Skip to main content
Top
Published in: Annals of Surgical Oncology 2/2019

01-02-2019 | Pancreatic Tumors

Increased SOX9 Expression in Premalignant and Malignant Pancreatic Neoplasms

Authors: Jennifer L. Gnerlich, MD, Xianzhong Ding, MD, Cara Joyce, PhD, Kevin Turner, BA, Christopher D. Johnson, MD, Haiyan Chen, MD, Gerard J. Abood, MD, Samuel G. Pappas, MD, Gerard V. Aranha, MD

Published in: Annals of Surgical Oncology | Issue 2/2019

Login to get access

Abstract

Background

SOX9, a progenitor cell marker, is important for pancreatic ductal development. Our goal was to examine SOX9 expression differences in intraductal papillary mucinous neoplasms (IPMNs) and ductal adenocarcinoma (PDAC) compared with benign pancreatic duct (BP).

Methods

SOX9 expression was evaluated by immunohistochemistry performed on 93 specimens: 37 BP, 24 low grade (LG) IPMN, 12 high grade (HG) IPMN, and 20 PDAC. A linear mixed-effects model was used to compare the percentage of cells expressing SOX9 by specimen type. A separate linear mixed-effects model evaluated differences in SOX9 expression by staining intensity in pancreatic epithelial cells.

Results

Nuclear SOX9 expression was detected in the epithelial cells of 98% HG IPMN, 93% LG IPMN, 83% PDAC, and 60% BP. Compared with BP, SOX9 was expressed from a significantly greater percentage of cells in LG IMPN, HG IMPN, and PDAC (p < 0.001 for each). BP and PDAC showed greater variability in SOX9 expression in epithelial cells compared with IPMNs which showed strong, homogenous SOX9 expression in almost all cells. Compared with BP, both LG and HG IPMN showed significantly greater SOX9 expression (p < 0.001 for each), but there was no significant difference in SOX9 expression between LG and HG IPMN (p > 0.05). PDAC had significantly higher expression of SOX9 compared with BP but significantly lower SOX9 expression compared with LG or HG IPMN (p < 0.001 for each).

