Transcriptomic Analysis and Meta-Analysis of Human Granulosa and Cumulus Cells

Tanja Burnik Papler, Eda Vrtacnik Bokal, Ales Maver, Andreja Natasa Kopitar, Luca Lovrečić, Tanja Burnik Papler, Eda Vrtacnik Bokal, Ales Maver, Andreja Natasa Kopitar, Luca Lovrečić

Abstract

Specific gene expression in oocytes and its surrounding cumulus (CC) and granulosa (GC) cells is needed for successful folliculogenesis and oocyte maturation. The aim of the present study was to compare genome-wide gene expression and biological functions of human GC and CC. Individual GC and CC were derived from 37 women undergoing IVF procedures. Gene expression analysis was performed using microarrays, followed by a meta-analysis. Results were validated using quantitative real-time PCR. There were 6029 differentially expressed genes (q < 10-4); of which 650 genes had a log2 FC ≥ 2. After the meta-analysis there were 3156 genes differentially expressed. Among these there were genes that have previously not been reported in human somatic follicular cells, like prokineticin 2 (PROK2), higher expressed in GC, and pregnancy up-regulated nonubiquitous CaM kinase (PNCK), higher expressed in CC. Pathways like inflammatory response and angiogenesis were enriched in GC, whereas in CC, cell differentiation and multicellular organismal development were among enriched pathways. In conclusion, transcriptomes of GC and CC as well as biological functions, are distinctive for each cell subpopulation. By describing novel genes like PROK2 and PNCK, expressed in GC and CC, we upgraded the existing data on human follicular biology.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Principal component analysis between granulosa…
Fig 1. Principal component analysis between granulosa and cumulus cells.
The axes of the plot represent first two main components of variance, obtained by performing principal component analysis (PCA) on global expression profiles. GC- granulosa cells; CC- cumulus cells.
Fig 2. Top network enriched in granulosa…
Fig 2. Top network enriched in granulosa cells (Enrichment score: 32) generated with Ingenuity Pathway Analysis (IPA).
The network is associated with hematological system development and function, cancer and cellular movement. Up-regulated genes in our study are marked in green colour; the colour intensity of the nodes indicates the degree of up-regulation. Transcripts in grey were not differentially expressed. Genes are represented as nodes, and the biological relationship between two nodes is represented as a line: the plain line indicates direct interaction, the dashed line indicates indirect interaction; the line without arrowhead indicates binding only, the line finishing with a vertical line indicates inhibition; the line with an arrowhead indicates ‘acts on’.
Fig 3. Top network enriched in cumulus…
Fig 3. Top network enriched in cumulus cells (Enrichment score: 41) generated with Ingenuity Pathway Analysis (IPA).
The network is associated with cellular development, skeletal and muscular system development and function and tissue development. Up-regulated genes in our study are marked in red colour; the colour intensity of the nodes indicates the degree of up-regulation. Transcripts in grey were not differentially expressed. Genes are represented as nodes, and the biological relationship between two nodes is represented as a line: the plain line indicates direct interaction, the dashed line indicates indirect interaction; the line without arrowhead indicates binding only, the line finishing with a vertical line indicates inhibition; the line with an arrowhead indicates ‘acts on’.
Fig 4. Validation of microarray results by…
Fig 4. Validation of microarray results by qPCR in additional 19 GC and 19 CC samples, derived from sixteen women.
For all genes, except ACE2, there was a statistically significant difference in expression between CC and GC. Data are presented as mean log2 fold change (FC) between expression in CC and GC. Blue bars represent log2 FC as measured by microarray; red bars represent log2 FC as measured by qPCR.
Fig 5. qPCR validation of microarray data.
Fig 5. qPCR validation of microarray data.
The results represent mean relative mRNA expression of the selected genes ± SEM.

References

    1. Moley KH, Schreiber JR. Ovarian follicular growth, ovulation and atresia. Endocrine, paracrine and autocrine regulation. Adv Exp Med Biol. 1995;77: 103–119.
    1. Greenwald GS, Terranova PF. Follicular selection and its control In: Knobil E, Neill JD, eds. The Physiology of Reproduction. New York: Raven Press; 1988; 387–445.
