Matched analysis of circulating selenium with the breast cancer selenotranscriptome: a multicentre prospective study

Kamil Demircan, Ylva Bengtsson, Thilo Samson Chillon, Johan Vallon-Christersson, Qian Sun, Christer Larsson, Martin Malmberg, Lao H Saal, Lisa Rydén, Åke Borg, Jonas Manjer, Lutz Schomburg, Kamil Demircan, Ylva Bengtsson, Thilo Samson Chillon, Johan Vallon-Christersson, Qian Sun, Christer Larsson, Martin Malmberg, Lao H Saal, Lisa Rydén, Åke Borg, Jonas Manjer, Lutz Schomburg

Abstract

Introduction: Low serum selenium and altered tumour RNA expression of certain selenoproteins are associated with a poor breast cancer prognosis. Selenoprotein expression stringently depends on selenium availability, hence circulating selenium may interact with tumour selenoprotein expression. However, there is no matched analysis to date.

Methods: This study included 1453 patients with newly diagnosed breast cancer from the multicentric prospective Sweden Cancerome Analysis Network - Breast study. Total serum selenium, selenoprotein P and glutathione peroxidase 3 were analysed at time of diagnosis. Bulk RNA-sequencing was conducted in matched tumour tissues. Fully adjusted Cox regression models with an interaction term were employed to detect dose-dependent interactions of circulating selenium with the associations of tumour selenoprotein mRNA expression and mortality.

Results: 237 deaths were recorded within ~ 9 years follow-up. All three serum selenium biomarkers correlated positively (p < 0.001). All selenoproteins except for GPX6 were expressed in tumour tissues. Single cell RNA-sequencing revealed a heterogeneous expression pattern in the tumour microenvironment. Circulating selenium correlated positively with tumour SELENOW and SELENON expression (p < 0.001). In fully adjusted models, the associations of DIO1, DIO3 and SELENOM with mortality were dose-dependently modified by serum selenium (p < 0.001, p = 0.020, p = 0.038, respectively). With increasing selenium, DIO1 and SELENOM associated with lower, whereas DIO3 expression associated with higher mortality. Association of DIO1 with lower mortality was only apparent in patients with high selenium [above median (70.36 µg/L)], and the HR (95%CI) for one-unit increase in log(FPKM + 1) was 0.70 (0.50-0.98).

Conclusions: This first unbiased analysis of serum selenium with the breast cancer selenotranscriptome identified an effect-modification of selenium on the associations of DIO1, SELENOM, and DIO3 with prognosis. Selenium substitution in patients with DIO1-expressing tumours merits consideration to improve survival.

Keywords: Glutathione peroxidase; Prognosis; SELENOP; Selenoproteins; Thyroid hormones.

Conflict of interest statement

LS holds shares of selenOmed GmbH, a company involved in selenium status measurement. The remaining authors have no competing interests to declare.

© 2023. BioMed Central Ltd., part of Springer Nature.

Figures

Fig. 1
Fig. 1
 A Study scheme. B Gene expression of selenoproteins in tumour samples of 1453 patients. C Spearman’s correlation matrix of tumour gene expression of selenoprotein genes and circulating selenium biomarker concentrations. D Spearman’s correlation of serum biomarkers with each other and serum biomarkers with SELENOW expression in the tumour. E Single cell RNA-expression of selenoprotein genes in different cells in breast cancer
Fig. 2
Fig. 2
 A Analysis scheme. B Cox regression models in the whole cohort and in low and high selenium subsets, divided according to median selenium concentration of the cohort, i.e. 70.36 µg/L. All models were adjusted for age, tumour size, histological grade, lymph node involvement, expression of HER2/ER/PGR-Receptor, laterality of the tumour, and histological type. P for interaction was tested by adding an interaction term between serum selenium and the gene of interest, marked by purple asterisk
Fig. 3
Fig. 3
 A Contour plot of the interaction between DIO1 expression and serum selenium concentrations on mortality. B Cox regression models depicting the association of DIO1 expression with mortality according to 10th, 50th and 90th quantiles of circulating selenium. C Contour plot of the interaction between DIO3 expression and serum selenium concentrations on mortality. D Cox regression models depicting the association of DIO3 expression with mortality according to 10th, 50th and 90th quantiles of circulating selenium. E Contour plot of the interaction between SELENOM expression and serum selenium concentrations on mortality. F Cox regression models depicting the association of SELENOM expression with mortality according to 10th, 50th and 90th quantiles of circulating selenium. All models were adjusted for age, tumour size, histological grade, lymph node involvement, expression of HER2/ER/PGR-Receptor, laterality of the tumour, and histological type

