MicroRNA-379-5p is associate with biochemical premature ovarian insufficiency through PARP1 and XRCC6

Yujie Dang, Xiaoyan Wang, Yajing Hao, Xinyue Zhang, Shidou Zhao, Jinlong Ma, Yingying Qin, Zi-Jiang Chen, Yujie Dang, Xiaoyan Wang, Yajing Hao, Xinyue Zhang, Shidou Zhao, Jinlong Ma, Yingying Qin, Zi-Jiang Chen

Abstract

Premature ovarian insufficiency (POI) imposes great challenges on women's fertility and lifelong health. POI is highly heterogeneous and encompasses occult, biochemical, and overt stages. MicroRNAs (miRNAs) are negative regulators of gene expression, whose roles in physiology and diseases like cancers and neurological disorders have been recognized, but little is known about the miRNAs profile and functional relevance in biochemical POI (bPOI). In this study, the expression of miRNAs and mRNAs in granulosa cells (GCs) of bPOI women was determined by two microarrays, respectively. MiR-379-5p, PARP1, and XRCC6 were differentially expressed in GCs of bPOI as revealed by microarrays. Subsequently, functional studies demonstrated that miR-379-5p overexpression inhibited granulosa cell proliferation and attenuated DNA repair efficiency. Furthermore, both PARP1 and XRCC6 showed lower levels in GCs from patients with bPOI and were identified as executives of miR-379-5p. Therefore, our data first uncovered potentially pathogenic miR-379-5p and two novel targets PARP1 and XRCC6 in bPOI, which corroborated the significance of DNA repair for POI, and brought up an epigenetic explanation for the disease.

Conflict of interest statement

The authors declare that they have no conflict of interest.

Figures

Fig. 1. MiR-379-5p, PARP1 , and XRCC6…
Fig. 1. MiR-379-5p, PARP1, and XRCC6 are differentially expressed in GCs of bPOI as revealed by microarray
a Heat map of miR-379-5p, PARP1, and XRCC6 based on microarray data. Each column represents a sample; red and green indicates up- and downregulation, respectively; r correlation coefficient. b, c, d Quantitative PCR (qPCR) analysis in GCs from women with and without bPOI shows that miR-379-5p increases (b), while PARP1 and XRCC6 decrease in bPOI patients (c, d). Two-tailed Mann–Whitney U-test
Fig. 2. PARP1 and XRCC6 are novel…
Fig. 2. PARP1 and XRCC6 are novel targets of miR-379-5p
a, b qRT-PCR and western blot analyses indicate that PARP1 and XRCC6 mRNA (a) and protein (b) levels are significantly reduced by forced expression of miR-379-5p in KGN cells. c, d Predicted target sites and relevant mutation sequences are shown. Luciferase assay demonstrates that overexpression of miR-379-5p leads to a decrease in PARP1 and XRCC6 with wild-type 3′-UTRs, while luciferase activity levels of mutated 3′-UTRs unchanged. Asterisk (*) indicates a significant difference. *P < 0.05, two-tailed Student's t test
Fig. 3. Overexpression of miR-379-5p leads to…
Fig. 3. Overexpression of miR-379-5p leads to proliferation inhibition and DNA repair impairment
a CCK-8 assay reveals that miR-379-5p overexpression led to suppression in cell proliferation. b, c EdU assay shows less EdU-positive cells in miR-379-5p overexpressed group. d PCNA level is much lower in KGN cells overexpressing miR-379-5p than negative control. e After exposed to MMC for 6 h, more of γH2AX is found in KGN cells overexpressing miR-379-5p. After recovery for 2 and 4 h, miR-379-5p delays the DNA damage repair. f, g The clonogenic survival of HeLa cells suffering MMC treatment and 2 h’ recovery. The clonogenic survival percent of miR-379-5p mimics group is significantly less than that of negative control. Asterisk (*) indicates a significant difference. *P < 0.05, **P < 0.01, two-tailed Student's t test
Fig. 4. PARP1 and XRCC6 are functional…
Fig. 4. PARP1 and XRCC6 are functional targets of miR-379-5p
a An efficient decrease in PARP1 and XRCC6 protein levels is confirmed by western blot in KGN cells 48 h post transfection. b, c, d Suppressed cell proliferation in PARP1 and XRCC6 knockdown group is indicated by CCK-8 assay (b) and EdU staining (c, d). e PCNA level is much lower in PARP1 and XRCC6 silencing cells than negative control cells. f After exposed to MMC for 6 h, and recovery for 2 and 4 h, PARP1 and XRCC6 silencing delayed the DNA damage repair. g, h The percent survival relative to the control is significantly lower in PARP1 and XRCC6 depletion cells. Asterisk (*) indicates a significant difference. *P < 0.05, **P < 0.01, ***P < 0.001, two-tailed Student's t test
Fig. 5. Reintroduction of PARP1 and XRCC6…
Fig. 5. Reintroduction of PARP1 and XRCC6 abrogates the miR-379-5p-induced effects partially
a An efficient increase in PARP1 and XRCC6 protein levels is confirmed by western blot in KGN cells 48 h post co-transfection infection. b, cPARP1 and XRCC6 overexpression could promote cell proliferation. d After exposed to MMC for 6 h, more of γH2AX is found in two groups overexpressing miR-379-5p. However, after 4 h recovery, PARP1 and XRCC6 overexpression group shows similarly less γH2AX as the NC + Ad-Null group. The results uncover that PARP1 and XRCC6 is required for efficient DNA damage repair process

