Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01-12-2018 | Research

The epigenetically downregulated factor CYGB suppresses breast cancer through inhibition of glucose metabolism

Authors: Yixiao Feng, Mingjun Wu, Shuman Li, Xiaoqian He, Jun Tang, Weiyan Peng, Beilei Zeng, Chuxia Deng, Guosheng Ren, Tingxiu Xiang

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2018

Login to get access

Abstract

Background

Recent studies suggested the globin family member cytoglobin (CYGB) as a potential tumor suppressor; however, the mechanism by which CYGB suppresses cancer is elusive. We investigated the role and mechanism of CYGB in suppressing breast cancer.

Methods

CYGB expression was examined by reverse transcription PCR, quantitative reverse transcription PCR and open database analysis. Promoter methylation was examined by methylation-specific PCR. Metabolomics and proteomics were analyzed by gas chromatography-mass spectrometry and isobaric tags for relative and absolute quantitation, respectively. The effects and mechanisms of ectopic CYGB expression in breast cancer cells were assessed with molecular biological and cellular approaches in vitro and with a xenograft tumor model in nude mice.

Results

CYGB expression was downregulated in breast cancer tissues and cell lines, which was associated with promoter methylation. Ectopic CYGB expression suppressed proliferation, migration, invasion and induced apoptosis in breast cancer cell lines MCF7 (p53WT) and MB231 (p53mt) in vitro, and inhibited xenograft tumor growth in vivo. By proteomics and metabolomics analysis, glucose metabolism was found to be one of the main pathways suppressed by CYGB. The CYGB-expressing cells had lower ATP and compromised glycolysis. Additionally, CYGB suppressed key glucose metabolism factors including GLUT1 and HXK2 in p53-dependent and -independent manners. Restoration of GLUT1 or HXK2 expression attenuated CYGB-mediated proliferation suppression and apoptosis induction.

