Upregulation of the oestrogen target gene SIX1 is associated with higher growth speed and decreased survival in HCV-positive women with hepatocellular carcinoma

Rosina Maria Critelli, Fabiola Milosa, Adriana Romanzi, Simone Lasagni, Gemma Marcelli, Lorenza Di Marco, Alessandra Pivetti, Filippo Schepis, Dante Romagnoli, Serena Mancarella, Francesco Dituri, Maria-Luz Martinez-Chantar, Gianluigi Giannelli, Erica Villa, Rosina Maria Critelli, Fabiola Milosa, Adriana Romanzi, Simone Lasagni, Gemma Marcelli, Lorenza Di Marco, Alessandra Pivetti, Filippo Schepis, Dante Romagnoli, Serena Mancarella, Francesco Dituri, Maria-Luz Martinez-Chantar, Gianluigi Giannelli, Erica Villa

Abstract

The male/female ratio of patients with hepatocellular carcinoma (HCC) is often unbalanced towards the male sex, indicating a sex predisposition for HCC development. A possible explanation may be attributed to different hormonal statuses, including the pro-inflammatory action of androgens in men and the protective effects of oestrogen against excessive inflammation in women. Although several studies have studied gene expression in patients with HCC, very few have attempted to identify features that could be distinctive between male and female patients. The present study aimed to identify distinctive signalling mechanisms between men and women that may be associated with HCC progression. The present study analysed a detailed microarray database that was obtained from the prospective study of 78 patients with HCC to study gene expression according to sex. In addition, the present study aimed to evaluate whether the differentially expressed genes were known oestrogen targets. Moreover, RNAs from the HCC cohort were evaluated for microRNA (miRNA/miR) expression, and a relationship between miRNA and gene expression according to sex was investigated. One gene, sineoculis homeobox homolog 1 (SIX1), which is known to be an oestrogen target gene, was revealed to be highly upregulated in hepatitis virus C (HCV)-positive female patients with HCC but not in HCV-positive male patients. In addition, SIX1 upregulation had a significant relationship with tumour growth speed (assessed as tumour doubling time in two CTs performed 6 weeks apart) and survival (P=0.009 and P=0.042, respectively) in female patients only. Furthermore, SIX1 upregulation was related with miR-421 and miR-9-5p only in male patients; however, in female patients, SIX1 upregulation had a direct relationship with miR-181b, miR-503-5p and miR-125b (miRNAs with potential oncogenic capacity), and an inverse correlation with miR139-5p, miR-26b, let7c-3p and let7c-5p (putatively oncosuppressive microRNAs). These data suggested a distinctive model for liver carcinogenesis in HCV-positive women, with downregulation of protective mechanisms against tumour progression and the activation of potential oncogenes, in relation to the oestrogen target gene SIX1. (IRB10/08_CE_UniRer; ClinicalTrials ID: NCT01657695).

Keywords: TGF-β; biologic aggressiveness; hepatocellular carcinoma; microRNA; sex; sineoculis homeobox homolog 1.

Conflict of interest statement

The authors declare that they have no competing interests.

Copyright: © Critelli et al.

Figures

Figure 1.
Figure 1.
Transcriptomic and proteomic levels of SIX1 in nontumor and tumour tissue according to sex. (A) mRNA levels were evaluated by microarray analysis (Agilent Whole Human Genome Oligo Microarrays). Expression levels in non-tumor tissue were non-significantly different between males and females. In the tumour tissue, SIX1 levels only moderately increased in males while in females a sharp increase was observed. Comparisons were performed by paired t-test between T and NT in males, and T and NT in females. T-test for independent groups was used to compared SIX1 in tumour tissue of males vs. females, as well as in non-tumor tissue of males vs. females. (*P

Figure 2.

Evaluation of growth speed (evaluated…

Figure 2.