References

    1. Nelson LM. Clinical practice. Primary ovarian insufficiency. N. Engl. J. Med. 2009;360:606–614. doi: 10.1056/NEJMcp0808697.
    1. European Society for Human Reproduction and Embryology (ESHRE) Guideline Group on POI. Webber L, et al. ESHRE guideline: management of women with premature ovarian insufficiency. Hum. Reprod. 2016;31:926–937. doi: 10.1093/humrep/dew027.
    1. Wu X, et al. Impact of premature ovarian failure on mortality and morbidity among Chinese women. PLoS ONE. 2014;9:e89597. doi: 10.1371/journal.pone.0089597.
    1. Welt CK. Primary ovarian insufficiency: a more accurate term for premature ovarian failure. Clin. Endocrinol. 2008;68:499–509. doi: 10.1111/j.1365-2265.2007.03073.x.
    1. Knauff EA, et al. Anti-Mullerian hormone, inhibin B, and antral follicle count in young women with ovarian failure. J. Clin. Endocrinol. Metab. 2009;94:786–792. doi: 10.1210/jc.2008-1818.
    1. De Vos M, Devroey P, Fauser BC. Primary ovarian insufficiency. Lancet. 2010;376:911–921. doi: 10.1016/S0140-6736(10)60355-8.
    1. Jorgez CJ, Klysik M, Jamin SP, Behringer RR, Matzuk MM. Granulosa cell-specific inactivation of follistatin causes female fertility defects. Mol. Endocrinol. 2004;18:953–967. doi: 10.1210/me.2003-0301.
    1. Edson MA, Nagaraja AK, Matzuk MM. The mammalian ovary from genesis to revelation. Endocr. Rev. 2009;30:624–712. doi: 10.1210/er.2009-0012.
    1. Matsuda F, Inoue N, Manabe N, Ohkura S. Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. J. Reprod. Dev. 2012;58:44–50. doi: 10.1262/jrd.2011-012.
    1. Dong J, et al. Growth differentiation factor-9 is required during early ovarian folliculogenesis. Nature. 1996;383:531–535. doi: 10.1038/383531a0.
    1. Lechowska A, et al. Premature ovarian failure in nobox-deficient mice is caused by defects in somatic cell invasion and germ cell cyst breakdown. J. Assist. Reprod. Genet. 2011;28:583–589. doi: 10.1007/s10815-011-9553-5.
    1. Sirotkin AV, Ovcharenko D, Grossmann R, Lauková M, Mlyncek M. Identification of microRNAs controlling human ovarian cell steroidogenesis via a genome-scale screen. J. Cell Physiol. 2009;219:415–420. doi: 10.1002/jcp.21689.
    1. Imbar T, Eisenberg I. Regulatory role of microRNAs in ovarian function. Fertil. Steril. 2014;101:1524–1530. doi: 10.1016/j.fertnstert.2014.04.024.
    1. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/S0092-8674(04)00045-5.
    1. Esteller M. Non-coding RNAs in human disease. Nat. Rev. Genet. 2011;12:861–874. doi: 10.1038/nrg3074.
    1. Rah H, et al. Association of miR-146aC>G, miR-196a2T>C, and miR-499A>G polymorphisms with risk of premature ovarian failure in Korean women. Reprod. Sci. 2013;20:60–68. doi: 10.1177/1933719112450341.
    1. Pan H, et al. The miR-449b polymorphism, rs10061133 A>G, is associated with premature ovarian insufficiency. Menopause. 2016;23:1009–1011. doi: 10.1097/GME.0000000000000659.
