Structural analysis of human SEPHS2 protein, a selenocysteine machinery component, over-expressed in triple negative breast cancer

Carmine Nunziata, Andrea Polo, Angela Sorice, Francesca Capone, Marina Accardo, Eliana Guerriero, Federica Zito Marino, Michele Orditura, Alfredo Budillon, Susan Costantini, Carmine Nunziata, Andrea Polo, Angela Sorice, Francesca Capone, Marina Accardo, Eliana Guerriero, Federica Zito Marino, Michele Orditura, Alfredo Budillon, Susan Costantini

Abstract

Selenophosphate synthetase 2 (SEPHS2) synthesizes selenide and ATP into selenophosphate, the selenium donor for selenocysteine (Sec), which is cotranslationally incorporated into selenoproteins. The action and regulatory mechanisms of SEPHS2 as well as its role in carcinogenesis (especially breast cancer) remain ambiguous and need further clarification. Therefore, lacking an experimentally determined structure for SEPHS2, we first analyzed the physicochemical properties of its sequence, modeled its three-dimensional structure and studied its conformational behavior to identify the key residues (named HUB nodes) responsible for protein stability and to clarify the molecular mechanisms by which it induced its function. Bioinformatics analysis evidenced higher amplification frequencies of SEPHS2 in breast cancer than in other cancer types. Therefore, because triple negative breast cancer (TNBC) is biologically the most aggressive breast cancer subtype and its treatment represents a challenge due to the absence of well-defined molecular targets, we evaluated SEPHS2 expression in two TNBC cell lines and patient samples. We demonstrated mRNA and protein overexpression to be correlated with aggressiveness and malignant tumor grade, suggesting that this protein could potentially be considered a prognostic marker and/or therapeutic target for TNBC.

Conflict of interest statement

The authors declare no competing interests.

Figures

Figure 1
Figure 1
Sequence analysis of human SEPHS2. The negatively and positively charged residues are shown in red and cyan, respectively. We report predictions for secondary structure (Jnet), disorder propensity (Meta_d and IUPred), globularity propensity (GlobPL), predicted and experimentally determined phosphorylation sites (NetPhos and Phosphosite), sulfination sites (Sulfinator), glycosylation sites (NetNGlyc and NetOGlyc), and molecular recognition features (MorfPred and ANCHOR).
Figure 2
Figure 2
BLAST alignment between human SEPHS2 and human SEPHS1. The 310-helices, α-helices and β-strands are shown in cyan, red and yellow, respectively.
Figure 3
Figure 3
Complete SEPHS2 model obtained by the molecular modeling approach. In detail, 310-helices and α-helices are reported in red, β-strands in yellow and loops in green.
Figure 4
Figure 4
(A) RMSD, (B) gyration radius, (C) H-bond, (D) RMSF, and (E) solvent accessible surface plots and (F) partial densities for SEPHS2 during MD simulations at neutral (in red) and acidic (in black) pH.
Figure 5
Figure 5
Projection plot and cluster analysis results for SEPHS2 during MD simulations at neutral (A,B) and acidic pH (C,D), respectively.
Figure 6
Figure 6
Residue interaction network for SEPHS2 at the end of the neutral pH simulation. HUB nodes are in pink, and the other residues are in light blue.
Figure 7
Figure 7
Residue interaction network for SEPHS2 at the end of the acidic pH simulation. HUB nodes are in pink, and the other residues are in light blue.
Figure 8
Figure 8
SEPHS2 levels in MDA-MB468 and MDA-MB231 cells compared to MCF10A cells and in TNBC tissues compared to their normal counterparts based on RT-qPCR analysis. The p-values

Figure 9

( A ) Immunohistochemical observation…

Figure 9

( A ) Immunohistochemical observation of human SEPHS2 expression at 200x magnification in…

Figure 9
(A) Immunohistochemical observation of human SEPHS2 expression at 200x magnification in normal mammary tissues and grade 2 (G2) and 3 (G3) TNBC patients. In the panel related to a tissue section from a G2 patient, only the neoplastic lesion is visible. In the panel related to a tissue section from a G3 patient, two black arrows indicate a lobular ductule of a normal mammary gland immersed in the adipose stroma; moreover, clusters of neoplastic cells are found in the same stroma. (B) Kaplan–Meier curves of the overall survival of TNBC patients. Blue line: score = 2 and green line: score = 3.
All figures (9)
Figure 9
Figure 9
(A) Immunohistochemical observation of human SEPHS2 expression at 200x magnification in normal mammary tissues and grade 2 (G2) and 3 (G3) TNBC patients. In the panel related to a tissue section from a G2 patient, only the neoplastic lesion is visible. In the panel related to a tissue section from a G3 patient, two black arrows indicate a lobular ductule of a normal mammary gland immersed in the adipose stroma; moreover, clusters of neoplastic cells are found in the same stroma. (B) Kaplan–Meier curves of the overall survival of TNBC patients. Blue line: score = 2 and green line: score = 3.