Conclusions

CYGB is a potential tumor suppressor in breast cancer that is epigenetically suppressed. The results for the first time suggest that CYGB suppresses breast cancer through inhibiting glucose metabolism, which could be exploited for breast cancer prevention and therapy.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference DeSantis CE, Fedewa SA, Goding Sauer A, Kramer JL, Smith RA, Jemal A. Breast cancer statistics, 2015: convergence of incidence rates between black and white women. CA Cancer J Clin. 2016;66(1):31–42.PubMedCrossRef DeSantis CE, Fedewa SA, Goding Sauer A, Kramer JL, Smith RA, Jemal A. Breast cancer statistics, 2015: convergence of incidence rates between black and white women. CA Cancer J Clin. 2016;66(1):31–42.PubMedCrossRef
3.
go back to reference Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.PubMedCrossRef Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66(2):115–32.PubMedCrossRef
4.
go back to reference Martens JW, Margossian AL, Schmitt M, Foekens J, Harbeck N. DNA methylation as a biomarker in breast cancer. Future Oncol. 2009;5(8):1245–56.PubMedCrossRef Martens JW, Margossian AL, Schmitt M, Foekens J, Harbeck N. DNA methylation as a biomarker in breast cancer. Future Oncol. 2009;5(8):1245–56.PubMedCrossRef
5.
go back to reference Palmisano WA, Crume KP, Grimes MJ, Winters SA, Toyota M, Esteller M, et al. Aberrant promoter methylation of the transcription factor genes PAX5 alpha and beta in human cancers. Cancer Res. 2003;63(15):4620–5.PubMed Palmisano WA, Crume KP, Grimes MJ, Winters SA, Toyota M, Esteller M, et al. Aberrant promoter methylation of the transcription factor genes PAX5 alpha and beta in human cancers. Cancer Res. 2003;63(15):4620–5.PubMed
6.
go back to reference Mu H, Wang N, Zhao L, Li S, Li Q, Chen L, et al. Methylation of PLCD1 and adenovirus-mediated PLCD1 overexpression elicits a gene therapy effect on human breast cancer. Exp Cell Res. 2015;332(2):179–89.PubMedCrossRef Mu H, Wang N, Zhao L, Li S, Li Q, Chen L, et al. Methylation of PLCD1 and adenovirus-mediated PLCD1 overexpression elicits a gene therapy effect on human breast cancer. Exp Cell Res. 2015;332(2):179–89.PubMedCrossRef
7.
go back to reference Yu JS, Koujak S, Nagase S, Li CM, Su T, Wang X, et al. PCDH8, the human homolog of PAPC, is a candidate tumor suppressor of breast cancer. Oncogene. 2008;27(34):4657–65.PubMedCrossRef Yu JS, Koujak S, Nagase S, Li CM, Su T, Wang X, et al. PCDH8, the human homolog of PAPC, is a candidate tumor suppressor of breast cancer. Oncogene. 2008;27(34):4657–65.PubMedCrossRef
8.
go back to reference Fordel E, Geuens E, Dewilde S, Rottiers P, Carmeliet P, Grooten J, et al. Cytoglobin expression is upregulated in all tissues upon hypoxia: an in vitro and in vivo study by quantitative real-time PCR. Biochem Biophys Res Commun. 2004;319(2):342–8.PubMedCrossRef Fordel E, Geuens E, Dewilde S, Rottiers P, Carmeliet P, Grooten J, et al. Cytoglobin expression is upregulated in all tissues upon hypoxia: an in vitro and in vivo study by quantitative real-time PCR. Biochem Biophys Res Commun. 2004;319(2):342–8.PubMedCrossRef
9.
go back to reference Nishi H, Inagi R, Kawada N, Yoshizato K, Mimura I, Fujita T, et al. Cytoglobin, a novel member of the globin family, protects kidney fibroblasts against oxidative stress under ischemic conditions. Am J Pathol. 2011;178(1):128–39.PubMedCrossRef Nishi H, Inagi R, Kawada N, Yoshizato K, Mimura I, Fujita T, et al. Cytoglobin, a novel member of the globin family, protects kidney fibroblasts against oxidative stress under ischemic conditions. Am J Pathol. 2011;178(1):128–39.PubMedCrossRef
10.
go back to reference Zhang S, Li X, Jourd'heuil FL, Qu S, Devejian N, Bennett E, et al. Cytoglobin promotes cardiac progenitor cell survival against oxidative stress via the upregulation of the NFkappaB/iNOS signal pathway and nitric oxide production. Sci Rep. 2017;7(1):10754.PubMedCrossRef Zhang S, Li X, Jourd'heuil FL, Qu S, Devejian N, Bennett E, et al. Cytoglobin promotes cardiac progenitor cell survival against oxidative stress via the upregulation of the NFkappaB/iNOS signal pathway and nitric oxide production. Sci Rep. 2017;7(1):10754.PubMedCrossRef