References

    1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–249.
    1. Dowsett M, Dunbier AK. Emerging biomarkers and new understanding of traditional markers in personalized therapy for breast cancer. Clin Cancer Res. 2008;14(24):8019–8026.
    1. Flowers B, Poles A, Kastrati I. Selenium and breast cancer: an update of clinical and epidemiological data. Arch Biochem Biophys. 2022;732:109465.
    1. Demircan K, Sun Q, Bengtsson Y, Seemann P, Vallon-Christersson J, Malmberg M, et al. Autoimmunity to selenoprotein P predicts breast cancer recurrence. Redox Biol. 2022;53:102346.
    1. Demircan K, Bengtsson Y, Sun Q, Brange A, Vallon-Christersson J, Rijntjes E, et al. Serum selenium, selenoprotein P and glutathione peroxidase 3 as predictors of mortality and recurrence following breast cancer diagnosis: a multicentre cohort study. Redox Biol. 2021;47:102145.
    1. Sandsveden M, Nilsson E, Borgquist S, Rosendahl AH, Manjer J. Prediagnostic serum selenium levels in relation to breast cancer survival and tumor characteristics. Int J Cancer. 2020;147(9):2424–2436.
    1. Lubinski J, Marciniak W, Muszynska M, Huzarski T, Gronwald J, Cybulski C, et al. Serum selenium levels predict survival after breast cancer. Breast Cancer Res Treat. 2018;167(2):591–598.
    1. Harris HR, Bergkvist L, Wolk A. Selenium intake and breast cancer mortality in a cohort of swedish women. Breast Cancer Res Treat. 2012;134(3):1269–1277.
    1. Labunskyy VM, Hatfield DL, Gladyshev VN. Selenoproteins: molecular pathways and physiological roles. Physiol Rev. 2014;94(3):739–777.
    1. Schomburg L. Selenium, selenoproteins and the thyroid gland: interactions in health and disease. Nat Rev Endocrinol. 2012;8(3):160–171.
    1. Steinbrenner H, Speckmann B, Klotz LO. Selenoproteins: antioxidant selenoenzymes and beyond. Arch Biochem Biophys. 2016;595:113–119.
    1. Xia Y, Hill KE, Li P, Xu J, Zhou D, Motley AK, et al. Optimization of selenoprotein P and other plasma selenium biomarkers for the assessment of the selenium nutritional requirement: a placebo-controlled, double-blind study of selenomethionine supplementation in selenium-deficient chinese subjects. Am J Clin Nutr. 2010;92(3):525–531.
    1. Hoffmann PR, Höge SC, Li PA, Hoffmann FW, Hashimoto AC, Berry MJ. The selenoproteome exhibits widely varying, tissue-specific dependence on selenoprotein P for selenium supply. Nucleic Acids Res. 2007;35(12):3963–3973.
    1. Goemann IM, Marczyk VR, Recamonde-Mendoza M, Wajner SM, Graudenz MS, Maia AL. Decreased expression of the thyroid hormone-inactivating enzyme type 3 deiodinase is associated with lower survival rates in breast cancer. Sci Rep. 2020;10(1):13914.
    1. Lou W, Ding B, Wang S, Fu P. Overexpression of GPX3, a potential biomarker for diagnosis and prognosis of breast cancer, inhibits progression of breast cancer cells in vitro. Cancer Cell Int. 2020;20(1):378.
    1. Cadenas C, Franckenstein D, Schmidt M, Gehrmann M, Hermes M, Geppert B, et al. Role of thioredoxin reductase 1 and thioredoxin interacting protein in prognosis of breast cancer. Breast Cancer Res. 2010;12(3):R44.
    1. Li Z, Ferguson L, Deol KK, Roberts MA, Magtanong L, Hendricks JM, et al. Ribosome stalling during selenoprotein translation exposes a ferroptosis vulnerability. Nat Chem Biol. 2022;18(7):751–761.
    1. Hilal T, Killam BY, Grozdanović M, Dobosz-Bartoszek M, Loerke J, Bürger J, et al. Structure of the mammalian ribosome as it decodes the selenocysteine UGA codon. Science. 2022;376(6599):1338–1343.
