Use of Protein-Protein Interaction Network for Biomarker Identification in Oocyte Maturation

Abstract

Background: Oocyte maturation begins at the embryonic stage and continues throughout life. The effect of Follicle- Stimulating hormone (FSH) on gene of genes was evaluated using GEO access codes for the data set GSE38345. Materials and


Methods: The microarray data containing the gene expression information from cow oocytes show that their maturation is influenced by FSH. Data analysis was performed using GEO2R. After identifying the genes and examining the different genes expressed, two gene groups with increased and decreased expression were identified. The interaction of each of the gene groups was examined using the STRING database, based on the co-expression information. The meaningful sub networks were explored using the Clusterone software. Gene ontology was performed using the comparative GO database. The miRNA-mRNA interaction network was also studied based on the miRWalk database. Finally, meaningful networks and subnets obtained by the Cytoscape software, were designed.


Results: Comparison of oocyte gene expression data between the pre-maturation and postmaturation stages after treatment with FSH revealed 5958 genes with increased expression and 4275 genes with decreased expression. Examination of the protein interaction network among the set of increased and decreased expression genes based on string information revealed 262 genes with increased expression and 147 genes with decreased expression (high confidence (0.7) data). RPS3, NUSAP1, TBL3, and ATP5H showed increased expression and were effective in the positive regulation of rRNA processing, cell division, mitochondrial ATP synthesis coupled proton, and in oxidative phosphorylation and progesterone-mediated functions. WDR46 and MRPL22 showed decreased expression, which were important in the regulation of SRP-dependent co-translational proteins targeting the membrane, RNA secondary structure, unwinding, and functional pathways of ribosomal and RNA polymerase. The most important miRNA genes in the protein network of increased and decreased gene expression were bta-miR-10b-5p and miR-29b-2-5p.


Conclusion: Examination of the genes expressed in the oocyte maturation pathway revealed nuclear, mitochondrial, and miRNA genes. Increasing and decreasing gene expression helps maintain equilibrium, which can be a biological marker.

References
[1] Segal E, Friedman N, Kaminski N, Regev A, Koller D. From signatures to models: understanding cancer using microarrays. Nature Genet. 2005;37:S38-45.

[2] Patel OV, Bettegowda A, Ireland JJ, Coussens PM, Lonergan P, Smith GW. Functional genomics studies of oocyte competence: Evidence that reduced transcript abundance for follistatin is associated with poor developmental competence of bovine oocytes. Reprod. 2007;133:95-106.

[3] Bader GD, Cary MP, Sander C. Pathguide: a pathway resource list. Nucleic Acids Res. 2006;1(34):D504-6.

[4] Behdani E, Bakhtiarizadeh MR. Construction of an integrated gene regulatory network link to stress-related immune system in cattle. Genetica. 2017;145(4-5):441-54.

[5] Liu J, Jing L, TU X. Weighted gene co-expression network analysis identifies specific modules and hub genes related to coronary artery disease. BMC Cardiovasc Disord. 2016;16:54

[6] Uddin RK, Singh SM. Hippocampal gene expression meta-analysis identifies aging and age-associated spatial learning impairment (ASLI) genes and pathways. PLoS One. 2013;8(7):e69768.

[7] Riquelme Medina I, Lubovac-Pilav Z. Gene Co-Expression Network Analysis for Identifying Modules and Functionally Enriched Pathways in Type 1 Diabetes. PLoS One. 2016; 3(11):6.

[8] Labrecque RM, Vigneault C, Blondin P, Sirard MA. Gene expression analysis of bovine oocytes with high developmental competence obtained from FSH-stimulated animals. Mol Reprod Dev. 2013;80(6):428-40.

[9] Von Mering C, Huynen M, Jaeggi D, Schmidt S, Bork P, Snel B. STRING: a database of predicted functional associations between proteins. Nucleic Acids Res. 2003;31(1):258-61.

[10] Tamás N, Haiyuan Y, Alberto P. Detecting overlapping protein complexes in proteinprotein interaction networks. Nat Methods. 2012;9(5):471-2.