11.
go back to reference le Thuy TT, Van Thuy TT, Matsumoto Y, Hai H, Ikura Y, Yoshizato K, et al. Absence of cytoglobin promotes multiple organ abnormalities in aged mice. Sci Rep. 2016;6:24990.PubMedCrossRef le Thuy TT, Van Thuy TT, Matsumoto Y, Hai H, Ikura Y, Yoshizato K, et al. Absence of cytoglobin promotes multiple organ abnormalities in aged mice. Sci Rep. 2016;6:24990.PubMedCrossRef
12.
go back to reference Shaw RJ, Omar MM, Rokadiya S, Kogera FA, Lowe D, Hall GL, et al. Cytoglobin is upregulated by tumour hypoxia and silenced by promoter hypermethylation in head and neck cancer. Br J Cancer. 2009;101(1):139–44.PubMedCrossRef Shaw RJ, Omar MM, Rokadiya S, Kogera FA, Lowe D, Hall GL, et al. Cytoglobin is upregulated by tumour hypoxia and silenced by promoter hypermethylation in head and neck cancer. Br J Cancer. 2009;101(1):139–44.PubMedCrossRef
13.
go back to reference Shivapurkar N, Stastny V, Okumura N, Girard L, Xie Y, Prinsen C, et al. Cytoglobin, the newest member of the globin family, functions as a tumor suppressor gene. Cancer Res. 2008;68(18):7448–56.PubMedCrossRef Shivapurkar N, Stastny V, Okumura N, Girard L, Xie Y, Prinsen C, et al. Cytoglobin, the newest member of the globin family, functions as a tumor suppressor gene. Cancer Res. 2008;68(18):7448–56.PubMedCrossRef
14.
go back to reference le Thuy TT, Morita T, Yoshida K, Wakasa K, Iizuka M, Ogawa T, et al. Promotion of liver and lung tumorigenesis in DEN-treated cytoglobin-deficient mice. Am J Pathol. 2011;179(2):1050–60.CrossRef le Thuy TT, Morita T, Yoshida K, Wakasa K, Iizuka M, Ogawa T, et al. Promotion of liver and lung tumorigenesis in DEN-treated cytoglobin-deficient mice. Am J Pathol. 2011;179(2):1050–60.CrossRef
15.
go back to reference Oleksiewicz U, Liloglou T, Tasopoulou KM, Daskoulidou N, Bryan J, Gosney JR, et al. Cytoglobin has bimodal: tumour suppressor and oncogene functions in lung cancer cell lines. Hum Mol Genet. 2013;22(16):3207–17.PubMedCrossRef Oleksiewicz U, Liloglou T, Tasopoulou KM, Daskoulidou N, Bryan J, Gosney JR, et al. Cytoglobin has bimodal: tumour suppressor and oncogene functions in lung cancer cell lines. Hum Mol Genet. 2013;22(16):3207–17.PubMedCrossRef
16.
go back to reference John R, Chand V, Chakraborty S, Jaiswal N, Nag A. DNA damage induced activation of Cygb stabilizes p53 and mediates G1 arrest. DNA Repair (Amst). 2014;24:107–12.CrossRef John R, Chand V, Chakraborty S, Jaiswal N, Nag A. DNA damage induced activation of Cygb stabilizes p53 and mediates G1 arrest. DNA Repair (Amst). 2014;24:107–12.CrossRef
17.
go back to reference Labuschagne CF, Zani F, Vousden KH. Control of metabolism by p53-Cancer and beyond. Biochim Biophys Acta Rev Cancer. 2018;1870(1):32–42. Labuschagne CF, Zani F, Vousden KH. Control of metabolism by p53-Cancer and beyond. Biochim Biophys Acta Rev Cancer. 2018;1870(1):32–42.
18.
go back to reference Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res. 2004;64(7):2627–33.PubMedCrossRef Schwartzenberg-Bar-Yoseph F, Armoni M, Karnieli E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res. 2004;64(7):2627–33.PubMedCrossRef
19.
go back to reference Shao B, Feng Y, Zhang H, Yu F, Li Q, Tan C, et al. The 3p14.2 tumour suppressor ADAMTS9 is inactivated by promoter CpG methylation and inhibits tumour cell growth in breast cancer. J Cell Mol Med. 2018;22(2):1257–71.PubMed Shao B, Feng Y, Zhang H, Yu F, Li Q, Tan C, et al. The 3p14.2 tumour suppressor ADAMTS9 is inactivated by promoter CpG methylation and inhibits tumour cell growth in breast cancer. J Cell Mol Med. 2018;22(2):1257–71.PubMed
20.