Evaluation of growth speed (evaluated by the estimate of the doubling time) and…

Figure 2.
Evaluation of growth speed (evaluated by the estimate of the doubling time) and of survival in HCV-positive females and males in relation to low or high SIX1 expression. (A and B) Probability of having elevated growth speed in relation to SIX1 median values in (A) females and (B) males. HCC in females who had elevated SIX1 median values was characterised by significantly higher growth speed (P=0.003 by log-rank test). (C) HCV-positive females with higher median SIX1 levels had significantly lower survival in comparison with those with lower median levels (P=0.020, log-rank test). (D) No difference was present in males in relation to SIX1 levels. (E and F) Survival analysis was performed according to SIX1 proteomic expression in the tumour tissue of (E) females and (F) males. Data confirm survival analysis according to transcriptomic data. The probability of having elevated speed growth and survival was evaluated by the Kaplan-Meier method. For panel 2C and 2E two-stage hazard rate comparison method was used. DT, doubling time.
Figure 2.
Figure 2.
Evaluation of growth speed (evaluated by the estimate of the doubling time) and of survival in HCV-positive females and males in relation to low or high SIX1 expression. (A and B) Probability of having elevated growth speed in relation to SIX1 median values in (A) females and (B) males. HCC in females who had elevated SIX1 median values was characterised by significantly higher growth speed (P=0.003 by log-rank test). (C) HCV-positive females with higher median SIX1 levels had significantly lower survival in comparison with those with lower median levels (P=0.020, log-rank test). (D) No difference was present in males in relation to SIX1 levels. (E and F) Survival analysis was performed according to SIX1 proteomic expression in the tumour tissue of (E) females and (F) males. Data confirm survival analysis according to transcriptomic data. The probability of having elevated speed growth and survival was evaluated by the Kaplan-Meier method. For panel 2C and 2E two-stage hazard rate comparison method was used. DT, doubling time.