[11] Assenov Y, Ramírez F, Schelhorn SE., Lengauer T, Albrecht M. Computing topological parameters of biological networks. Bioinformatics. 2008;24:282-4.

[12] Fruzangohar M, Ebrahimie E, Ogunniyi AD, Mahdi LK, Paton JC, Adelson DL. Comparative GO: A web application for comparative gene ontology and gene ontology-based gene selection in bacteria. PLoS One. 2013;8(3):e58759.

[13] Sticht C, De La Torre C, Parveen A, Gretz N. miRWalk: An online resource for prediction of microRNA binding sites. PLoS One. 2018;13(10):e0206239.

[14] Jang CY, Kim HD, Zhang X, Chang JS, Kim J. Ribosomal protein S3 localizes on the mitotic spindle and functions as a microtubule associated protein in mitosis. Biochem Biophys Res Commun. 2012;429(1-2):57-62.

[15] Susor A, Kubelka M. Translational regulation in the mammalian oocyte. Results and probl in cell differ. 2017;63:257-95.

[16] Mayer S, Wrenzycki C, Tomek W. Inactivation of mTor arrests bovine oocytes in the metaphase-I stage, despite reversible inhibition of 4E-BP1 phosphorylation. Mol Reprod Dev. 2014;81(4):363-75.

[17] Orozco-Lucero E, Sirard MA. Molecular markers of fertility in cattle oocytes and embryos: progress and challenges. Anim. Reprod. 2014;11(3):183-94.

[18] Torner H, Ghanem N, Ambros C, Hölker M, Tomek W, Phatsara C, et al. Molecular and subcellular characterisation of oocytes screened for their developmental competence based on glucose-6-phosphate dehydrogenase activity. Reprod. 2008;135(2):197-212.

[19] Raemaekers T, Ribbeck K, Beaudouin J, Annaert W, Van Camp M, Stockmans I, et al. NuSAP, a novel microtubule-associated protein involved in mitotic spindle organization. J Cell Biol. 2003;162(6):1017-29.

[20] Ribbeck K, Aaron C, Groen AC, Santarella R, Bohnsack TB, Raemaekers T, et al. NuSAP, a mitotic RanGTP target that stabilizes and cross-links microtubules. MBoC. 2006;17(6):2646-60.

[21] Yu J, Lan X, Chen X, Yu C, Xu Y, Liu Y, et al. Protein synthesis and degradation are essential to regulate germ line stem cell homeostasis in Drosophila testes. Development. 2016;143(16):2930-45.

[22] Peddinti D, Memili E, Burgess SC. Proteomics-Based Systems Biology Modeling of Bovine Germinal Vesicle Stage Oocyte and Cumulus Cell Interaction. PLoS One. 2010;5(6):e11240.

[23] Tanghe S, Van Soom A, Nauwynck H, Coryn M, de Kruif A. Minireview: Functions of the cumulus oophorus during oocyte maturation, ovulation, and fertilization. Mol Reprod Dev. 2002;61(3):414-24.

[24] Buccione R, Schroeder AC, Eppig JJ. Interactions between somatic cells and germ cells throughout. mammalian oogenesis. Biol Reprod. 1990;43(4):543-7.

[25] Tripurani SK, Xiao C, Salem M, Yao J. Cloning and analysis of fetal ovary microRNAs in cattle. Anim Reprod Sci. 2010;120(1-4):16-22.

[26] Fleckenstein B, Daniel MD, Hunt RD, Werner RD, Falk LA, Mulder C. Tumour induction with DNA of oncogenic primate herpes viruses. Nature. 1978;274:57-9.

[27] Irvine KR, Rao JB, Rosenberg SA, Restifo NP. Cytokine enhancement of DNA immunization leads to effective treatment of established pulmonary metastases. J Immunol. 1996;156(1):238-45.

[28] Israel MA, Chan HW, Hourihan SL, Rowe WP, Martin MA. Biological activity of polyoma virus DNA in mice and hamsters. J Virol. 1979;29(3):990-6.