    1. Kidder GM, Vanderhyden BC. Bidirectional communication between oocytes and follicle cells: ensuring oocyte developmental competence. Can J Physiol Pharmacol. 2010;88: 399–413. 10.1139/y10-009
    1. Knight PG, Glister C. Focus on TGF-b Signalling: TGF-b superfamily members and ovarian follicle development. Reproduction. 2006; 132: 191–206.
    1. Gilchrist RB, Lane M, Thompson JG. Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Hum Reprod Update. 2008;14: 159–177. 10.1093/humupd/dmm040
    1. Li Q, McKenzie LJ, Matzuk MM. Revisiting oocyte-somatic cell interactions: in search of novel intrafollicular predictors and regulators of oocyte developmental competence. Mol Hum Reprod. 2008;14: 673–678. 10.1093/molehr/gan064
    1. Binelli M, Murphy BD. Coordinated regulation of follicle development by germ and somatic cells. Rep Fertil Dev. 2010;22: 1–12.
    1. Thomas FH, Ethier JF, Shimasaki S, Vanderhyden BC. Follicle-stimulating hormone regulates oocyte growth by modulation of expression of oocyte and granulosa cell factors. Endocrinology. 2005;146: 941–949.
    1. Wang JM, Chao JR, Chen W, Kuo ML, Yen JJ, Yang-Yen HF. The antiapoptotic gene mcl-1 is upregulated by the phosphatidylinositol 3'-kinase/factor complex containing CREB. Mol Cell Biol. 1999;19: 6195–6206.
    1. Kõks S, Velthut A, Sarapik A, Altmäe S, Reinmaa E, Schalkwyk LC, et al. The differential transcriptome and ontology profiles of floating and cumulus granulosa cells in stimulated human antral follicles. Mol Hum Reprod. 2010;16: 229–240. 10.1093/molehr/gap103
    1. Grøndahl ML, Andersen CY, Bogstad J, Borgbo T, Boujida VH, Borup R. Specific genes are selectively expressed between cumulus and granulosa cells from individual human pre-ovulatory follicles. Mol Hum Reprod. 2012;18: 572–584. 10.1093/molehr/gas035
    1. Bonnet A, Cabau C, Bouchez O, Sarry J, Marsaud N, Foissac S, et al. An overview of gene expression dynamics during early ovarian folliculogenesis: specificity of follicular compartments and bi-directional dialog. BMC Genomics. 2013;9: 904.
    1. Cooper TG, Noonan E, von Eckardstein S, Auger J, Baker HW, Hermann M, et al. World Health Organization reference values for human semen characteristics. Hum Reprod Update. 2010;16: 231–245. 10.1093/humupd/dmp048
    1. Assou S, Haouzi D, De Vos J, Hamamah S. Human cumulus cells as biomarkers for embryo and pregnancy outcomes. Mol Hum Rep. 2010;16: 531–538.
    1. Szaniszlo P, Wang N, Sinha M, Reece LM, Van Hook JW, Luxon BA, et al. Getting the right cells to the array: Gene expression microarray analysis of cell mixtures and sorted cells. Cytometry A. 2004;59: 191–202.
    1. Smyth GK. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat Appl Genet Mol Biol. 2004: Article3
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Roy Statist Soc: Series B (Statistical Methodology). 1995; 289–300.
    1. Sean D, Meltzer PS. GEOquery: a bridge between the Gene Expression Omnibus (GEO) and BioConductor. Bioinformatics. 2007;23: 1846–1847.
    1. Hong F, Breitling R, McEntee CW, Wittner BS, Nemhauser JL, Chory J. RankProd: a bioconductor package for detecting differentially expressed genes in meta-analysis. Bioinformatics. 2006;22: 2825–2827.
    1. Reiner A, Yekutieli D, Benjamini Y. Identifying differentially expressed genes using false discovery rate controlling procedures. Bioinformatics. 2003;19: 368–375.
    1. Nogales-Cadenas R, Carmona-Saez P, Vazquez M, Vicente C, Yang X, Tirado F, et al. GeneCodis: interpreting gene lists through enrichment analysis and integration of diverse biological information. Nucleic Acids Res. 2009;37: W317–322. 10.1093/nar/gkp416
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25: 402–408.