    1. Yang X, et al. Differentially expressed plasma microRNAs in premature ovarian failure patients and the potential regulatory function of mir-23a in granulosa cell apoptosis. Reproduction. 2012;144:235–244. doi: 10.1530/REP-11-0371.
    1. Zhang Q, et al. MicroRNA-181a suppresses mouse granulosa cell proliferation by targeting activin receptor IIA. PLoS ONE. 2013;8:e59667. doi: 10.1371/journal.pone.0059667.
    1. Dang Y, et al. MicroRNA-22-3p is down-regulated in the plasma of Han Chinese patients with premature ovarian failure. Fertil. Steril. 2015;103:802–807.e1. doi: 10.1016/j.fertnstert.2014.12.106.
    1. Chen X, Xie M, Liu D, Shi K. Down regulation of microRNA-146a inhibits ovarian granulosa cell apoptosis by simultaneously targeting interleukin-1 receptor-associated kinase and tumor necrosis factor receptor-associated factor 6. Mol. Med. Rep. 2015;12:5155–5162. doi: 10.3892/mmr.2015.4036.
    1. da Rocha ST, Edwards CA, Ito M, Ogata T, Ferguson-Smith AC. Genomic imprinting at the mammalian Dlk1-Dio3 domain. Trends Genet. 2008;24:306–316. doi: 10.1016/j.tig.2008.03.011.
    1. Stadtfeld M, et al. Aberrant silencing of imprinted genes on chromosome 12qF1 in mouse induced pluripotent stem cells. Nature. 2010;465:175–181. doi: 10.1038/nature09017.
    1. Gardiner E, et al. Imprinted DLK1-DIO3 region of 14q32 defines a schizophrenia-associated miRNA signature in peripheral blood mononuclear cells. Mol. Psychiatry. 2012;17:827–840. doi: 10.1038/mp.2011.78.
    1. Labialle S, et al. The miR-379/miR-410 cluster at the imprinted Dlk1-Dio3 domain controls neonatal metabolic adaptation. EMBO J. 2014;33:2216–2230. doi: 10.15252/embj.201387038.
    1. Khan S, et al. MiR-379 regulates cyclin B1 expression and is decreased in breast cancer. PLoS ONE. 2013;8:e68753. doi: 10.1371/journal.pone.0068753.
    1. Gururajan M, et al. miR-154* and miR-379 in the DLK1-DIO3 microRNA mega-cluster regulate epithelial to mesenchymal transitionand bone metastasis of prostate cancer. Clin. Cancer Res. 2014;20:6559–6569. doi: 10.1158/1078-0432.CCR-14-1784.
    1. Chen JS, et al. MicroRNA-379-5p inhibits tumor invasion and metastasis by targeting FAK/AKT signaling in hepatocellular carcinoma. Cancer Lett. 2016;375:73–83. doi: 10.1016/j.canlet.2016.02.043.
    1. AlAsiri S, et al. Exome sequencing reveals MCM8 mutation underlies ovarian failure and chromosomal instability. J. Clin. Invest. 2015;125:258–262. doi: 10.1172/JCI78473.
    1. Dou X, et al. Minichromosome maintenance complex component 8 mutations cause primary ovarian insufficiency. Fertil. Steril. 2016;106:1485–1489.e2. doi: 10.1016/j.fertnstert.2016.08.018.
    1. Wood-Trageser MA, et al. MCM9 mutations are associated with ovarian failure, short stature, and chromosomal instability. Am. J. Hum. Genet. 2014;95:754–762. doi: 10.1016/j.ajhg.2014.11.002.