[29] Ahmad N, Townsend E, Dailey R, Inskeep E. Relationships of hormonal patterns and fertility to occurrence of two or three waves of ovarian follicles, before and after breeding, in beef cows and heifers. Anim Reprod Sci. 1997;49(1):13-28.

[30] Lubzens E, Young G, Bobe J, Cerdà J. Oogenesis in teleosts: How fish eggs are formed. Gen. Comp. Endocrinol. 2010;165(3):367-89.

[31] Wegnez M, Monier R, Denis H. Sequence heterogeneity of 5S RNA in Xenopus laevis. FEBS Lett. 1972;25(1):13-20.

[32] Tsafriri A, Channing CP. Influence of follicular maturation and culture conditions on the meiosis of pig oocytes in vitro. J Reprod Fertil. 1975;43(1):149-52.

[33] Motlik J, Crozet N, Fulka J. Meiotic competence in vitro of pig oocytes isolated from early antral follicles. J Reprod Fertil. 1984;72(2):323-8.

[34] Nicole C, Kanka J, Motlik J, Fulka, J. Nucleolar fine structure and RNA synthesis in bovine oocytes from antral follicles. Gamete Res. 1986;14(1):65-73.

[35] Motlík J, Kopecný V, Trávnik P, Pivko J. RNA synthesis in pig follicular oocytes. Auto radiographic and cytochemical study. Biol Cell. 1984;50(3):229-35.

[36] Leary DJ, Huang S. Regulation of ribosome biogenesis within the nucleolus. FEBS Lett. 2001;509(2):145-50.

[37] Wachtler F, Stahl A. The nucleolus: a structural and functionallinterpretation. Micron. 1993;24:473-505.

[38] Grummt I. Regulation of Mammalian Ribosomal Gene Transcription by RNA Polymerase I. Prog Nucleic Acid Res Mol Biol. 1998;62:109-54.

[39] Bell SP, Learned RM, Jantzen HM, Tjian R. Functional cooperativity between transcription factors UBF1 and SL1 mediates human ribosomal RNA synthesis. Science. 1988;241(4870):1192-7.

[40] Comai L, Tanese N, Tjian R. The TATA-binding protein and associated factors are integral components of the RNA polymerase I transcription factor, SL1. Cell. 1992;68(5):965-76.

[41] Learned RM, Learned TK, Haltiner MM, Tjian RT. Human rRNA transcription is modulated by the coordinate binding of two factors to an upstream control element. Cell. 1986;45(6):847-57.

[42] Tuan JC, Zhai W, Comai L. Recruitment of TATA-binding protein-TAFI complex SL1 to the human ribosomal DNA promoter is mediated by the carboxy-terminal activation domain of upstream binding factor (UBF) and is regulated by UBF phosphorylation. Mol Cell Biol. 1999;19(4):2872-9.

[43] Leese HJ, Barton AM. Pyruvate and glucose uptake by mouse ova and preimplantation embryos. J Reprod Fertil. 1984;72(1):9-13.

[44] Johnson MT, Freeman EA, Gardner DK, Hunt PA. Oxidative metabolism of pyruvateis required for meiotic maturation of murine oocytes in vivo. Biol Reprod. 2007;77(1):2-8.

[45] Tarlatzis BC, Pazaitou K, Bili H, Bontis J, Papadimas J, Lagos S, et al. Growth hormone, oestradiol, progesterone and testosterone concentrations in follicular fluid after ovarian stimulation with various regimes for assisted reproduction. Hum Reprod. 1993;8(10):1612-6.

[46] Leroy JL, Vanholder T, Delanghe JR, Opsomer G, Van Soom A, Bols PE, et al. Metabolic changes in follicular fluid of the dominant follicle in high-yielding dairy cows early post partum. Theriogenology. 2004;62(6):1131-43.

[47] Barzaghi Firooz Abadi F. Laboratory effects of ascorbic acid and FSH on oocyte maturation and mouse follicles. JSSU. 2011;19(5):586-97.