Conclusions

IPMNs demonstrated the highest expression levels of SOX9. SOX9 expression in BP and PDAC demonstrated much more heterogeneity compared with the strong, uniform expression in IPMN.
Literature
1.
go back to reference Hotz HG, Hotz B, Buhr HJ. Genes associated with epithelial-mesenchymal transition: possible therapeutic targets in ductal pancreatic adenocarcinoma? Anticancer Agents Med Chem. 2011;11(5):448–54.CrossRefPubMed Hotz HG, Hotz B, Buhr HJ. Genes associated with epithelial-mesenchymal transition: possible therapeutic targets in ductal pancreatic adenocarcinoma? Anticancer Agents Med Chem. 2011;11(5):448–54.CrossRefPubMed
2.
3.
go back to reference Lahat G, Lubezky N, Loewenstein S, et al. Epithelial-to-mesenchymal transition (EMT) in intraductal papillary mucinous neoplasm (IPMN) is associated with high tumor grade and adverse outcomes. Ann Surg Oncol. 2014; 21 Suppl 4:S750–7.CrossRefPubMed Lahat G, Lubezky N, Loewenstein S, et al. Epithelial-to-mesenchymal transition (EMT) in intraductal papillary mucinous neoplasm (IPMN) is associated with high tumor grade and adverse outcomes. Ann Surg Oncol. 2014; 21 Suppl 4:S750–7.CrossRefPubMed
4.
go back to reference Russell R, Perkhofer L, Liebau S, et al. Loss of ATM accelerates pancreatic cancer formation and epithelial-mesenchymal transition. Nat Commun. 2015;6:7677.CrossRefPubMedPubMedCentral Russell R, Perkhofer L, Liebau S, et al. Loss of ATM accelerates pancreatic cancer formation and epithelial-mesenchymal transition. Nat Commun. 2015;6:7677.CrossRefPubMedPubMedCentral
5.
go back to reference Castellanos JA, Merchant NB, Nagathihalli NS. Emerging targets in pancreatic cancer: epithelial-mesenchymal transition and cancer stem cells. OncoTargets Ther. 2013;6:1261–7.PubMedPubMedCentral Castellanos JA, Merchant NB, Nagathihalli NS. Emerging targets in pancreatic cancer: epithelial-mesenchymal transition and cancer stem cells. OncoTargets Ther. 2013;6:1261–7.PubMedPubMedCentral
6.
go back to reference Su A, He S, Tian B, et al. MicroRNA-221 mediates the effects of PDGF-BB on migration, proliferation, and the epithelial-mesenchymal transition in pancreatic cancer cells. PLoS ONE. 2013;8(8):e71309.CrossRefPubMedPubMedCentral Su A, He S, Tian B, et al. MicroRNA-221 mediates the effects of PDGF-BB on migration, proliferation, and the epithelial-mesenchymal transition in pancreatic cancer cells. PLoS ONE. 2013;8(8):e71309.CrossRefPubMedPubMedCentral
7.
go back to reference Lee JM, Dedhar S, Kalluri R, Thompson EW. The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol. 2006;172(7):973–81.CrossRefPubMedPubMedCentral Lee JM, Dedhar S, Kalluri R, Thompson EW. The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol. 2006;172(7):973–81.CrossRefPubMedPubMedCentral
8.
go back to reference Savagner P. Leaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays. 2001;23(10):912–23.CrossRefPubMed Savagner P. Leaving the neighborhood: molecular mechanisms involved during epithelial-mesenchymal transition. Bioessays. 2001;23(10):912–23.CrossRefPubMed
9.
go back to reference Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.CrossRefPubMed Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.CrossRefPubMed
11.
go back to reference Weber CE, Li NY, Wai PY, Kuo PC. Epithelial-mesenchymal transition, TGF-beta, and osteopontin in wound healing and tissue remodeling after injury. J Burn Care Res. 2012;33(3):311–8.CrossRefPubMedPubMedCentral Weber CE, Li NY, Wai PY, Kuo PC. Epithelial-mesenchymal transition, TGF-beta, and osteopontin in wound healing and tissue remodeling after injury. J Burn Care Res. 2012;33(3):311–8.CrossRefPubMedPubMedCentral
12.
go back to reference Nakamura M, Tokura Y. Epithelial-mesenchymal transition in the skin. J Dermatol Sci. 2011;61(1):7–13.CrossRefPubMed Nakamura M, Tokura Y. Epithelial-mesenchymal transition in the skin. J Dermatol Sci. 2011;61(1):7–13.CrossRefPubMed
13.
go back to reference Hudson LG, Newkirk KM, Chandler HL, et al. Cutaneous wound reepithelialization is compromised in mice lacking functional Slug (Snai2). J Dermatol Sci. 2009;56(1):19–26.CrossRefPubMedPubMedCentral Hudson LG, Newkirk KM, Chandler HL, et al. Cutaneous wound reepithelialization is compromised in mice lacking functional Slug (Snai2). J Dermatol Sci. 2009;56(1):19–26.CrossRefPubMedPubMedCentral
14.