    1. Saal LH, Vallon-Christersson J, Häkkinen J, Hegardt C, Grabau D, Winter C, et al. The Sweden Cancerome Analysis network-breast (SCAN-B) Initiative: a large-scale multicenter infrastructure towards implementation of breast cancer genomic analyses in the clinical routine. Genome Med. 2015;7(1):20.
    1. Löfgren L, Eloranta S, Krawiec K, Asterkvist A, Lönnqvist C, Sandelin K, et al. Validation of data quality in the Swedish National Register for breast cancer. BMC Public Health. 2019;19(1):495.
    1. Staaf J, Häkkinen J, Hegardt C, Saal LH, Kimbung S, Hedenfalk I, et al. RNA sequencing-based single sample predictors of molecular subtype and risk of recurrence for clinical assessment of early-stage breast cancer. NPJ Breast Cancer. 2022;8(1):94.
    1. Wu SZ, Al-Eryani G, Roden DL, Junankar S, Harvey K, Andersson A, et al. A single-cell and spatially resolved atlas of human breast cancers. Nat Genet. 2021;53(9):1334–1347.
    1. Nappi A, De Stefano MA, Dentice M, Salvatore D. Deiodinases and cancer. Endocrinology. 2021;162:4.
    1. Goemann IM, Marczyk VR, Romitti M, Wajner SM, Maia AL. Current concepts and challenges to unravel the role of iodothyronine deiodinases in human neoplasias. Endocr Relat Cancer. 2018;25(12):R625–r645.
    1. Robinson DR, Wu YM, Vats P, Su F, Lonigro RJ, Cao X, et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat Genet. 2013;45(12):1446–1451.
    1. Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3(2):224–237.
    1. Brueffer C, Vallon-Christersson J, Grabau D, Ehinger A, Häkkinen J, Hegardt C et al. Clinical value of RNA sequencing–based classifiers for prediction of the five conventional breast Cancer biomarkers: a Report from the Population-Based Multicenter Sweden Cancerome Analysis Network—Breast Initiative. JCO Precis Oncol. 2018;(2):1–18.
    1. Sotiriou C, Pusztai L. Gene-expression signatures in breast cancer. N Engl J Med. 2009;360(8):790–800.
    1. Reis-Filho JS, Pusztai L. Gene expression profiling in breast cancer: classification, prognostication, and prediction. Lancet. 2011;378(9805):1812–1823.
    1. Driscoll DM, Copeland PR, MECHANISM AND REGULATION OF SELENOPROTEIN SYNTHESIS Annu Rev Nutr. 2003;23(1):17–40.
    1. Turanov AA, Everley RA, Hybsier S, Renko K, Schomburg L, Gygi SP, et al. Regulation of selenocysteine content of human selenoprotein P by dietary selenium and insertion of cysteine in place of selenocysteine. PLoS ONE. 2015;10(10):e0140353.
    1. Xia Y, Hill KE, Byrne DW, Xu J, Burk RF. Effectiveness of selenium supplements in a low-selenium area of China2. Am J Clin Nutr. 2005;81(4):829–834.
    1. Köhrle J, Frädrich C. Deiodinases control local cellular and systemic thyroid hormone availability. Free Radic Biol Med. 2022;193:59–79.
    1. Krashin E, Silverman B, Steinberg DM, Yekutieli D, Giveon S, Fabian O, et al. Pre-diagnosis thyroid hormone dysfunction is associated with cancer mortality. Endocr Relat Cancer. 2021;28(11):705–713.
    1. Sandsveden M, Borgquist S, Rosendahl AH, Manjer J. Low thyroid hormone receptor alpha-2 (THRα-2) tumor expression is associated with unfavorable tumor characteristics and high breast cancer mortality. Breast Cancer Res. 2021;23(1):117.
    1. Journy NMY, Bernier MO, Doody MM, Alexander BH, Linet MS, Kitahara CM. Hyperthyroidism, hypothyroidism, and cause-specific mortality in a large cohort of women. Thyroid. 2017;27(8):1001–1010.
    1. Brandt J, Borgquist S, Almquist M, Manjer J. Thyroid function and survival following breast cancer. BJS (Br J Surg) 2016;103(12):1649–1657.