    1. Allison DB, Cui X, Page GP, Sabripour M. Microarray data analysis: from disarray to consolidation and consensus. Nat Rev Genet. 2006;7: 55–65.
    1. Ghosh D, Barette TR, Rhodes D, Chinnaiyan AM. Statistical issues and methods for meta-analysis of microarray data: a case study in prostate cancer. Funct Integr Genomics. 2003;3: 180–188.
    1. Li M, Bullock CM, Knauer DJ, Ehlert FJ, Zhou QY. Identification of two prokineticin cDNAs: Recombinant proteins potently contract gastrointestinal smith muscle. Mol Pharmacol. 2001;59: 692–698.
    1. Lin R. Characterization of endocrine gland-derived vascular endothelial growth factor signaling in adrenal cortex capillary endothelial cells. J Biol Chem. 2002;277: 8724–8729.
    1. Le Couter J, Lin R, Tejada M, Frantz G, Peale F, Hillan KJ, et al. The endocrine-gland-derived VEGF homologue Bv8 promotes angiogenesis in the testis: Localization of Bv8 receptors to endothelial cells. Proc Natl Acad Sci USA. 2003;100: 2685–2690.
    1. Zhou QY, Cheng MY. Prokineticin 2 and circadian clock output. FEBS J. 2005;272: 5703–5709.
    1. Balasubramanian R, Plummer L, Sidis Y, Pitteloud N, Martin C,Zhou QY, et al. The puzzles of the prokineticin 2 pathway in human reproduction. Mol Cell Endocrinol. 2011;346: 44–50. 10.1016/j.mce.2011.05.040
    1. Ng KL, Li JD, Cheng MY, Leslie FM, Lee AG, Zhou QY.Dependence of olfactory bulb neurogenesis on prokineticin 2 signaling. Science. 2005;308: 1923–1927.
    1. Matsumoto S. Abnormal development of the olfactory bulb and reproductive system in mice lacking prokineticin receptor PKR2. Proc Natl Acad Sci USA. 2006;103: 4140–4145.
    1. Pitteloud N, Zhang C, Pignatelli D, Li JD, Raivio T, Cole LW, et al. Loss-of-function mutation in the prokineticin 2 gene causes Kallmann syndrome and normosmic idiopathic hypogonadotropic hypogonadism. Proc Natl Acad Sci USA. 2007;104: 17447–17452.
    1. Dode C, Teixeira L, Levilliers J, Fouveaut C, Bouchard P, Kottler ML, et al. Kallmann syndrome: mutations in the genes encoding prokineticin-2 and prokineticin receptor-2. PLoS Genetics. 2006;2: e175
    1. Suzuki T, Sasano H, Takaya R, Fukaya T, Yajima ANagura H. Cyclic changes of vasculature and vascular phenotypes in normal human ovaries. Hum Rep. 1998;13: 953–959.
    1. Lam PM, Haines C. Vascular endothelial growth factor plays more than an angiogenic role in the female reproductive system. Fertil Steril. 2005;84: 1775–1778.
    1. Kisliouk T, Podlovni H, Spanel-Borowski K, Ovadia O, Zhou QY, Meidan R. Prokineticins (endocrine gland-derived vascular endothelial growth factor and BV8) in the bovine ovary: expression and role as mitogens and survival factors for corpus luteum-derived endothelial cells. Endocrinology. 2005;146: 3950–3958.
    1. Murdoch WJ, Lund SA. Prostaglandin-independent anovulatory mechanism of indomethacin action: inhibition of tumor necrosis factor alpha-induced sheep ovarian cell apoptosis. Biol Reprod. 1999;61: 1655–1659.
    1. Naito Y, Watanabe Y, Yokokura H, Sugita R, Nishio M, Hidaka H. Isoform-specific activation and structural diversity of calmodulin kinase I. J Biol Chem. 1997;272: 32704–32708.
    1. Jusuf AA, Rina S, Sakagami H, Terashima T. Expression of Ca2+/calmodulin-dependent protein kinase (CaMK) I beta 2 in developing rat CNS. Neuroscience. 2002;109: 407–420.
    1. Gardner HP, Rajan JV, Ha SI, Copeland NG, Gilbert DJ, Jenkins NA, et al. Cloning, characterization, and chromosomal localization of Pnck, a Ca2+/calmodulin-dependent protein kinase. Genomics. 2000;63: 279–288.