    1. Desai S, et al. MCM8 and MCM9 nucleotide variants in women with primary ovarian insufficiency. J. Clin. Endocrinol. Metab. 2017;102:576–582.
    1. Qin Y, et al. CSB-PGBD3 mutations cause premature ovarian failure. PLoS Genet. 2015;11:e1005419. doi: 10.1371/journal.pgen.1005419.
    1. Guo T, et al. Mutations in MSH5 in primary ovarian insufficiency. Hum. Mol. Genet. 2017;26:1452–1457. doi: 10.1093/hmg/ddx044.
    1. He C, et al. Genome-wide association studies identify loci associated with age at menarche and age at natural menopause. Nat. Genet. 2009;41:724–728. doi: 10.1038/ng.385.
    1. He C, et al. A large-scale candidate gene association study of age at menarche and age at natural menopause. Hum. Genet. 2010;128:515–527. doi: 10.1007/s00439-010-0878-4.
    1. Stolk L, et al. Meta-analyses identify 13 loci associated with age at menopause and highlight DNA repair and immune pathways. Nat. Genet. 2012;44:260–268. doi: 10.1038/ng.1051.
    1. Day FR, et al. Large-scale genomic analyses link reproductive aging to hypothalamic signaling, breast cancer susceptibility and BRCA1-mediated DNA repair. Nat. Genet. 2015;47:1294–1303. doi: 10.1038/ng.3412.
    1. Rouleau M, Patel A, Hendzel MJ, Kaufmann SH, Poirier GG. PARP inhibition: PARP1 and beyond. Nat. Rev. Cancer. 2010;10:293–301. doi: 10.1038/nrc2812.
    1. Celli GB, Denchi EL, de Lange T. Ku70 stimulates fusion of dysfunctional telomeres yet protects chromosome ends from homologous recombination. Nat. Cell Biol. 2006;8:885–890. doi: 10.1038/ncb1444.
    1. Jiang L, et al. MicroRNA-93 promotes ovarian granulosa cells proliferation through targeting CDKN1A in polycystic ovarian syndrome. J. Clin. Endocrinol. Metab. 2015;100:E729–E738. doi: 10.1210/jc.2014-3827.
    1. Gao F, et al. Wt1 functions in ovarian follicle development by regulating granulosa cell differentiation. Hum. Mol. Genet. 2014;23:333–341. doi: 10.1093/hmg/ddt423.
    1. Titus S, et al. Impairment of BRCA1-related DNA double-strand break repair leads to ovarian aging in mice and humans. Sci. Transl. Med. 2013;5:172ra21. doi: 10.1126/scitranslmed.3004925.
    1. Oktay K, Turan V, Titus S, Stobezki R, Liu L. BRCA mutations, DNA repair deficiency, and ovarian aging. Biol. Reprod. 2015;93:67. doi: 10.1095/biolreprod.115.132290.
    1. Zhang D, et al. Increased DNA damage and repair deficiency in granulosa cells are associated with ovarian aging in rhesus monkey. J. Assist. Reprod. Genet. 2015;32:1069–1078. doi: 10.1007/s10815-015-0483-5.
    1. Bürkle A, Beneke S, Muiras ML. Poly(ADP-ribosyl)ation and aging. Exp. Gerontol. 2004;39:1599–1601. doi: 10.1016/j.exger.2004.07.010.
    1. Bai P, Cantó C. The role of PARP-1 and PARP-2 enzymes in metabolic regulation and disease. Cell Metab. 2012;16:290–295. doi: 10.1016/j.cmet.2012.06.016.
    1. Shimizu I, Yoshida Y, Suda M, Minamino T. DNA damage response and metabolic disease. Cell Metab. 2014;20:967–977. doi: 10.1016/j.cmet.2014.10.008.