[48] Lydon JP, Demayo FJ, Funk CR, Mani SK, Hughes AR, Montgomery CA, et al. Mice lacking progesterone receptor exhibit pleiotropic reproductive abnormalities. Genes Dev. 1995;9(18):2266-78.

[49] Kim J, Bagchi IC, Bagchi MK. Control of ovulation in mice by progesterone receptorregulated gene networks. Mol Hum Reprod. 2009;15(12):821-8.

[50] Akison LK, Robker RL. The critical roles of progesterone receptor (PGR) in ovulation, oocyte developmental competence and oviductal transport in mammalian reproduction. Reprod Domest Anim. 2012; 47(4):288-96.

[51] Shimada M, Yamashita Y, Ito J, Okazaki T, Kawahata K, Nishibori M. Expression of two progesterone receptor isoforms in cumulus cells and their roles during meiotic resumption of porcine oocytes. J Mol Endocrinol. 2004;33:209-25.

[52] Hunter RHF. Sperm release from oviduct epithelial binding is controlled hormonally by peri-ovulatory Graafian follicles. Mol Reprod Dev. 2008;75(1):167-74.

[53] Bureau M, Bailey JL, Sirard MA. Binding regulation of porcine spermatozoa to oviductal vesicles in vitro. J Androl. 2002;23(2):188-93.

[54] Yamashita Y, Shimada M, Okazaki T, Maeda T, Terada T. Production of progesterone from de novo-synthesized cholesterol in cumulus cells and its physiological role during meiotic resumption of porcine oocytes. Biol Reprod. 2003;68(4):1193-8.

[55] Hirai Y, Louvet E, Oda T, Kumeta M, Watanabe Y, Horigome T, et al. Nucleolar scaffold protein, WDR46, determines the granular compartmental localization of nucleolin and DDX21. Genes Cells. 2013;18(9):780-97.

[56] Lu S, Arthos J, Montefiori DC, Yasutomi Y, Manson K, Mustafa F, et al. Simian immunodeficiency virus DNA vaccine trial in macaques. J Virol. 1996;70(6):3978-91.

[57] Alexeyev MF, Venediktova N, Pastukh V, Shokolenko I, Bonilla G, Wilson GL. Selective elimination of mutant mitochondrial genomes as therapeutic strategy for the treatment of NARP and MILS syndromes. Gene Ther. 2008;15(7):516-23.

[58] Ma W, Zhang D, Hou Y, Li YH, Sun QY, Sun XF, et al. Reduced expression of MAD2, BCL2, and MAP kinase activity in pig oocytes after in vitro aging are associated with defects in sister chromatid segregation during meiosis II and embryo fragmentation after activation. Biol Reprod. 2005;72(2):373-83.

[59] Gordo AC, Rodrigues P, Kurokawa M, Jellerette T, Exley GE, Warner C, et al. Intracellular calcium oscillations signal apoptosis rather than activation in in vitro aged mouse eggs. Biol Reprod. 2002;66(6):1828-37

[60] Wang Z. MicroRNA: A matter of life or death. World J Biol Chem. 2010;1(4):41-54.

[61] Kim MR, Tilly JL. Current concepts in Bcl-2 family member regulation of female germ cell development and survival. Mol Cell Research. 2003;1644(2-3):205-10.

[62] Lord T, Aitken RJ. Oxidative stress and ageing of the post-ovulatory oocyte. Reprod. 2013;146(6):217-27.

[63] Sommerville J, Brumwell CL, Ritland Politz JC, Pederson T. Signal recognition particle assembly in relation to the function of amplified nucleoli of Xenopus oocytes. J Cell Sci. 2005;118(6):1299-307.

[64] Akopian D, Kuang S, Xin Z, Shu-ou S. Signal recognition particle: An essential protein targeting machine. Annu Rev Biochem. 2013;82:693-721.

[65] Jaramillo M, Dever TE, Merrick WC, Sonenberg N. RNA unwinding in translation: assembly of helicase complex intermediates comprising eukaryotic initiation factors eIF-4F and eIF-4B. Mol Cell Biol. 1991;11(12):5992-7.