    1. Deb TB, Coticchia CM, Barndt R, Zuo H, Dickson RB, Johnson MD. Pregnancy upregulated nonubiquitous calmodulin kinase induces ligand-independent EGFR degradation. Am J Physiol Cell Physiol. 2008;295: C365–C377. 10.1152/ajpcell.00449.2007
    1. Fan HY, Liu Z, Shimada M, Sterneck E, Johnson PF, Hedrick SM, et al. MAPK3/1 (ERK1/2) in ovarian granulosa cells are essential for female fertility. Science. 2009;324: 938–941. 10.1126/science.1171396
    1. Parrott JA, Whaley PD, Skinner MK. Extrahepatic expression of fibrinogen by granulosa cells: potential role in ovulation. Endocrinology. 1993;133: 1645–1649.
    1. Rinn JL, Chang HY. Genome regulation by long noncoding RNAs. Annu Rev Biochem. 2012;81: 145–166. 10.1146/annurev-biochem-051410-092902
    1. Ponting CP, Oliver PL, Reik W. Evolution and functions of long noncoding RNAs. Cell. 2009;136: 629–641. 10.1016/j.cell.2009.02.006
    1. Yerushalmi GM, Salmon-Divon M, Yung Y, Maman E, Kedem A, Ophir L, et al. Characterization of the human cumulus cell transcriptome during final follicular maturation and ovulation. Mol Hum Reprod. 2014;20: 719–735. 10.1093/molehr/gau031
    1. Xu XF, Li J, Cao YX, Chen DW, Zhang ZG, He XJ, et al. Differential Expression of Long Noncoding RNAs in Human Cumulus Cells Related to Embryo Developmental Potential: A Microarray Analysis. Reprod Sci. 2015;22: 672–678. 10.1177/1933719114561562
    1. Espey LL. Current status of the hypothesis that mammalian ovulation is comparable to an inflammatory reaction. Biol Reprod. 1994;50: 233–238.
    1. Fleming JS, Beaugie CR, Haviv I, Chenevix-Trench G, Tan OL. Incessant ovulation, inflammation and epithelial ovarian carcinogenesis: revisiting old hypotheses. Mol Cell Endocrinol. 2006;247: 4–21.
    1. Dinarello CA. The interleukin-1 family: 10 years of discovery. FASEB J. 1994;8: 1314–1325.
    1. Sims JE, Dower SK. Interleukin-1 receptors. Eur Cytokine Netw. 1991;5: 539–546.
    1. Arend WP. Interleukin 1 receptor antagonist: A new member of the interleukin 1 family. J Clin Invest. 1991;88: 1445–1451.
    1. Piquette GN, Simon C, el Danasouri I, Frances A, Polan ML. Gene regulation of interleukin-1 beta, interleukin-1 receptor type I, and plasminogen activator inhibitor-1 and -2 in human granulosa-luteal cells. Fertil Steril. 1994;62: 760–770.
    1. Martoriati A, Gérard N. Interleukin-1 (IL-1) system gene expression in granulosa cells: kinetics during terminal preovulatory follicle maturation in the mare. Reprod Biol Endocrinol. 2003;6:42.
    1. Gérard N, Caillaud M, Martoriati A, Goudet G, Lalmanach AC. The interleukin-1 system and female reproduction. J Endocrinol. 2004;180: 203–212.
    1. Rae MT, Niven D, Ross A, Forster T, Lathe R, Critchley HO, et al. Steroid signalling in human ovarian surface epithelial cells: the response to interleukin-1alpha determined by microarray analysis. J Endocrinol. 2004;183: 19–28.
    1. Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC. Ovarian surface epithelium: biology, endocrinology, and pathology. Endocr Rev. 2001;22: 255–288.
    1. Kawasaki F, Kawano Y, Kosay Hasan Z, Narahara H, Miyakawa I. The clinical role of interleukin-6 and interleukin-6 soluble receptor in human follicular fluids. Clin Exp Med. 2003;3: 27–31.