    1. Downs JA, Jackson SP. A means to a DNA end: the many roles of Ku. Nat. Rev. Mol. Cell Biol. 2004;5:367–378. doi: 10.1038/nrm1367.
    1. Ménissier de Murcia J, et al. Functional interaction between PARP-1 and PARP-2 in chromosome stability and embryonic development in mouse. EMBO J. 2003;22:2255–2263. doi: 10.1093/emboj/cdg206.
    1. Piskunova T. S. et al. Deficiency in poly (ADP-ribose) polymerase-1 (PARP-1) accelerates aging and spontaneous carcinogenesis in mice. Curr. Gerontol. Geriatr. Res.2008, 754190 (2008).
    1. Gu Y, et al. Growth retardation and leaky SCID phenotype of Ku70-deficient mice. Immunity. 1997;7:653–665. doi: 10.1016/S1074-7613(00)80386-6.
    1. Li H, Vogel H, Holcomb VB, Gu Y, Hasty P. Deletion of Ku70, Ku80, or both causes early aging without substantially increased cancer. Mol. Cell Biol. 2007;27:8205–8214. doi: 10.1128/MCB.00785-07.
    1. Ma T, et al. Microarray analysis of differentially expressed microRNAs in non-regressed and regressed bovine corpus luteum tissue; microRNA-378 may suppress luteal cell apoptosis by targeting the interferon gamma receptor 1 gene. J. Appl. Genet. 2011;52:481–486. doi: 10.1007/s13353-011-0055-z.
    1. Tesfaye D, et al. Identification and expression profiling of microRNAs during bovine oocyte maturation using heterologous approach. Mol. Reprod. Dev. 2009;76:665–677. doi: 10.1002/mrd.21005.
    1. da Silveira JC, Veeramachaneni DN, Winger QA, Carnevale EM, Bouma GJ. Cell-secreted vesicles in equine ovarian follicular fluid contain miRNAs and proteins: a possible new form of cell communication within the ovarian follicle. Biol. Reprod. 2012;86:71. doi: 10.1095/biolreprod.111.093252.
    1. Jiao X. et al. Premature ovarian insufficiency: phenotypic characterization within different etiologies. J. Clin. Endocrinol. Metab. (2017)..
    1. Xu X, et al. Impaired telomere length and telomerase activity in peripheral blood leukocytes and granulosa cells in patients with biochemical primary ovarian insufficiency. Hum. Reprod. 2017;32:201–207.
    1. Nishi Y, et al. Establishment and characterization of a steroidogenic human granulosa-like tumor cell line, KGN, that expresses functional follicle-stimulating hormone receptor. Endocrinology. 2001;142:437–445. doi: 10.1210/endo.142.1.7862.
    1. Patel AG, Sarkaria JN, Kaufmann SH. Nonhomologous end joining drives poly(ADP-ribose) polymerase (PARP) inhibitor lethality in homologous recombination-deficient cells. Proc. Natl Acad. Sci. USA. 2011;108:3406–3411. doi: 10.1073/pnas.1013715108.
    1. Ström CE, Johansson F, Uhlén M, Szigyarto CA, Erixon K, Helleday T. Poly (ADP-ribose) polymerase (PARP) is not involved in base excision repair but PARP inhibition traps a single-strand intermediate. Proc. Nucleic Acids Res. 2011;39:3166–3175. doi: 10.1093/nar/gkq1241.

Source: PubMed

3
Subskrybuj