References

    1. Villa E, Baldini GM, Pasquinelli C, Melegari M, Cariani E, Di Chirico G, Manenti F. Risk factors for hepatocellular carcinoma in Italy. Male sex, hepatitis B virus, non-A non-B infection, and alcohol. Cancer. 1988;62:611–615. doi: 10.1002/1097-0142(19880801)62:3<611::AID-CNCR2820620328>;2-0.
    1. Zhang X, El-Serag HB, Thrift AP. Sex and race disparities in the incidence of hepatocellular carcinoma in the United States examined through age-period-cohort analysis. Cancer Epidemiol Biomarkers Prev. 2020;29:88–94. doi: 10.1158/1055-9965.EPI-19-1052.
    1. McGlynn KA, Petrick JL, El-Serag HB. Epidemiology of hepatocellular carcinoma. Hepatology. 2021;73((Suppl 1)):S4–S13. doi: 10.1002/hep.31288.
    1. Ma WL, Lai HC, Yeh S, Cai X, Chang C. Androgen receptor roles in hepatocellular carcinoma, cirrhosis, and hepatitis. Endocr Relat Cancer. 2014;21:R165–R182. doi: 10.1530/ERC-13-0283.
    1. Li Y, Xu A, Jia S, Huang J. Recent advances in the molecular mechanism of sex disparity in hepatocellular carcinoma. Oncol Lett. 2019;17:4222–4228.
    1. Villa E. Role of estrogen in liver cancer. Womens Health (Lond) 2008;4:41–50. doi: 10.2217/17455057.4.1.41.
    1. Shi L, Feng Y, Lin H, Ma R, Cai X. Role of estrogen in hepatocellular carcinoma: Is inflammation the key? J Transl Med. 2014;12:93. doi: 10.1186/1479-5876-12-93.
    1. Naugler WE, Sakurai T, Kim S, Maeda S, Kim K, Elsharkawy AM, Karin M. Gender disparity in liver cancer due to sex differences in MyD88-dependent IL-6 production. Science. 2007;317:121–124. doi: 10.1126/science.1140485.
    1. Villa E, Critelli R, Lei B, Marzocchi G, Cammà C, Giannelli G, Pontisso P, Cabibbo G, Enea M, Colopi S, et al. Neoangiogenesis-related genes are hallmarks of fast-growing hepatocellular carcinomas and worst survival. Results from a prospective study. Gut. 2016;65:861–869. doi: 10.1136/gutjnl-2014-308483.
    1. Berglund JA, Voelker R, Barber P, Diegel J, Mahady A, Bodner M. Oregon Univ Eugene; 2011. RNA regulation by estrogen.
    1. Li H, Han D, Hou Y, Chen H, Chen Z. Statistical inference methods for two crossing survival curves: A comparison of methods. PLoS One. 2015;10:e0116774. doi: 10.1371/journal.pone.0116774.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Martik ML, McClay DR. Deployment of a retinal determination gene network drives directed cell migration in the sea urchin embryo. Elife. 2015;4:e08827. doi: 10.7554/eLife.08827.
    1. Liu Y, Han N, Zhou S, Zhou R, Yuan X, Xu H, Zhang C, Yin T, Wu K. The DACH/EYA/SIX gene network and its role in tumor initiation and progression. Int J Cancer. 2016;138:1067–1075. doi: 10.1002/ijc.29560.
    1. Micalizzi DS, Christensen KL, Jedlicka P, Coletta CD, Barón AE, Harrel JC, Horwitz KB, Billheimer D, Heichman KA, Welm AL, et al. The Six1 homeoprotein induces human mammary carcinoma cells to undergo epithelial-mesenchymal transition and metastasis in mice through increasing TGF-beta signaling. J Clin Invest. 2009;119:2678–2690. doi: 10.1172/JCI37815.
    1. Micalizzi DS, Wang CA, Farabaugh SM, Schiemann WP, Ford HL. Homeoprotein Six1 increases TGF-beta type I receptor and converts TGF-beta signaling from suppressive to supportive for tumor growth. Cancer Res. 2010;70:10371–10380. doi: 10.1158/0008-5472.CAN-10-1354.
    1. Min WP, Wei XF. Silencing SIX1 inhibits epithelial mesenchymal transition through regulating TGF-β/Smad2/3 signaling pathway in papillary thyroid carcinoma. Auris Nasus Larynx. 2021;48:487–495. doi: 10.1016/j.anl.2020.10.002.
    1. Liu W, Gao M, Li L, Chen Y, Fan H, Cai Q, Shi Y, Pan C, Liu J, Cheng L, et al. Homeoprotein SIX1 compromises antitumor immunity through TGF-β-mediated regulation of collagens. Cell Mol Immunol. 2021;18:2660–2672. doi: 10.1038/s41423-021-00800-x.
    1. Critelli R, Milosa F, Faillaci F, Condello R, Turola E, Marzi L, Lei B, Dituri F, Andreani S, Sighinolfi P, et al. Microenvironment inflammatory infiltrate drives growth speed and outcome of hepatocellular carcinoma: A prospective clinical study. Cell Death Dis. 2017;8:e3017. doi: 10.1038/cddis.2017.395.
    1. Christensen KL, Patrick AN, McCoy EL, Ford HL. The six family of homeobox genes in development and cancer. Adv Cancer Res. 2008;101:93–126. doi: 10.1016/S0065-230X(08)00405-3.
    1. Behbakht K, Qamar L, Aldridge CS, Coletta RD, Davidson SA, Thorburn A, Ford HL. Six1 overexpression in ovarian carcinoma causes resistance to TRAIL-mediated apoptosis and is associated with poor survival. Cancer Res. 2007;67:3036–3042. doi: 10.1158/0008-5472.CAN-06-3755.
    1. Cheng Q, Ning D, Chen J, Li X, Chen XP, Jiang L. SIX1 and DACH1 influence the proliferation and apoptosis of hepatocellular carcinoma through regulating p53. Cancer Biol Ther. 2018;19:381–390. doi: 10.1080/15384047.2018.1423920.