[66] Palma GA, Arganaraz ME, Barrera AD, Rodler D, Mutto AA, Sinowatz F. Biology and biotechnology of follicle development. Sci World J. 2012;1-14.

[67] Yenuganti VR, Baddela VS, Baufeld A, Singh D, Vanselow J. The gene expression pattern induced by high plating density in cultured bovine and buffalo granulose cells might be regulated by specific miRNA species. Sci World J. 2015;61(2):154-60.

[68] Bashirullah A, Cooperstock RL, Lipshitz HD. Spatial and temporal control of RNA stability. Proc Natl Acad Sci USA. 2001;98(13):7025-8.

[69] Audic Y, Garbrecht M, Fritz B, Sheetts MD. Zygotic control of maternal Cyclin A1 translation and mRNA stability. Dev Dyn. 2002;225(4):511-21.

[70] Schultz RM. Regulation of Zygotic gene activation in mouse. Bioessay. 1993;15(8):531- 8.

[71] Gebauer F, Xu W, Cooper GM, Richter JD. Translational control by cytoplasmic polyadenylation of c-mos mRNA is necessary for oocyte maturation in the mouse. EMBO J. 1994;13(23):5712-20.

[72] Wahle E. A Novel poly(A) binding protein acts as a specific factor in the second phase of messenger RNA polyadenylation. Cell. 1991;66(4):759-68.

[73] Hershey WB. Translational control in mammalian cells. Annu Rev Biochem. 1991;60:717-55.

[74] Pause A, Belsham GJ, Gingras AC, Donzé O, Lin TA, Lawrence JC Jr, et al. Insulindependent stimulation of protein synthesis by phosphorylation of a regulator of 59- cap function. Nature. 1994;371:162-7.

[75] Richter J, Lasko P. Translational control in oocyte development. Cold Spring Harb Perspect Biol. 2011;3(9):a002758.

[76] Blondin P, Bousquet D, Twagiramungu H, Barnes F, Sirard MA. Manipulation of follicular development to produce developmentally competent bovine oocytes. Biol Reprod. 2002;66:38-43.

[77] Nivet al, Bunel A, Labrecque R, Belanger J, Vigneault C, Blondin P, et al. FSH withdrawal improves developmental competence of oocytes in the bovine model. Reprod. 2012;143:165-71.

[78] Gilbert I, Scantland S, Sylvestre EL, Gravel C, Laflamme I, Sirard, MA, et al. The dynamics of gene products fluctuation during bovine pre-hatching development. Mol Reprod Dev. 2009;76:762-72.

[79] Coenen K, Massicotte L, Sirard MA. Study of newly synthesized proteins during bovine oocyte maturation in vitro using image analysis of two-dimensional gel electrophoresis. Mol Reprod Dev. 2004;67:313-22.

[80] Fair T, Hyttel P, Greve T. Bovine oocyte diameter in relation to maturational competence and transcriptional activity. Mol Reprod Dev. 1995;42:437-42.

[81] LoddeV, Modina S, Maddox-Hyttel P, Franciosi F, Lauria A, Luciano AM. Oocyte morphology and transcriptional silencing in relation to chromatin remodeling during the final phases of bovine oocyte growth. Mol Reprod Dev. 2008;75:915-24.

[82] Richter JD. Cytoplasmic poly adenylation in development and beyond. Microbiol Mol Biol Rev. 1999;63:446-56.

[83] Ghanem N, Holker M, Rings F, Jennen D, Tholen E, Sirard MA, et al. Alterations in transcript abundance of bovine oocytes recovered at growth and dominance phases of the first follicular wave. BMC Develop Biol. 2007;7:90.

[84] Schatten H, Sun QY. Centrosome and microtubule functions and dysfunctions in meiosis: Implications for age-related infertility and developmental disorders. Reprod Fertil Dev. 2015;27:934-43.

[85] Bennabi I, Terret ME, Verlhac MH. Meiotic spindle assembly and chromosome segregation in oocytes. J Cell Biol. 2016;215, 611–619.

