Cholesterol de novo Biosynthesis in Paired Samples of Breast Cancer and Adjacent Histologically Normal Tissue: Association with Proliferation Index, Tumor Grade, and Recurrence-Free Survival De novo cholesterol biosynthesis in breast tissue

Danila Coradini (1), Federico Ambrogi (2), Gabriele Infante (3)
(1) Laboratory of Medical Statistics and Biometry, Department of Clinical Sciences and Community Health, Campus Cascina Rosa, University of Milan, Italy, Italy,
(2) Laboratory of Medical Statistics and Biometry, Department of Clinical Sciences and Community Health, Campus Cascina Rosa, University of Milan, Italy, Scientific Directorate, IRCCS Policlinico San Donato, San Donato Milanese, Italy, Italy,
(3) Laboratory of Medical Statistics and Biometry, Department of Clinical Sciences and Community Health, Clinical Epidemiology and Trial Organization Unit, Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy, Italy

Abstract

Background: Cholesterol is an essential component of cell membranes whose local de novo biosynthesis may occur in response to additional cellular requirements, especially cell proliferation. In this study, we investigated: (1) the differential expression of the genes coding for the main enzymes involved in cholesterol biosynthesis (ACAT2, HMGCS1, HMGCR, FDFT1, SQLE, LSS, and NSDHL), or the proteins that control their activity (SREBF2, SCAP, and INSIG1), in patient-matched samples of breast cancer and adjacent histologically normal (HN) tissue; (2) their association with the expression of MKI67 or the histologic tumor grade (in particular, G2); (3) their association with recurrence-free survival (RFS).


Methods: Nonparametric rank-based models for longitudinal data were applied to assess the differential gene expression between the tumor and the adjacent HN tissue from the same patient or between the classes of tumor grade. Spearman’s rank correlation and Cox proportional hazard models were used to assess the correlation between the genes and their association with RFS.


Results: Compared to the adjacent HN tissue, HMGCS1, HMGCR, SQLE, and NSDHL genes were more expressed in the tumor. Their expression progressively increased according to tumor grade and correlated positively with MKI67. ACAT2, HMGCR, and NSDHL genes were associated with a high risk of recurrence even when adjusted for age, tumor grade, or immunohistochemical Ki67.


Conclusion: The findings indicated that some genes involved in cholesterol biosynthesis were more expressed in cancerous tissue, correlated positively with tumor grade and MKI67 expression, and were associated with RFS, thus substantiating the relationship between de novo cholesterol biosynthesis and tumor aggressiveness.

Full text article

Generated from XML file

References

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-674. doi: 10.1016/j.cell.2011.02.013.

Allred DC, Harvey JM, Berardo M, Clark GM. Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol. 1998;11(2):155-68.

Early Breast Cancer Trialists’ Collaborative Group. Tamoxifen for early breast cancer: an overview of the randomised trials. Lancet. 1998;351(9114):1451-67.

Yerushalmi R, Woods R, Ravdin PM, Hayes MM, Gelmon KA. Ki67 in breast cancer: prognostic and predictive potential. Lancet Oncol. 2010;11(12):174-83. doi: 10.1016/S1470-2045(09)70262-1.

Dowsett M, Nielsen TO, A'Hern R, Bartlett J, Coombes RC, Cuzick J, et al. Assessment of Ki67 in breast cancer: recommendations from the International Ki67 in Breast Cancer Working Group. J Natl Cancer Inst. 2011;103(22):1656-64. doi: 10.1093/jnci/djr393.

Scholzen T, Gerdes J. The Ki-67 protein: from the known and the unknown. J Cell Physiol. 2000;182(3):311-22. doi: 10.1002/(SICI)1097-4652(200003)182:3<311::AID-JCP1>3.0.CO;2-9.

Elston CW, Ellis IO. Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology. 1991;19(5):403-10. doi: 10.1111/j.1365-2559.1991.tb00229.x.

Sotiriou C, Wirapati P, Loi S, Harris A, Fox S, Smeds J, et al. Gene expression profiling in breast cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst. 2006;98(4):262-72. doi: 10.1093/jnci/djj052.

Matikas A, Foukakis T, Swain S, Bergh J. Avoiding over- and undertreatment in patients with resected node-positive breast cancer with the use of gene expression signatures: are we there yet? Ann Oncol. 2019;30(7):1044-50. doi: 10.1093/annonc/mdz126.

Griffiths B, Lewis CA, Bensaad K, Ros S, Zhang Q, Ferber EC, et al. Sterol regulatory element binding protein-dependent regulation of lipid synthesis supports cell survival and tumor growth. Cancer Metab 2013;1(1):3. doi: 10.1186/2049-3002-1-3.

Clendening JW, Pandyra A, Boutros PC, El Ghamrasni S, Khosravi F, Trentin GA, et al. Dysregulation of the mevalonate pathway promotes transformation. Proc Natl Acad Sci. 2010;107(34):15051-6. doi: 10.1073/pnas.0910258107.