    1. Ford HL, Kabingu EN, Bump EA, Mutter GL, Pardee AB. Abrogation of the G2 cell cycle checkpoint associated with overexpression of HSIX1: A possible mechanism of breast carcinogenesis. Proc Natl Acad Sci USA. 1998;95:12608–12613. doi: 10.1073/pnas.95.21.12608.
    1. Jin J, Jin T, Quan M, Piao Y, Lin Z. Ezrin overexpression predicts the poor prognosis of gastric adenocarcinoma. Diagn Pathol. 2012;7:135. doi: 10.1186/1746-1596-7-135.
    1. Kahlert C, Lerbs T, Pecqueux M, Herpel E, Hoffmeister M, Jansen L, Brenner H, Chang-Claude J, Bläker H, Kloor M, et al. Overexpression of SIX1 is an independent prognostic marker in stage I–III colorectal cancer. Int J Cancer. 2015;137:2104–2113. doi: 10.1002/ijc.29596.
    1. Kong J, Zhou X, Liu S, Jin T, Piao Y, Liu C, Lin Z. Overexpression of sineoculis homeobox homolog 1 predicts poor prognosis of hepatocellular carcinoma. Int J Clin Exp Pathol. 2014;7:3018–3027.
    1. Chen K, Wei H, Pan J, Chen Z, Pan D, Gao T, Huang J, Huang M, Ou M, Zhong W. Six1 is negatively correlated with poor prognosis and reduces 5-fluorouracil sensitivity via attenuating the stemness of hepatocellular carcinoma cells. Eur J Pharmacol. 2019;861:172599. doi: 10.1016/j.ejphar.2019.172599.
    1. Cui Q, Kong D, Li Z, Ahiable P, Wang K, Wu K, Wu G. Dachshund 1 is differentially expressed between male and female breast cancer: A matched case-control study of clinical characteristics and prognosis. Clinical Breast Cancer. 2018;18:e875–e882. doi: 10.1016/j.clbc.2018.01.011.
    1. Suen AA, Jefferson WN, Wood CE, Padilla-Banks E, Bae-Jump VL, Williams CJ. SIX1 oncoprotein as a biomarker in a model of hormonal carcinogenesis and in human endometrial cancer. Mol Cancer Res. 2016;14:849–858. doi: 10.1158/1541-7786.MCR-16-0084.
    1. Jefferson WN, Padilla-Banks E, Phelps JY, Gerrish KE, Williams CJ. Permanent oviduct posteriorization after neonatal exposure to the phytoestrogen genistein. Environ Health Perspect. 2011;119:1575–1582. doi: 10.1289/ehp.1104018.
    1. Jefferson WN, Chevalier DM, Phelps JY, Cantor AM, Padilla-Banks E, Newbold RR, Archer TK, Kinyamu HK, Williams CJ. Persistently altered epigenetic marks in the mouse uterus after neonatal estrogen exposure. Mol Endocrinol. 2013;27:1666–1677. doi: 10.1210/me.2013-1211.
    1. Rossini GP, Baldini GM, Villa E, Manenti F. Characterization of estrogen receptor from human liver. Gastroenterology. 1989;96:1102–1109. doi: 10.1016/0016-5085(89)91629-6.
    1. Villa E, Vukotic R, Cammà C, Petta S, Di Leo A, Gitto S, Turola E, Karampatou A, Losi L, Bernabucci V, et al. Reproductive status is associated with the severity of fibrosis in women with hepatitis C. PLoS One. 2012;7:e44624. doi: 10.1371/journal.pone.0044624.
    1. Meng F, Glaser SS, Francis H, DeMorrow S, Han Y, Passarini JD, Stokes A, Cleary JP, Liu X, Venter J, et al. Functional analysis of microRNAs in human hepatocellular cancer stem cells. J Cell Mol Med. 2012;16:160–173. doi: 10.1111/j.1582-4934.2011.01282.x.
    1. Wang B, Hsu SH, Majumder S, Kutay H, Huang W, Jacob ST, Ghoshal K. TGFbeta-mediated upregulation of hepatic miR-181b promotes hepatocarcinogenesis by targeting TIMP3. Oncogene. 2010;29:1787–1797. doi: 10.1038/onc.2009.468.
    1. Zhou Q, Zheng X, Chen L, Xu B, Yang X, Jiang J, Wu C. Smad2/3/4 pathway contributes to TGF-β-induced MiRNA-181b expression to promote gastric cancer metastasis by targeting Timp3. Cell Physiol Biochem. 2016;39:453–466. doi: 10.1159/000445638.
    1. Li B, Liu L, Li X, Wu L. miR-503 suppresses metastasis of hepatocellular carcinoma cell by targeting PRMT1. Biochem Biophys Res Commun. 2015;464:982–987. doi: 10.1016/j.bbrc.2015.06.169.
    1. Xiao Z, Shen J, Zhang L, Li M, Hu W, Cho C. Therapeutic targeting of noncoding RNAs in hepatocellular carcinoma: Recent progress and future prospects. Oncol Lett. 2018;15:3395–3402.
    1. Xiao Y, Tian Q, He J, Huang M, Yang C, Gong L. MiR-503 inhibits hepatocellular carcinoma cell growth via inhibition of insulin-like growth factor 1 receptor. Onco Targets Ther. 2016;9:3535–3544.
    1. Yang X, Zang J, Pan X, Yin J, Xiang Q, Yu J, Gan R, Lei X. miR-503 inhibits proliferation making human hepatocellular carcinoma cells susceptible to 5-fluorouracil by targeting EIF4E. Oncol Rep. 2017;37:563–570. doi: 10.3892/or.2016.5220.
    1. Qiu G, Lin Y, Zhang H, Wu D. miR-139-5p inhibits epithelial-mesenchymal transition, migration and invasion of hepatocellular carcinoma cells by targeting ZEB1 and ZEB2. Biochem Biophys Res Commun. 2015;463:315–321. doi: 10.1016/j.bbrc.2015.05.062.
    1. Li J, Fang L, Yu W, Wang Y. MicroRNA-125b suppresses the migration and invasion of hepatocellular carcinoma cells by targeting transcriptional coactivator with PDZ-binding motif. Oncol Lett. 2015;9:1971–1975. doi: 10.3892/ol.2015.2973.
    1. Yang J, Li Z, Pang Y, Zhou T, Sun J, Cheng XY, Zheng WV. MicroRNA-139-5p negatively regulates NME1 expression in hepatocellular carcinoma cells. Adv Clin Exp Med. 2022;31:655–670. doi: 10.17219/acem/146579.

Source: PubMed

3
Subskrybuj