[86] Wilding M, Dale B, Marino M, di Matteo L, Alviggi C, Pisaturo ML, et al. Mitochondrial aggregation patterns and activity in human oocytes and preimplantation embryos. Hum Reprod. 2001;16(5):909-17.

[87] Curtis D, Lehmann R, Zamore PD. Translational regulation in development. Cell. 1995;81:171-8.

[88] Zeng F, Schultz RM. Requirement for RNAbinding activity of MYS2 for cytoplasmic localization and retention in mouse oocytes. Dev Biol. 2003;255:249-62.

[89] Ferreira EM, Vireque A, Adona PR, Meirelles FV, Ferriani RA, Navarro PA. Cytoplasmic maturation of bovine oocytes: structural and biochemical modifications and acquisition of developmental competence. Theriogenology. 2009;71(5):836-48.

[90] Robin ED, Wong R. Mitochondrial DNA molecules and virtual number of mitochondria per cell in mammalian cells. J Cell Physiol. 1988;136:507-13.

[91] Shuster RC, Rubenstein AJ, WallaceBiochem DC. Mitochondrial DNA in anucleate human blood cells. Biophys Res Commun. 1988;155:1360-5.

[92] Daniel MC, Kirchhoff F, Czajak SC, Sehgal PK, Desrosiers RC. Protective effects of a live attenuated SIV vaccine with a deletion in the nef gene. Science. 1992;258:1938-41.

[93] Gosden RG. Oogenesis as a foundation for embryogenesis. Mol Cell Endocrinol. 2002;186(2):149-53

[94] Jansen RP, de Boer K. The bottleneck: mitochondrial imperatives in oogenesis and ovarian follicular fate. Mol Cell Endocrinol. 1998;145:81-8.

[95] Chen X, Prosser R, Simonetti S, Sadlock J, Jagiello G, Schon EA. Rearranged mitochondrial genomes are present in human oocytes. Am J Hum Genet. 1995;57:239-47.

[96] Van Blerkom J. Mitochondria in human oogenesis and preimplantation embryogenesis: engines of metabolism, ionic regulation and developmental competence. Reprod. 2004;128 (3):269-80.

[97] Khanaki K, Abedinzade M, Gazor R, Norasfard M, Jafari-Shakib R. Effect of Lamium album on mitochondrial oxidative stress in diabetic rats. Res Mol Med. 2017;5(2):9-13.

[98] Susin, SA, Zamzami N, Castedo M, Hirsch T, Marchetti P, MachoV, et al. BCL-2 inhibits the mitochondrial release of an apoptogenic protease. J Exp Med. 1996;184:1331-41.

[99] Suen DF, Norris KL, Youle RJ. Mitochondrial dynamics and apoptosis. Genes Dev. 2008;22:1577-90.

[100] Chandel NS. Mitochondria and cancer. Cancer Metab. 2014;2:8.

[101] Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, et al. Dicer is essential for mouse development. Nat Genet. 2003;35(3):215-7.

[102] Assou S, Al-edani T, Haouzi D, Philippe N, Lecellier CH, Piquemal D, et al. MicroRNAs: New candidates for the regulation of the human cumulus oocyte complex. Hum Reprod. 2013;28(11):3038-49.

[103] Tong XH, Xu B, Zhang YW, Liu YS, Ma CH. Research resources: Comparative microRNA profiles in human corona radiata cells and cumulus oophorus cells detected by next-generation small RNA sequencing. PLoS One. 2014;9(9):e106706.

[104] Tanghe S, Van Soom A, Nauwynck H, Coryn M, de Kruif A. Minireview: Functions of the cumulus oophorus during oocyte maturation, ovulation, and fertilization. Mol Reprod Dev. 2002;61(3):414-24.

[105] Shahedi A, Hosseini A, Khalili MA, Yeganeh F. Effects of vitrification on nuclear maturation and gene expression of immature human oocytes. Res Mol Med. 2017;5(1):27-33.