References

    1. Turanov AA, et al. Biosynthesis of selenocysteine, the 21st amino acid in the genetic code, and a novel pathway for cysteine biosynthesis. Adv. Nutr. 2011;2:122–8. doi: 10.3945/an.110.000265.
    1. Xu XM, et al. Selenophosphatesynthetase 2 is essential for selenoprotein biosynthesis. Biochem. J. 2007;404:115–20. doi: 10.1042/BJ20070165.
    1. Xu XM, et al. Biosynthesis of selenocysteine on its tRNA in eukaryotes. PLoS Biol. 2007;5:e4. doi: 10.1371/journal.pbio.0050004.
    1. Palioura S, Sherrer RL, Steitz TA, Söll D, Simonovic M. The human SepSecS-tRNASec complex reveals the mechanism of selenocysteine formation. Science. 2009;325:321–325. doi: 10.1126/science.1173755.
    1. Oudouhou F, Casu B, Dopgwa Puemi AS, Sygusch J, Baron C. Analysis of Novel Interactions between Components of the Selenocysteine Biosynthesis Pathway, SEPHS1, SEPHS2, SEPSECS, and SECp43. Biochemistry. 2017;56:2261–2270. doi: 10.1021/acs.biochem.6b01116.
    1. Davis CD, Tsuji PA, Milner JA. Selenoproteins and cancer prevention. Annu. Rev. Nutr. 2012;32:73–95. doi: 10.1146/annurev-nutr-071811-150740.
    1. Lan X, et al. Decreased Expression of Selenoproteins as a Poor Prognosticator of Gastric Cancer in Humans. Biol. Trace Elem. Res. 2017;178:22–28. doi: 10.1007/s12011-016-0908-8.
    1. Maciel-Dominguez A, Swan D, Ford D, Hesketh J. Selenium alters miRNA profile in an intestinal cell line: evidence that miR-185 regulates expression of GPX2 and SEPSH2. Mol. Nutr. Food Res. 2013;57:2195–205. doi: 10.1002/mnfr.201300168.
    1. Dudaladava V, et al. Gene Expression Profiling in Hereditary, BRCA1-linked Breast Cancer: Preliminary Report. Hered. Cancer Clin. Pract. 2006;4:28–38. doi: 10.1186/1897-4287-4-1-28.
    1. Braicu C, et al. Novel insight into triple-negative breast cancers, the emerging role of angiogenesis, and antiangiogenic therapy. Expert Rev. Mol. Med. 2016;18:e18. doi: 10.1017/erm.2016.17.
    1. Xu F, et al. Repositioning antipsychotic fluphenazine hydrochloride for treating triple negative breast cancer with brain metastases and lung metastases. Am. J. Cancer Res. 2019;9(3):459–478.
    1. Gasteiger, E. et al. Protein identification and Analysis tools on the ExPASy Server; in John M. Walker (ed.): The Proteomics Protocols Handbook. Humana Press. 571–607 (2005).
    1. Das RK, Pappu RV. Conformations of intrinsically disordered proteins are influenced by linear sequence distributions of oppositely charged residues. Proc. Natl. Acad. Sci. USA. 2013;110:13392–7. doi: 10.1073/pnas.1304749110.
    1. Cuff JA, Barton GJ. Application of multiple sequence alignment profiles to improve protein secondary structure prediction. Proteins. 2000;40:502–511. doi: 10.1002/1097-0134(20000815)40:3<502::AID-PROT170>;2-Q.
    1. Kozlowski LP, Bujnicki JM. MetaDisorder: a meta-server for the prediction of intrinsic disorder proteins. BMC Bioinformatics. 2012;13:111. doi: 10.1186/1471-2105-13-111.
    1. Wright PE, Dyson HJ. Intrinsically disordered proteins in cellular signalling and regulation. Nat. Rev. Mol. Cell. Biol. 2015;16:18–29. doi: 10.1038/nrm3920.
    1. Blom N, Gammeltoft S, Brunak S. Sequence and structure-based prediction of eukaryotic protein phosphorylation sites. J. Mol. Biol. 1999;294:1351–62. doi: 10.1006/jmbi.1999.3310.
    1. Hornbeck PV, et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 2014;43:D512–20. doi: 10.1093/nar/gku1267.
    1. Monigatti F, Gasteiger E, Bairoch A, Jung E. The Sulfinator: Predicting tyrosine sulfination sites in protein sequences. Bioinformatics. 2002;18:769–70. doi: 10.1093/bioinformatics/18.5.769.
    1. Steentoft C, et al. Precision mapping of the human O-GalNAcglycoproteome trough SimpleCell technology. EMBO J. 2013;32:1478–88. doi: 10.1038/emboj.2013.79.
    1. Meireles LM, Dömling AS, Camacho CJ. ANCHOR: a web server and database for analysis of protein-protein interaction binding pockets for drug discovery. Nucleic Acids Res. 2010;38:W407–11. doi: 10.1093/nar/gkq502.