go back to reference Xiang T, Li L, Fan Y, Jiang Y, Ying Y, Putti TC, et al. PLCD1 is a functional tumor suppressor inducing G(2)/M arrest and frequently methylated in breast cancer. Cancer Biol Ther. 2010;10(5):520–7.PubMedCrossRef Xiang T, Li L, Fan Y, Jiang Y, Ying Y, Putti TC, et al. PLCD1 is a functional tumor suppressor inducing G(2)/M arrest and frequently methylated in breast cancer. Cancer Biol Ther. 2010;10(5):520–7.PubMedCrossRef
21.
go back to reference Yin X, Xiang T, Li L, Su X, Shu X, Luo X, et al. DACT1, an antagonist to Wnt/beta-catenin signaling, suppresses tumor cell growth and is frequently silenced in breast cancer. Breast Cancer Res. 2013;15(2):R23.PubMedCrossRef Yin X, Xiang T, Li L, Su X, Shu X, Luo X, et al. DACT1, an antagonist to Wnt/beta-catenin signaling, suppresses tumor cell growth and is frequently silenced in breast cancer. Breast Cancer Res. 2013;15(2):R23.PubMedCrossRef
22.
go back to reference Zhao L, Li S, Gan L, Li C, Qiu Z, Feng Y, et al. Paired box 5 is a frequently methylated lung cancer tumour suppressor gene interfering beta-catenin signalling and GADD45G expression. J Cell Mol Med. 2016;20(5):842–54.PubMedCrossRef Zhao L, Li S, Gan L, Li C, Qiu Z, Feng Y, et al. Paired box 5 is a frequently methylated lung cancer tumour suppressor gene interfering beta-catenin signalling and GADD45G expression. J Cell Mol Med. 2016;20(5):842–54.PubMedCrossRef
23.
go back to reference Yin X, Xiang T, Mu J, Mao H, Li L, Huang X, et al. Protocadherin 17 functions as a tumor suppressor suppressing Wnt/beta-catenin signaling and cell metastasis and is frequently methylated in breast cancer. Oncotarget. 2016;7(32):51720–32.PubMedCrossRef Yin X, Xiang T, Mu J, Mao H, Li L, Huang X, et al. Protocadherin 17 functions as a tumor suppressor suppressing Wnt/beta-catenin signaling and cell metastasis and is frequently methylated in breast cancer. Oncotarget. 2016;7(32):51720–32.PubMedCrossRef
24.
go back to reference Liu Y, Pu QH, Wu MJ, Yu C. Proteomic analysis for the impact of hypercholesterolemia on expressions of hepatic drug transporters and metabolizing enzymes. Xenobiotica. 2016;46(10):940–7.PubMedCrossRef Liu Y, Pu QH, Wu MJ, Yu C. Proteomic analysis for the impact of hypercholesterolemia on expressions of hepatic drug transporters and metabolizing enzymes. Xenobiotica. 2016;46(10):940–7.PubMedCrossRef
25.
go back to reference Xinarianos G, McRonald FE, Risk JM, Bowers NL, Nikolaidis G, Field JK, et al. Frequent genetic and epigenetic abnormalities contribute to the deregulation of cytoglobin in non-small cell lung cancer. Hum Mol Genet. 2006;15(13):2038–44.PubMedCrossRef Xinarianos G, McRonald FE, Risk JM, Bowers NL, Nikolaidis G, Field JK, et al. Frequent genetic and epigenetic abnormalities contribute to the deregulation of cytoglobin in non-small cell lung cancer. Hum Mol Genet. 2006;15(13):2038–44.PubMedCrossRef
26.
go back to reference Mai WX, Gosa L, Daniels VW, Ta L, Tsang JE, Higgins B, et al. Cytoplasmic p53 couples oncogene-driven glucose metabolism to apoptosis and is a therapeutic target in glioblastoma. Nat Med. 2017;23(11):1342–51.PubMedPubMedCentral Mai WX, Gosa L, Daniels VW, Ta L, Tsang JE, Higgins B, et al. Cytoplasmic p53 couples oncogene-driven glucose metabolism to apoptosis and is a therapeutic target in glioblastoma. Nat Med. 2017;23(11):1342–51.PubMedPubMedCentral
27.
go back to reference Gomes AS, Ramos H, Soares J, Saraiva L. p53 and glucose metabolism: an orchestra to be directed in cancer therapy. Pharmacol Res. 2018;131:75–86.PubMedCrossRef Gomes AS, Ramos H, Soares J, Saraiva L. p53 and glucose metabolism: an orchestra to be directed in cancer therapy. Pharmacol Res. 2018;131:75–86.PubMedCrossRef
28.
go back to reference Xiang T, Li L, Yin X, Zhong L, Peng W, Qiu Z, et al. Epigenetic silencing of the WNT antagonist Dickkopf 3 disrupts normal Wnt/beta-catenin signalling and apoptosis regulation in breast cancer cells. J Cell Mol Med. 2013;17(10):1236–46.PubMedCrossRef Xiang T, Li L, Yin X, Zhong L, Peng W, Qiu Z, et al. Epigenetic silencing of the WNT antagonist Dickkopf 3 disrupts normal Wnt/beta-catenin signalling and apoptosis regulation in breast cancer cells. J Cell Mol Med. 2013;17(10):1236–46.PubMedCrossRef