    1. Shimada M, Hernandez-Gonzalez I, Gonzalez-Robanya I, Richards JS. Induced expression of pattern recognition receptors in cumulus oocyte complexes: novel evidence for innate immune-like functions during ovulation. Mol Endocrinol. 2006;20: 3228–3239.
    1. Liu Z, Shimada M, Richards JS. The involvement of the toll-like receptor family in ovulation. J Assist Reprod Gen. 2008;25: 223–228.
    1. Salustri A, Garlanda C, Hirsch E, De Acetis M, Maccagno A, Bottazzi B, et al. PTX3 plays a key role in the organization of the cumulus oophorus extracellular matrix and in in vivo fertilization. Development. 2004;131: 1577–1586.
    1. Shimada M, Yanai Y, Okazaki T, Noma N, Kawashima I, Mori T, et al. Hyaluronan fragments generated by sperm-secreted hyaluronidase stimulate cytokine/chemokine production via the TLR2 and TLR4 pathway in cumulus cells of ovulated COCs, which may enhance fertilization. Development. 2008;135: 2001–2011. 10.1242/dev.020461
    1. Woods DC, Schorey JS, Johnson AL. Toll-like receptor signaling in hen ovarian granulosa cells is dependent on stage of follicle maturation. Reproduction. 2009;137: 987–996. 10.1530/REP-08-0320
    1. Zhang Z, Schluesener HJ. Mammalian toll-like receptors: from endogenous ligands to tissue regeneration. Cell Mol Life Sci. 2006;63: 2901–2907.
    1. Pricci F, Pugliese G, Romano G, Romeo G, Locuratolo N, Pugliese F, et al. Insulin-like growth factors I and II stimulate extracellular matrix production in human glomerular mesangial cells. Comparison with transforming growth factor-beta. Endocrinol. 1996;137: 879–885.
    1. Giudice LC. Insulin-like growth factors and ovarian development. Endocr Rev. 1992;13: 641–669.
    1. Monget P, Bondy C. Importance of the IGF system in early folliculogenesis. Mol Cell Endocrinol. 2000;163: 89–93.
    1. Ingman WV, Owens PC, Armstrong DT. Differential regulation by FSH and IGF-I of extracellular matrix IGFBP-5 in bovine granulosa cells: effect of association with the oocyte. Mol Cell Endocrinol. 2000;164: 53–58.
    1. Familiari G, Verlengia C, Nottola SA, Renda T, Micara G, Aragona C, et al. Heterogeneous distribution of fibronectin, tenascin-C, and laminin immunoreactive material in the cumulus-corona cells surrounding mature human oocytes from IVF-ET protocols-evidence that they are composed of different subpopulations: an immunohistochemical study using scanning confocal laser and fluorescence microscopy. Mol Reprod Dev. 1996;43: 392–402.
    1. Erickson H, Bourdon M. Tenascin: an extracellular matrix protein prominent in specialized embryonic tissues and tumours. Annu Rev Cell Biol. 1989;5: 71–92.
    1. Wilson KE, Langdon SP, Lessells AM, Miller WR. Expression of the extracellular matrix protein tenascin in malignant and benign ovarian tumours. Br J Cancer. 1996;74: 999–1004.
    1. Wilson KE, Bartlett JM, Miller EP, Smyth JF, Mullen P, Miller WR, et al. Regulation and function of the extracellular matrix protein tenascin-C in ovarian cancer cell lines. Br J Cancer. 1999;80: 685–692.
    1. Vanderhyden BC, Macdonald EA, Nagyova E, Dhawan A. Evaluation of members of the TGF beta superfamily as candidates for the oocyte factors that control mouse cumulus expansion and steroidogenesis. Hum Reprod Suppl. 2003;61: 55–70.
    1. Dragovic RA, Ritter LJ, Schulz SJ, Amato F, Armstrong DT, Gilchrist RB. Role of oocyte-secreted growth differentiation factor 9 in the regulation of mouse cumulus expansion. Endocrinol. 2005;146: 2798–2806.
    1. Gilchrist RB, Ritter LJ, Myllymaa S, Kaivo-Oja N, Dragovic RA, Hickey TE, et al. Molecular basis of oocyte-paracrine signalling that promotes granulosa cell proliferation. J Cell Sci. 2006;119: 3811–3821.

Source: PubMed

3
S'abonner