Llaverias G, Danilo C, Mercier I, Daumer K, Capozza F, Williams TM, et al. Role of cholesterol in the development and progression of breast cancer. Am J Pathol. 2011;178(1):402-12. doi: 10.1016/j.ajpath.2010.11.005.

Gustbée E, Tryggvadottir H, Markkula A, Simonsson M, Nodin B, Jirström K, et al. Tumor-specific expression of HMG-CoA reductase in a population-based cohort of breast cancer patients. BMC Clin Pathol. 2015;15:8. doi: 10.1186/s12907-015-0008-2.

Tamimi RM, Baer HJ, Marotti J, Galan M, Galaburda L, Fu Y, et al. Comparison of molecular phenotypes of ductal carcinoma in situ and invasive breast cancer. Breast Cancer Res. 2008;10(4):R67. doi: 10.1186/bcr2128.

Sisti JS, Collins LC, Beck AH, Tamimi RM, Rosner BA, Eliassen AH. Reproductive risk factors in relation to molecular subtypes of breast cancer: Results from the nurses' health studies. Int J Cancer. 2016;138(10):2346-56. doi: 10.1002/ijc.29968.

Wang J, Zhang X, Beck AH, Collins LC, Chen WY, Tamim RM, et al. Alcohol Consumption and Risk of Breast Cancer by Tumor Receptor Expression Horm Cancer. 2015; 6(0): 237–246. doi: 10.1007/s12672-015-0235-0.

Brunner E, Bathke AC, Konietschke F. Two-Factor Crossed Designs. In: Rank and pseudo-rank procedures for independent observations in factorial designs. Springer International Publishing. 2018;263-331. doi: 10.1007/978-3-030-02914-2.

Brunner E, Dette H, Munk A. Box-type approximations in nonparametric factorial designs. J Am Stat Ass. 1997;92(440):1494-502. doi: 10.1080/01621459.1997.10473671.

Walsh CA, Akrap N, Garre E, Magnusson Y, Harrison H, Andersson D, et al. The mevalonate precursor enzyme HMGCS1 is a novel marker and key mediator of cancer stem cell enrichment in luminal and basal models of breast cancer. PLoS One. 2020;15(7):e0236187. doi: 10.1371/journal.pone.0236187.

Gill S, Stevenson J, Kristiana I, Brown AJ. Cholesterol-dependent degradation of squalene monooxygenase, a control point in cholesterol synthesis beyond HMG-CoA reductase. Cell Metab. 2011;13(3):260-73. doi: 10.1016/j.cmet.2011.01.015.

Yoon SH, Kim HS, Kim RN, Jung SY, Hong BS, Kang EJ, et al. NAD(P)-dependent steroid dehydrogenase-like is involved in breast cancer cell growth and metastasis. BMC Cancer. 2020;20(1):375. doi: 10.1186/s12885-020-06840-2.

Xue T, Zhang Y, Zhang L, Yao L, Hu X, Xu LX. Proteomic analysis of two metabolic proteins with potential to translocate to plasma membrane associated with tumor metastasis development and drug targets. J Proteome Res. 2013;12(4):1754-63. doi: 10.1021/pr301100r.

Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y, Aebersold R, et al. Crucial step in cholesterol homeostasis: sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell. 2002;110(4):489-500. doi: 10.1016/s0092-8674(02)00872-3.

Sorrentino G, Ruggeri N, Specchia V, Cordenonsi M, Mano M, Dupont S, et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat Cell Biol. 2014;16(4):357-66. doi: 10.1038/ncb2936.

Zhao B, Wei X, Li W, Udan RS, Yang Q, Kim J, et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 2007;21(21): 2747-61. doi: 10.1101/gad.1602907.

Lei QY, Zhang H, Zhao B, Zha ZY, Bai F, Pei XH, et al. TAZ promotes cell proliferation and epithelial-mesenchymal transition and is inhibited by the hippo pathway. Mol Cell Biol. 2008;28(7):2426-36. doi: 10.1128/MCB.01874-07.

Hao Y, Chun A, Cheung K, Rashidi B, Yang X. Tumor suppressor LATS1 is a negative regulator of oncogene YAP. J Biol Chem. 2008;283(9):5496-5509. doi: 10.1074/jbc.M709037200.

Authors

Danila Coradini
danila.coradini@gmail.com (Primary Contact)
Federico Ambrogi
Gabriele Infante
1.
Coradini D, Ambrogi F, Infante G. Cholesterol de novo Biosynthesis in Paired Samples of Breast Cancer and Adjacent Histologically Normal Tissue: Association with Proliferation Index, Tumor Grade, and Recurrence-Free Survival: De novo cholesterol biosynthesis in breast tissue. Arch Breast Cancer [Internet]. 2023 Mar. 11 [cited 2024 Jul. 16];10(2):187-99. Available from: https://archbreastcancer.com/index.php/abc/article/view/690

Article Details