Cancer-associated fibroblasts predict poor outcome and promote periostin-dependent invasion in oesophageal adenocarcinoma

Timothy J Underwood, Annette L Hayden, Mathieu Derouet, Edwin Garcia, Fergus Noble, Michael J White, Steve Thirdborough, Abbie Mead, Nicholas Clemons, Massimiliano Mellone, Chudy Uzoho, John N Primrose, Jeremy P Blaydes, Gareth J Thomas, Timothy J Underwood, Annette L Hayden, Mathieu Derouet, Edwin Garcia, Fergus Noble, Michael J White, Steve Thirdborough, Abbie Mead, Nicholas Clemons, Massimiliano Mellone, Chudy Uzoho, John N Primrose, Jeremy P Blaydes, Gareth J Thomas

Abstract

Interactions between cancer cells and cancer-associated fibroblasts (CAFs) play an important role in tumour development and progression. In this study we investigated the functional role of CAFs in oesophageal adenocarcinoma (EAC). We used immunochemistry to analyse a cohort of 183 EAC patients for CAF markers related to disease mortality. We characterized CAFs and normal oesophageal fibroblasts (NOFs) using western blotting, immunofluorescence and gel contraction. Transwell assays, 3D organotypic culture and xenograft models were used to examine the effects on EAC cell function and to dissect molecular mechanisms regulating invasion. Most EACs (93%) contained CAFs with a myofibroblastic (α-SMA-positive) phenotype, which correlated significantly with poor survival [p = 0.016; HR 7. 1 (1.7-29.4)]. Primary CAFs isolated from EACs have a contractile, myofibroblastic phenotype and promote EAC cell invasion in vitro (Transwell assays, p ≤ 0.05; organotypic culture, p < 0.001) and in vivo (p ≤ 0.05). In vitro, this pro-invasive effect is modulated through the matricellular protein periostin. Periostin is secreted by CAFs and acts as a ligand for EAC cell integrins αvβ3 and αvβ5, promoting activation of the PI3kinase-Akt pathway. In patient samples, periostin expression at the tumour cell-stromal interface correlates with poor overall and disease-free survival. Our study highlights the importance of the tumour stroma in EAC progression. Paracrine interaction between CAF-secreted periostin and EAC-expressed integrins results in PI3 kinase-Akt activation and increased tumour cell invasion. Most EACs contain a myofibroblastic CAF-rich stroma; this may explain the aggressive, highly infiltrative nature of the disease, and suggests that stromal targeting may produce therapeutic benefit in EAC patients.

Keywords: CAFs; oesophageal cancer; periostin; tumour microenvironment.

© 2014 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.

Figures

Figure 1
Figure 1
CAFs in the microenvironment are important in EAC. (A) Kaplan–Meier curve of overall survival after resection in patients with α-SMA-positive versus α-SMA-negative tumours; representative IHC images of negative/low and positive/high are in the right-hand panel. (B) Western blot for pan-cytokeratin and vimentin in FLO-1, OE33 and primary oesophageal NOFs and CAFs. (C) Immunocytochemistry for α-SMA in primary oesophageal NOFs and CAFs. (D) Photograph of a representative gel contraction assay at 72 h and gel weight with the incorporation of CAFs compared to NOFs (p < 0.01; n = 3). (E) Western blot and immunocytochemistry for α-SMA in NOFs and NOFs + TGFβ1, and a representative gel contraction assay
Figure 2
Figure 2
CAFs promote EAC cell invasion in vitro and tumour growth in vivo. (A) Transwell invasion assays comparing the effect of NOFs and CAFs conditioned medium on invasion of the EAC cell lines FLO-1 and OE33. (B) Organotypic models of EAC comparing the effect of NOFs and CAFs to promote invasion of FLO-1 and OE33. (C) Kaplan–Meier curves of overall survival of mice injected with OE33 alone or in combination with NOFs or CAFs. Mice were culled when the tumour volume reached 500 mm3. (D) Immunohistochemistry for Pan-CK and α-SMA in tumour xenografts containing OE33 and CAFs
Figure 3
Figure 3
Periostin is secreted by CAFs and promotes EAC cell invasion. (A) Unsupervised hierarchical clustering of the ECM module genes differentiated between normal oesophageal epithelium and EAC. (B) Exported network weights in BioLayout Express3D, graphically representing periostin as a central gene within the ECM module, with many connections to other ECM module genes and genes associated with the cell-cycle process. (C) Western blot of conditioned medium and immunocytochemistry, comparing the expression and secretion of periostin in NOFs and CAFs. (D) Transwell invasion assays, demonstrating the invasion of FLO-1 in response to CAF-conditioned medium ± periostin silencing, as indicated (*p < 0.05; **p < 0.001), with western blot of conditioned medium normalized to cell number confirming periostin knockdown. (E) Organotypic cultures of FLO-1 and CAFs, with and without periostin silencing. (F) Transwell invasion assays of FLO-1 towards CAF-conditioned medium, with or without periostin immunodepletion and rescue with recombinant periostin, as indicated (*p < 0.05). (G) Transwell invasion assay of FLO-1 towards conditioned medium from NOFs or NOFs + TGFβ1, with and without periostin silencing, and western blot analysis of periostin expression under the same conditions (**p < 0.001). (H) Transwell invasion assay of FLO-1 towards conditioned medium from NOFs ± recombinant periostin or CAFs, as indicated (**p < 0.001)
Figure 4
Figure 4
Periostin-dependent invasion of EAC cells is suppressed through inhibition of αvβ3 and αvβ5 integrins and PI3K. (A) Western blot for total and phospho-Akt in FLO-1 up to 120 min after stimulation with recombinant periostin. (B) Western blot for total and phospho-Akt in FLO-1, 30 min after periostin stimulation, in the presence or absence of blocking antibodies against integrins αvβ3 or αvβ5, as indicated. (C) Western blot for total and phospho-Akt in FLO-1, 30 min after periostin stimulation, in the presence or absence of the PI3K inhibitor LY294002. (D) Transwell invasion assays of FLO-1 towards NOFs or CAFs conditioned medium under varying conditions. Invasion towards NOF-conditioned medium is used as the internal control. Periostin was added to the NOF-conditioned medium and either LY294002 or blocking antibodies against integrins αvβ3 or αvβ5 were added to the FLO-1 cells, as indicated. (E) Organotypic models of FLO-1 + CAFs in the presence or absence of LY294002, including staining by H&E, pan-cytokeratin and Ki67. (F) Immunohistochemistry for p-Akt in representative human EAC tumours with strongest staining adjacent to CAFs. (G) Immunohistochemistry for periostin and p-Akt in tumour xenografts containing OE33 and CAFs
Figure 5
Figure 5
Stromal periostin expression correlates with poor survival in EAC. (A) Kaplan–Meier curves of overall survival after resection in patients with periostin-positive versus periostin-negative tumours. (B) Kaplan–Meier curves of disease-free survival after resection in patients with periostin-positive versus periostin-negative tumours. (C) Immunohistochemistry for periostin expression in human normal oesophagus (limited to the walls of blood vessels) and EAC. (D) Immunohistochemistry to compare expression patterns of periostin and α-SMA in EAC