    1. Disfani FM, et al. MoRFpred, a computational tool for sequenced-based prediction and characterization of short disorder-to-order transitioning binding regions in proteins. Bioinformatics. 2012;28:i75–83. doi: 10.1093/bioinformatics/bts209.
    1. Polo A, Colonna G, Guariniello S, Ciliberto G, Costantini S. Deducing functional characteristics of the human selenoprotein SELK from the structural properties of its intrinsically disordered C-terminal domain. Mol. BioSyst. 2016;12:758–72. doi: 10.1039/C5MB00679A.
    1. Polo A, Guariniello S, Colonna G, Ciliberto G, Costantini S. A study on the structural features of SELK, an over-expressed protein in hepatocellular carcinoma, by molecular dynamics simulations in a lipid-water system. Mol. Biosyst. 2016;10:3209–22. doi: 10.1039/C6MB00469E.
    1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ. Best local alignment search tool. J. Mol. Biol. 1990;215:403–410. doi: 10.1016/S0022-2836(05)80360-2.
    1. Wang KT, Wang J, Li LF, Su XD. Crystal structures of catalytic intermediates of human selenophosphatesynthetase 1. J. Mol. Biol. 2009;390:747–59. doi: 10.1016/j.jmb.2009.05.032.
    1. Sali A, Blundell TL. Comparative protein modelling by satisfaction of spatial restraints. J. Mol. Biol. 1993;234:779–815. doi: 10.1006/jmbi.1993.1626.
    1. Gopalakrishnan K, Sowmiya G, Sheik SS, Sekar K. Ramachandran plot on the web (2.0) Protein Pept. Lett. 2007;14:669–71. doi: 10.2174/092986607781483912.
    1. Sippl MJ. Recognition of errors in three-dimensional structure of proteins. Proteins. 1993;17:355–62. doi: 10.1002/prot.340170404.
    1. Wu S, Zhang Y. MUSTER: Improving protein sequence profile-profile alignments by using multiple sources of structure information. Proteins. 2008;72:547–556. doi: 10.1002/prot.21945.
    1. Van Der Spoel D, et al. GROMACS: fast, flexible, and free. J. Comput. Chem. 2005;26:1701–1718. doi: 10.1002/jcc.20291.
    1. Kato Y, et al. Acidic extracellular microenvironment and cancer. Cancer Cell Int. 2013;13:89. doi: 10.1186/1475-2867-13-89.
    1. Tina KG, Bhadra R, Srinivasan N. PIC: Protein Interactions Calculator. Nucleic Acids Res. 2007;35:W473–W476. doi: 10.1093/nar/gkm423.
    1. McDonald IK, Thornton JM. Satisfying hydrogen bonding potential in proteins. J. Mol. Biol. 1994;283:777–93. doi: 10.1006/jmbi.1994.1334.
    1. Vangone A, Spinelli R, Scarano V, Cavallo L, Oliva R. COCOMAPS: a web application to analyze and visualize contacts at the interface of biomolecular complexes. Bioinformatics. 2011;27:2915–2916. doi: 10.1093/bioinformatics/btr484.
    1. Assenov Y, Ramírez F, Schelhorn SE, Lengauer T, Albrecht M. Computing topological parameters of biological networks. Bioinformatics. 2008;24:82–284. doi: 10.1093/bioinformatics/btm554.
    1. Doncheva NT, Klein K, Dominques FS, Albrecht M. Analyzing and visualizing residue networks of protein structures. Trends Biochem. Sci. 2011;36:179–82. doi: 10.1016/j.tibs.2011.01.002.
    1. Gao J, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013;6:pl1. doi: 10.1126/scisignal.2004088.
    1. Cerami E, et al. The cBio Cancer Genomics Portal: An Open Platform for Exploring Multidimensional Cancer Genomics Data. Cancer Discov. 2012;2:401–404. doi: 10.1158/-12-0095.
    1. Holliday DL, Speirs V. Choosing the right cell line for breast cancer research. Breast Cancer Res. 2011;13:215. doi: 10.1186/bcr2889.
    1. Raymond F, et al. Selenoprotein P- Expression, Functions, and Roles in Mammals. Biochim Biophys Acta. 2009;1790:1441–1447. doi: 10.1016/j.bbagen.2009.03.026.
    1. Chen G, et al. The serum activity of thioredoxin reductases 1 (TrxR1) is correlated with the poor prognosis in EGFR wild-type and ALK negative non-small cell lung cancer. Oncotarget. 2017;8:115270–115279.
    1. Arnogiannaki N, et al. BCL-2, BAX and P53 expression profiles in endometrial carcinoma as studied by real-time PCR and immunohistochemistry. Anticancer Res. 2009;29:3977–3982.
    1. Sinicrope, F. A. et al. Bcl-2 and p53 oncoprotein expression during colorectal tumorigenesis. Cancer Res. 55, 237-241 (1995).

Source: PubMed

3
订阅