29.
go back to reference Xiao Y, Xiang T, Luo X, Li C, Li Q, Peng W, et al. Zinc-finger protein 545 inhibits cell proliferation as a tumor suppressor through inducing apoptosis and is disrupted by promoter methylation in breast cancer. PLoS One. 2014;9(10):e110990.PubMedCrossRef Xiao Y, Xiang T, Luo X, Li C, Li Q, Peng W, et al. Zinc-finger protein 545 inhibits cell proliferation as a tumor suppressor through inducing apoptosis and is disrupted by promoter methylation in breast cancer. PLoS One. 2014;9(10):e110990.PubMedCrossRef
30.
go back to reference Daskalos A, Oleksiewicz U, Filia A, Nikolaidis G, Xinarianos G, Gosney JR, et al. UHRF1-mediated tumor suppressor gene inactivation in nonsmall cell lung cancer. Cancer. 2011;117(5):1027–37.PubMedCrossRef Daskalos A, Oleksiewicz U, Filia A, Nikolaidis G, Xinarianos G, Gosney JR, et al. UHRF1-mediated tumor suppressor gene inactivation in nonsmall cell lung cancer. Cancer. 2011;117(5):1027–37.PubMedCrossRef
31.
go back to reference le Thuy TT, Matsumoto Y, Thuy TT, Hai H, Suoh M, Urahara Y, et al. Cytoglobin deficiency promotes liver cancer development from hepatosteatosis through activation of the oxidative stress pathway. Am J Pathol. 2015;185(4):1045–60.PubMedCrossRef le Thuy TT, Matsumoto Y, Thuy TT, Hai H, Suoh M, Urahara Y, et al. Cytoglobin deficiency promotes liver cancer development from hepatosteatosis through activation of the oxidative stress pathway. Am J Pathol. 2015;185(4):1045–60.PubMedCrossRef
32.
go back to reference Wojnarowicz PM, Provencher DM, Mes-Masson AM, Tonin PN. Chromosome 17q25 genes, RHBDF2 and CYGB, in ovarian cancer. Int J Oncol. 2012;40(6):1865–80.PubMed Wojnarowicz PM, Provencher DM, Mes-Masson AM, Tonin PN. Chromosome 17q25 genes, RHBDF2 and CYGB, in ovarian cancer. Int J Oncol. 2012;40(6):1865–80.PubMed
33.
go back to reference Emara M, Turner AR, Allalunis-Turner J. Hypoxic regulation of cytoglobin and neuroglobin expression in human normal and tumor tissues. Cancer Cell Int. 2010;10:33.PubMedCrossRef Emara M, Turner AR, Allalunis-Turner J. Hypoxic regulation of cytoglobin and neuroglobin expression in human normal and tumor tissues. Cancer Cell Int. 2010;10:33.PubMedCrossRef
34.
go back to reference Demirci S, Dogan A, Apdik H, Tuysuz EC, Gulluoglu S, Bayrak OF, et al. Cytoglobin inhibits migration through PI3K/AKT/mTOR pathway in fibroblast cells. Mol Cell Biochem. 2018;437(1–2):133–42.PubMedCrossRef Demirci S, Dogan A, Apdik H, Tuysuz EC, Gulluoglu S, Bayrak OF, et al. Cytoglobin inhibits migration through PI3K/AKT/mTOR pathway in fibroblast cells. Mol Cell Biochem. 2018;437(1–2):133–42.PubMedCrossRef
35.
go back to reference Madan E, Gogna R, Bhatt M, Pati U, Kuppusamy P, Mahdi AA. Regulation of glucose metabolism by p53: emerging new roles for the tumor suppressor. Oncotarget. 2011;2(12):948–57.PubMedCrossRef Madan E, Gogna R, Bhatt M, Pati U, Kuppusamy P, Mahdi AA. Regulation of glucose metabolism by p53: emerging new roles for the tumor suppressor. Oncotarget. 2011;2(12):948–57.PubMedCrossRef
36.
go back to reference Liu J, Zhang C, Hu W, Feng Z. Tumor suppressor p53 and its mutants in cancer metabolism. Cancer Lett. 2015;356(2 Pt A):197–203.PubMedCrossRef Liu J, Zhang C, Hu W, Feng Z. Tumor suppressor p53 and its mutants in cancer metabolism. Cancer Lett. 2015;356(2 Pt A):197–203.PubMedCrossRef
Metadata
Title
The epigenetically downregulated factor CYGB suppresses breast cancer through inhibition of glucose metabolism
Authors
Yixiao Feng
Mingjun Wu
Shuman Li
Xiaoqian He
Jun Tang
Weiyan Peng
Beilei Zeng
Chuxia Deng
Guosheng Ren
Tingxiu Xiang
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2018
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0979-9

Other articles of this Issue 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine