Landberg G, et al. Targeting CXCR1/2 significantly reduces breast cancer stem cell activity and increases the efficacy of inhibiting HER2 via HER2-dependent and -independent mechanisms. Clin Cancer Res. 2013;19(3):643–56. https://doi.org/10.1158/1078-0432.CCR-12-1063.

    Article  CAS  PubMed  Google Scholar 

  107. Dhawan P, Richmond A. Role of CXCL1 in tumorigenesis of melanoma. J Leukoc Biol. 2002;72(1):9–18.

    CAS  PubMed  Google Scholar 

  108. Wang Z, Wang Z, Li G, Wu H, Sun K, Chen J, et al. CXCL1 from tumor-associated lymphatic endothelial cells drives gastric cancer cell into lymphatic system via activating integrin beta1/FAK/AKT signaling. Cancer Lett. 2017;385:28–38. https://doi.org/10.1016/j.canlet.2016.10.043.

    Article  CAS  PubMed  Google Scholar 

  109. Kuo PL, Shen KH, Hung SH, Hsu YL. CXCL1/GROalpha increases cell migration and invasion of prostate cancer by decreasing fibulin-1 expression through NF-kappaB/HDAC1 epigenetic regulation. Carcinogenesis. 2012;33(12):2477–87. https://doi.org/10.1093/carcin/bgs299.

    Article  CAS  PubMed  Google Scholar 

  110. Kemp DM, Pidich A, Larijani M, Jonas R, Lash E, Sato T, et al. Ladarixin, a dual CXCR1/2 inhibitor, attenuates experimental melanomas harboring different molecular defects by affecting malignant cells and tumor microenvironment. Oncotarget. 2017;8(9):14428–42. https://doi.org/10.18632/oncotarget.14803.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Chang Q, Bournazou E, Sansone P, Berishaj M, Gao SP, Daly L, et al. The IL-6/JAK/Stat3 feed-forward loop drives tumorigenesis and metastasis. Neoplasia. 2013;15(7):848–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Song L, Turkson J, Karras JG, Jove R, Haura EB. Activation of Stat3 by receptor tyrosine kinases and cytokines regulates survival in human non-small cell carcinoma cells. Oncogene. 2003;22(27):4150–65. https://doi.org/10.1038/sj.onc.1206479.

    Article  CAS  PubMed  Google Scholar 

  113. Cheng N, Chytil A, Shyr Y, Joly A, Moses HL. Enhanced hepatocyte growth factor signaling by type II transforming growth factor-beta receptor knockout fibroblasts promotes mammary tumorigenesis. Cancer Res. 2007;67(10):4869–77. https://doi.org/10.1158/0008-5472.CAN-06-3381.

    Article  CAS  PubMed  Google Scholar 

  114. Elliott BE, Hung WL, Boag AH, Tuck AB. The role of hepatocyte growth factor (scatter factor) in epithelial-mesenchymal transition and breast cancer. Can J Physiol Pharmacol. 2002;80(2):91–102.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Iman Jokar for technical assistance.

Funding

These studies were supported by funds from KU Endowment and the National Institutes of Health/National Cancer Institute: R01CA172764 to N. Cheng.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nikki Cheng.

Electronic supplementary material

ESM 1

(PPTX 2.09 mb)

ESM 2

(PPTX 689 kb)

ESM 3

(PPTX 76.3 kb)

ESM 4

(PPTX 2.15 mb)

ESM 5

(PPTX 543 kb)

ESM 6

(PPTX 703 kb)

ESM 7

(PPTX 79.2 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bernard, S., Myers, M., Fang, W.B. et al. CXCL1 Derived from Mammary Fibroblasts Promotes Progression of Mammary Lesions to Invasive Carcinoma through CXCR2 Dependent Mechanisms. J Mammary Gland Biol Neoplasia 23, 249–267 (2018). https://doi.org/10.1007/s10911-018-9407-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10911-018-9407-1

Keywords

Navigation