References

    1. Clinical Effectiveness Unit R, AUGIS, BSG, NCASP. National Oesophago-Gastric Cancer Audit. National Report. Report No. IC15100510. NHS Information Centre: Leeds, 2010
    1. Pennathur A, Gibson MK, Jobe BA. Oesophageal carcinoma. Lancet. 2013;381:400–412.
    1. Wang DH, Souza RF. Biology of Barrett's esophagus and esophageal adenocarcinoma. Gastrointest Endosc Clin N Am. 2011;21:25–38.
    1. Fitzgerald RC. Molecular basis of Barrett's oesophagus and oesophageal adenocarcinoma. Gut. 2006;55:1810–1820.
    1. Spechler SJ, Fitzgerald RC, Prasad GA, et al. History, molecular mechanisms, and endoscopic treatment of Barrett's esophagus. Gastroenterology. 2010;138:854–869.
    1. Hu M, Polyak K. Microenvironmental regulation of cancer development. Curr Opin Genet Dev. 2008;18:27–34.
    1. De Monte L, Reni M, Tassi E, et al. Intratumor T helper type 2 cell infiltrate correlates with cancer-associated fibroblast thymic stromal lymphopoietin production and reduced survival in pancreatic cancer. J Exp Med. 2011;208:469–478.
    1. Marsh D, Suchak K, Moutasim KA, et al. Stromal features are predictive of disease mortality in oral cancer patients. J Pathol. 2011;223:470–481.
    1. Surowiak P, Murawa D, Materna V, et al. Occurence of stromal myofibroblasts in the invasive ductal breast cancer tissue is an unfavourable prognostic factor. Anticancer Res. 2007;27:2917–2924.
    1. Tsujino T, Seshimo I, Yamamoto H, et al. Stromal myofibroblasts predict disease recurrence for colorectal cancer. Clin Cancer Res. 2007;13:2082–2090.
    1. Wu Y, Grabsch H, Ivanova T, et al. Comprehensive genomic meta-analysis identifies intra-tumoural stroma as a predictor of survival in patients with gastric cancer. Gut. 2013;62:1100–1111.
    1. Saadi A, Shannon NB, Lao-Sirieix P, et al. Stromal genes discriminate preinvasive from invasive disease, predict outcome, and highlight inflammatory pathways in digestive cancers. Proc Natl Acad Sci USA. 2010;107:2177–2182.
    1. Shimoda M, Mellody KT, Orimo A. Carcinoma-associated fibroblasts are a rate-limiting determinant for tumour progression. Semin Cell Dev Biol. 2010;21:19–25.
    1. Massarelli G, Tanda F, Bosincu L, et al. Myofibroblasts in the epithelial-stromal junction of basal cell carcinoma. Appl Pathol. 1983;1:25–30.
    1. Ohtani H, Sasano N. Stromal cell changes in human colorectal adenomas and carcinomas. An ultrastructural study of fibroblasts, myofibroblasts, and smooth muscle cells. Virchows Archiv A Pathol Anat Histopathol. 1983;401:209–222.
    1. Kawashiri S, Tanaka A, Noguchi N, et al. Significance of stromal desmoplasia and myofibroblast appearance at the invasive front in squamous cell carcinoma of the oral cavity. Head Neck. 2009;31:1346–1353.
    1. Merika EE, Syrigos KN, Saif MW. Desmoplasia in pancreatic cancer. Can we fight it? Gastroenterol Res Pract. 2012;2012 781765.
    1. Bailey JM, Swanson BJ, Hamada T, et al. Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin Cancer Res. 2008;14:5995–6004.
    1. Yamashita M, Ogawa T, Zhang X, et al. Role of stromal myofibroblasts in invasive breast cancer: stromal expression of α-smooth muscle actin correlates with worse clinical outcome. Breast Cancer. 2012;19:170–176.
    1. Lai D, Ma L, Wang F. Fibroblast activation protein regulates tumor-associated fibroblasts and epithelial ovarian cancer cells. Int J Oncol. 2012;41:541–550.
    1. Lewis MP, Lygoe KA, Nystrom ML, et al. Tumour-derived TGF-β1 modulates myofibroblast differentiation and promotes HGF/SF-dependent invasion of squamous carcinoma cells. Br J Cancer. 2004;90:822–832.
    1. Vong S, Kalluri R. The role of stromal myofibroblast and extracellular matrix in tumor angiogenesis. Genes Cancer. 2011;2:1139–1145.
    1. Gaggioli C, Hooper S, Hidalgo-Carcedo C, et al. Fibroblast-led collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 2007;9:1392–1400.
    1. De Wever O, Demetter P, Mareel M, et al. Stromal myofibroblasts are drivers of invasive cancer growth. Int J Cancer. 2008;123:2229–2238.
    1. Thannickal VJ, Lee DY, White ES, et al. Myofibroblast differentiation by transforming growth factor-β1 is dependent on cell adhesion and integrin signaling via focal adhesion kinase. J Biol Chem. 2003;278:12384–12389.
    1. Hinz B, Phan SH, Thannickal VJ, et al. Recent developments in myofibroblast biology: paradigms for connective tissue remodeling. Am J Pathol. 2012;180:1340–1355.
    1. Courrech Staal EF, Smit VT, van Velthuysen ML, et al. Reproducibility and validation of tumour stroma ratio scoring on oesophageal adenocarcinoma biopsies. Eur J Cancer. 2011;47:375–382.
    1. Schoppmann SF, Jesch B, Riegler MF, et al. Podoplanin expressing cancer associated fibroblasts are associated with unfavourable prognosis in adenocarcinoma of the esophagus. Clin Exp Metast. 2013;30:441–446.