go back to reference Savagner P, Kusewitt DF, Carver EA, et al. Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. J Cell Physiol. 2005;202(3):858–66.CrossRefPubMed Savagner P, Kusewitt DF, Carver EA, et al. Developmental transcription factor slug is required for effective re-epithelialization by adult keratinocytes. J Cell Physiol. 2005;202(3):858–66.CrossRefPubMed
15.
go back to reference Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2(6):442–54.CrossRefPubMed Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002;2(6):442–54.CrossRefPubMed
16.
go back to reference Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14(6):818–29.CrossRefPubMed Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14(6):818–29.CrossRefPubMed
17.
go back to reference Yamada S, Fuchs BC, Fujii T, et al. Epithelial-to-mesenchymal transition predicts prognosis of pancreatic cancer. Surgery. 2013;154(5):946–54.CrossRefPubMed Yamada S, Fuchs BC, Fujii T, et al. Epithelial-to-mesenchymal transition predicts prognosis of pancreatic cancer. Surgery. 2013;154(5):946–54.CrossRefPubMed
18.
go back to reference Schonleben F, Qiu W, Bruckman KC, et al. BRAF and KRAS gene mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/IPMC) of the pancreas. Cancer Lett. 2007;249(2):242–8.CrossRefPubMed Schonleben F, Qiu W, Bruckman KC, et al. BRAF and KRAS gene mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/IPMC) of the pancreas. Cancer Lett. 2007;249(2):242–8.CrossRefPubMed
19.
go back to reference Wada K. p16 and p53 gene alterations and accumulations in the malignant evolution of intraductal papillary-mucinous tumors of the pancreas. J Hepatobiliary Pancreat Surg. 2002;9(1):76–85.CrossRefPubMed Wada K. p16 and p53 gene alterations and accumulations in the malignant evolution of intraductal papillary-mucinous tumors of the pancreas. J Hepatobiliary Pancreat Surg. 2002;9(1):76–85.CrossRefPubMed
20.
go back to reference Cheung M, Briscoe J. Neural crest development is regulated by the transcription factor Sox9. Development. 2003;130(23):5681–93.CrossRefPubMed Cheung M, Briscoe J. Neural crest development is regulated by the transcription factor Sox9. Development. 2003;130(23):5681–93.CrossRefPubMed
21.
go back to reference Foster JW, Dominguez-Steglich MA, Guioli S, et al. Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature. 1994;372(6506):525–30.CrossRefPubMed Foster JW, Dominguez-Steglich MA, Guioli S, et al. Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature. 1994;372(6506):525–30.CrossRefPubMed
22.
go back to reference Gordon CT, Tan TY, Benko S, et al. Long-range regulation at the SOX9 locus in development and disease. J Med Genet. 2009;46(10):649–56.CrossRefPubMed Gordon CT, Tan TY, Benko S, et al. Long-range regulation at the SOX9 locus in development and disease. J Med Genet. 2009;46(10):649–56.CrossRefPubMed
23.
go back to reference Lynn FC, Smith SB, Wilson ME, et al. Sox9 coordinates a transcriptional network in pancreatic progenitor cells. Proc Natl Acad Sci U S A. 2007;104(25):10500–5.CrossRefPubMedPubMedCentral Lynn FC, Smith SB, Wilson ME, et al. Sox9 coordinates a transcriptional network in pancreatic progenitor cells. Proc Natl Acad Sci U S A. 2007;104(25):10500–5.CrossRefPubMedPubMedCentral
24.
go back to reference Seymour PA, Freude KK, Tran MN, et al. SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proc Natl Acad Sci U S A. 2007;104(6):1865–70.CrossRefPubMedPubMedCentral Seymour PA, Freude KK, Tran MN, et al. SOX9 is required for maintenance of the pancreatic progenitor cell pool. Proc Natl Acad Sci U S A. 2007;104(6):1865–70.CrossRefPubMedPubMedCentral
25.
go back to reference Chakravarty G, Moroz K, Makridakis NM, et al. Prognostic significance of cytoplasmic SOX9 in invasive ductal carcinoma and metastatic breast cancer. Exp Biol Med (Maywood). 2011;236(2):145–55.CrossRefPubMed Chakravarty G, Moroz K, Makridakis NM, et al. Prognostic significance of cytoplasmic SOX9 in invasive ductal carcinoma and metastatic breast cancer. Exp Biol Med (Maywood). 2011;236(2):145–55.CrossRefPubMed
26.
go back to reference Darido C, Buchert M, Pannequin J, et al. Defective claudin-7 regulation by Tcf-4 and Sox-9 disrupts the polarity and increases the tumorigenicity of colorectal cancer cells. Cancer Res. 2008;68(11):4258–68.CrossRefPubMed Darido C, Buchert M, Pannequin J, et al. Defective claudin-7 regulation by Tcf-4 and Sox-9 disrupts the polarity and increases the tumorigenicity of colorectal cancer cells. Cancer Res. 2008;68(11):4258–68.CrossRefPubMed