    1. Tosovic A, Bondeson A-G, Bondeson L, Ericsson U-B, Manjer J. Triiodothyronine levels in relation to mortality from breast cancer and all causes: a population-based prospective cohort study. Eur J Endocrinol. 2013;168(4):483–490.
    1. Pascual A, Aranda A. Thyroid hormone receptors, cell growth and differentiation. Biochim Biophys Acta. 2013;1830(7):3908–3916.
    1. Kowalik MA, Puliga E, Cabras L, Sulas P, Petrelli A, Perra A, et al. Thyroid hormone inhibits hepatocellular carcinoma progression via induction of differentiation and metabolic reprogramming. J Hepatol. 2020;72(6):1159–1169.
    1. Dentice M, Luongo C, Huang S, Ambrosio R, Elefante A, Mirebeau-Prunier D, et al. Sonic hedgehog-induced type 3 deiodinase blocks thyroid hormone action enhancing proliferation of normal and malignant keratinocytes. Proc Natl Acad Sci. 2007;104(36):14466–71.
    1. Labunskyy VM, Hatfield DL, Gladyshev VN. The Sep15 protein family: roles in disulfide bond formation and quality control in the endoplasmic reticulum. IUBMB Life. 2007;59(1):1–5.
    1. Ferguson AD, Labunskyy VM, Fomenko DE, Araç D, Chelliah Y, Amezcua CA, et al. NMR structures of the selenoproteins Sep15 and SelM reveal redox activity of a new thioredoxin-like family*. J Biol Chem. 2006;281(6):3536–3543.
    1. Davis CD, Tsuji PA, Milner JA. Selenoproteins and cancer prevention. Annu Rev Nutr. 2012;32(1):73–95.
    1. Zigrossi A, Hong LK, Ekyalongo RC, Cruz-Alvarez C, Gornick E, Diamond AM, et al. SELENOF is a new tumor suppressor in breast cancer. Oncogene. 2022;41(9):1263–1268.
    1. Lan X, Xing J, Gao H, Li S, Quan L, Jiang Y, et al. Decreased expression of Selenoproteins as a poor prognosticator of gastric cancer in humans. Biol Trace Elem Res. 2017;178(1):22–28.
    1. Dai X, Thongchot S, Dokduang H, Loilome W, Khuntikeo N, Titapun A, et al. Potential of Selenium Compounds as New Anticancer Agents for Cholangiocarcinoma. Anticancer Res. 2016;36(11):5981–5988.
    1. Schomburg L, Schweizer U. Hierarchical regulation of selenoprotein expression and sex-specific effects of selenium. Biochim Biophys Acta (BBA) Gen Subj. 2009;1790(11):1453–62.
    1. Bermano G, Nicol F, Dyer JA, Sunde RA, Beckett GJ, Arthur JR, et al. Tissue-specific regulation of selenoenzyme gene expression during selenium deficiency in rats. Biochem J. 1995;311(2):425–430.
    1. Baker JR, Umesh S, Jenab M, Schomburg L, Tjønneland A, Olsen A, et al. Prediagnostic blood selenium status and mortality among patients with colorectal cancer in western european populations. Biomedicines. 2021;9(11):1521.
    1. Bleys J, Navas-Acien A, Guallar E. Serum selenium levels and all-cause, cancer, and cardiovascular mortality among US adults. Arch Intern Med. 2008;168(4):404–410.
    1. Rogoża-Janiszewska E, Malińska K, Baszuk P, Marciniak W, Derkacz R, Lener M, et al. Serum selenium level and 10-year survival after melanoma. Biomedicines. 2021;9(8):991.
    1. Lubiński J, Marciniak W, Muszynska M, Jaworowska E, Sulikowski M, Jakubowska A, et al. Serum selenium levels and the risk of progression of laryngeal cancer. PLoS ONE. 2018;13(1):e0184873.
    1. Pietrzak S, Wójcik J, Scott RJ, Kashyap A, Grodzki T, Baszuk P, et al. Influence of the selenium level on overall survival in lung cancer. J Trace Elem Med Biol. 2019;56:46–51.
    1. Vallon-Christersson JRNA. Sequencing-based single sample predictors of molecular subtype and risk of recurrence for clinical assessment of early-stage breast cancer. 3 edn. Mendeley Data 2023.

Source: PubMed

3
Abonnieren