    1. Underwood TJ, Derouet M, White MJ, et al. A comparison of primary oesophageal squamous epithelial cells with HET-1A in organotypic culture. Biol Cell. 2010;102:635–644.
    1. Clemons NJ, Do H, Fennell C, et al. Characterization of a novel tumorigenic esophageal adenocarcinoma cell line: OANC1. Dig Dis Sci. 2014;59:78–88.
    1. Bergman LM, Birts CN, Darley M, et al. CtBPs promote cell survival through the maintenance of mitotic fidelity. Mol Cell Biol. 2009;29:4539–4551.
    1. Moutasim KA, Nystrom ML, Thomas GJ. Cell migration and invasion assays. Methods Mol Biol. 2011;731:333–343.
    1. Jenei V, Nystrom ML, Thomas GJ. Measuring invasion in an organotypic model. Methods Mol Biol. 2011;769:223–232.
    1. Oldham MC, Konopka G, Iwamoto K, et al. Functional organization of the transcriptome in human brain. Nat Neurosci. 2008;11:1271–1282.
    1. Chen Y, Zhu J, Lum PY, et al. Variations in DNA elucidate molecular networks that cause disease. Nature. 2008;452:429–435.
    1. Rice TW, Blackstone EH, Rusch VW. 7th edition of the AJCC Cancer Staging Manual, esophagus and esophagogastric junction. Ann Surg Oncol. 2010;17:1721–1724.
    1. UICC. TNM Classification of Malignant Tumours. 7th edn. New York: Wiley-Blackwell; 2009.
    1. Langfelder P, Horvath S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformat. 2008;9:559.
    1. Kimchi ET, Posner MC, Park JO, et al. Progression of Barrett's metaplasia to adenocarcinoma is associated with the suppression of the transcriptional programs of epidermal differentiation. Cancer Res. 2005;65:3146–3154.
    1. Wang Q, Ma C, Kemmner W. Wdr66 is a novel marker for risk stratification and involved in epithelial–mesenchymal transition of esophageal squamous cell carcinoma. BMC Cancer. 2013;13:137.
    1. Koppert LB, Wijnhoven BP, van Dekken H, et al. The molecular biology of esophageal adenocarcinoma. J Surg Oncol. 2005;92:169–190.
    1. Weaver JM, Ross-Innes CS, Shannon N, et al. Ordering of mutations in preinvasive disease stages of esophageal carcinogenesis. Nat Genet. 2014;46:837–843.
    1. Dulak AM, Stojanov P, Peng S, et al. Exome and whole-genome sequencing of esophageal adenocarcinoma identifies recurrent driver events and mutational complexity. Nat Genet. 2013;45:478–486.
    1. Agrawal N, Jiao Y, Bettegowda C, et al. Comparative genomic analysis of esophageal adenocarcinoma and squamous cell carcinoma. Cancer Discov. 2012;2:899–905.
    1. Baril P, Gangeswaran R, Mahon PC, et al. Periostin promotes invasiveness and resistance of pancreatic cancer cells to hypoxia-induced cell death: role of the β4 integrin and the PI3k pathway. Oncogene. 2007;26:2082–2094.
    1. Li G, Jin R, Norris RA, et al. Periostin mediates vascular smooth muscle cell migration through the integrins αvβ3 and αvβ5 and focal adhesion kinase (FAK) pathway. Atherosclerosis. 2010;208:358–365.
    1. Utispan K, Sonongbua J, Thuwajit P, et al. Periostin activates integrin α5β1 through a PI3K/AKTdependent pathway in invasion of cholangiocarcinoma. Int J Oncol. 2012;41:1110–1118.
    1. Ruan K, Bao S, Ouyang G. The multifaceted role of periostin in tumorigenesis. Cell Mol Life Sci. 2009;66:2219–2230.
    1. Malanchi I, Santamaria-Martinez A, Susanto E, et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature. 2012;481:85–89.
    1. Lee YJ, Kim IS, Park SA, et al. Periostin-binding DNA aptamer inhibits breast cancer growth and metastasis. Mol Ther. 2013;21:1004–1013.
    1. Michaylira CZ, Wong GS, Miller CG, et al. Periostin, a cell adhesion molecule, facilitates invasion in the tumor microenvironment and annotates a novel tumor-invasive signature in esophageal cancer. Cancer Res. 2010;70:5281–5292.
    1. Conway SJ, Izuhara K, Kudo Y, et al. The role of periostin in tissue remodeling across health and disease. Cell Mol Life Sci. 2014;71:1279–1288.
    1. Sheppard K, Kinross KM, Solomon B, et al. Targeting PI3 kinase/AKT/mTOR signaling in cancer. Crit Rev Oncog. 2012;17:69–95.
    1. Bruhn MA, Pearson RB, Hannan RD, et al. AKT-independent PI3-K signaling in cancer – emerging role for SGK3. Cancer Manag Res. 2013;5:281–292.
    1. Bioinformatics. 2004;20:307–315. affy – analysis of Affymetrix GeneChip data at the probe level Gautier L, Cope L, Bolstad BM, et al.
    1. Oldham MC, Langfelder P, Horvath S. Network methods for describing sample relationships in genomic datasets: application to Huntington's disease. BMC Syst Biol. 2012;6:63.
    1. Theocharidis A, van Dongen S, Enright AJ, et al. Network visualization and analysis of gene expression data using BioLayout Express (3D) Nat Protoc. 2009;4:1535–1550.

Source: PubMed

3
Abonnieren