27.
go back to reference Zalzali H, Naudin C, Bastide P, et al. CEACAM1, a SOX9 direct transcriptional target identified in the colon epithelium. Oncogene. 2008;27(56):7131–8.CrossRefPubMed Zalzali H, Naudin C, Bastide P, et al. CEACAM1, a SOX9 direct transcriptional target identified in the colon epithelium. Oncogene. 2008;27(56):7131–8.CrossRefPubMed
28.
go back to reference Sashikawa Kimura M, Mutoh H, Sugano K. SOX9 is expressed in normal stomach, intestinal metaplasia, and gastric carcinoma in humans. J Gastroenterol. 2011;46(11):1292–9.CrossRefPubMed Sashikawa Kimura M, Mutoh H, Sugano K. SOX9 is expressed in normal stomach, intestinal metaplasia, and gastric carcinoma in humans. J Gastroenterol. 2011;46(11):1292–9.CrossRefPubMed
29.
go back to reference Jiang SS, Fang WT, Hou YH, et al. Upregulation of SOX9 in lung adenocarcinoma and its involvement in the regulation of cell growth and tumorigenicity. Clin Cancer Res. 2010;16(17):4363–73.CrossRefPubMed Jiang SS, Fang WT, Hou YH, et al. Upregulation of SOX9 in lung adenocarcinoma and its involvement in the regulation of cell growth and tumorigenicity. Clin Cancer Res. 2010;16(17):4363–73.CrossRefPubMed
30.
go back to reference Wang H, Leav I, Ibaragi S, et al. SOX9 is expressed in human fetal prostate epithelium and enhances prostate cancer invasion. Cancer Res. 2008;68(6):1625–30.CrossRefPubMed Wang H, Leav I, Ibaragi S, et al. SOX9 is expressed in human fetal prostate epithelium and enhances prostate cancer invasion. Cancer Res. 2008;68(6):1625–30.CrossRefPubMed
31.
go back to reference Kopp JL, von Figura G, Mayes E, et al. Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;22(6):737–50.CrossRefPubMedPubMedCentral Kopp JL, von Figura G, Mayes E, et al. Identification of Sox9-dependent acinar-to-ductal reprogramming as the principal mechanism for initiation of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;22(6):737–50.CrossRefPubMedPubMedCentral
32.
go back to reference Shroff S, Rashid A, Wang H, et al. SOX9: a useful marker for pancreatic ductal lineage of pancreatic neoplasms. Hum Pathol. 2014;45(3):456–63.CrossRefPubMed Shroff S, Rashid A, Wang H, et al. SOX9: a useful marker for pancreatic ductal lineage of pancreatic neoplasms. Hum Pathol. 2014;45(3):456–63.CrossRefPubMed
33.
go back to reference Meng F, Takaori K, Ito T, et al. Expression of SOX9 in intraductal papillary mucinous neoplasms of the pancreas. Pancreas. 2014;43(1):7–14.CrossRefPubMed Meng F, Takaori K, Ito T, et al. Expression of SOX9 in intraductal papillary mucinous neoplasms of the pancreas. Pancreas. 2014;43(1):7–14.CrossRefPubMed
34.
go back to reference Pan JJ, Yang MH. The role of epithelial-mesenchymal transition in pancreatic cancer. J Gastrointest Oncol. 2011;2(3):151–6.PubMedPubMedCentral Pan JJ, Yang MH. The role of epithelial-mesenchymal transition in pancreatic cancer. J Gastrointest Oncol. 2011;2(3):151–6.PubMedPubMedCentral
35.
go back to reference Furuyama K, Kawaguchi Y, Akiyama H, et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet. 2011;43(1):34–41.CrossRefPubMed Furuyama K, Kawaguchi Y, Akiyama H, et al. Continuous cell supply from a Sox9-expressing progenitor zone in adult liver, exocrine pancreas and intestine. Nat Genet. 2011;43(1):34–41.CrossRefPubMed
36.
go back to reference Tanaka T, Kuroki T, Adachi T, et al. Evaluation of SOX9 expression in pancreatic ductal adenocarcinoma and intraductal papillary mucinous neoplasm. Pancreas. 2013;42(3):488–93.CrossRefPubMed Tanaka T, Kuroki T, Adachi T, et al. Evaluation of SOX9 expression in pancreatic ductal adenocarcinoma and intraductal papillary mucinous neoplasm. Pancreas. 2013;42(3):488–93.CrossRefPubMed
Metadata
Title
Increased SOX9 Expression in Premalignant and Malignant Pancreatic Neoplasms
Authors
Jennifer L. Gnerlich, MD
Xianzhong Ding, MD
Cara Joyce, PhD
Kevin Turner, BA
Christopher D. Johnson, MD
Haiyan Chen, MD
Gerard J. Abood, MD
Samuel G. Pappas, MD
Gerard V. Aranha, MD
Publication date
01-02-2019
Publisher
Springer International Publishing
Published in
Annals of Surgical Oncology / Issue 2/2019
Print ISSN: 1068-9265
Electronic ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-018-6925-4

Other articles of this Issue 2/2019

Annals of Surgical Oncology 2/2019 Go to the issue