The microRNAs important for ovarian and early embryonic development in cattle

Recent progress in genomic analysis and other biochemical methods has led to the discovery of a large population of microRNAs (miRNAs), which have been demonstrated to play important roles in diseases and a wide range of developmental processes. Characterization of miRNA expression profiles in different stages of ovarian follicular development and early embryogenesis has suggested the potential roles of miRNAs in follicular development, maturation of oocytes, and preimplantation embryonic development. This review focuses on the current studies of miRNAs involved in ovarian and early embryonic development in cattle.

The microRNAs important for ovarian and early embryonic development in cattle

Recent progress in genomic analysis and other biochemical methods has led to the discovery of a large population of microRNAs (miRNAs), which have been demonstrated to play important roles in diseases and a wide range of developmental processes. Characterization of miRNA expression profiles in different stages of ovarian follicular development and early embryogenesis has suggested the potential roles of miRNAs in follicular development, maturation of oocytes, and preimplantation embryonic development. This review focuses on the current studies of miRNAs involved in ovarian and early embryonic development in cattle.

___

  • Kropp J, Penagaricano F, Salih SM, Khatib H. Invited review: Genetic contributions underlying the development of preimplantation bovine embryos. J Dairy Sci 2014; 97: 1187– 1201.
  • Evans ACO, Walsh SW. The physiology of multifactorial problems limiting the establishment of pregnancy in dairy cattle. Reprod Fert Develop 2011; 24: 233–237.
  • Memili E, First NL. Zygotic and embryonic gene expression in cow: a review of timing and mechanisms of early gene expression as compared with other species. Zygote 1999; 8: 87–96.
  • Hossain MM, Salilew-Wondim D, Schellander K, Tesfaye D. The role of microRNAs in mammalian oocytes and embryos. Anim Reprod Sci 2012; 134: 36–44. 8. Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 1993; 75: 843–854.
  • Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009; 136: 215–233.
  • Place RF, Li LC, Pookot D, Noonan EJ, Dahiya R. MicroRNA-373 induces expression of genes with complementary promoter sequences. P Natl Acad Sci USA 2008; 105: 1608–1613.
  • Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004; 116: 281–297.
  • Rodriguez A, Griffiths-Jones S, Ashurst JL, Bradley A. Identification of mammalian microRNA host genes and transcription units. Genome Res 2004; 14: 1902–1910.
  • Corcoran DL, Pandit KV, Gordon B, Bhattacharjee A, Kaminski N, Benos PV. Features of mammalian microRNA promoters emerge from polymerase II chromatin immunoprecipitation data. PLoS One 2009; 4: e5279.
  • Han L, Witmer PD, Casey E, Valle D, Sukumar S. DNA methylation regulates microRNA expression. Cancer Biol Ther 2007; 6: 1284–1288.
  • Borchert GM, Lanier W, Davidson BL. RNA polymerase III transcribes human microRNAs. Nat Struct Mol Biol 2006; 13: 1097–1101.
  • Winter J, Jung S, Keller S, Gregory RI, Diederichs S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat Cell Biol 2009; 11: 228–234.
  • Bohnsack MT, Czaplinski K, Görlich D. Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs. RNA 2004; 10: 185–191.
  • Chendrimada TP, Gregory RI, Kumaraswamy E, Norman J, Cooch N, Nishikura K, Shiekhattar R. TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing. Nature 2005; 436: 740–744.
  • Khvorova A, Reynolds A, Jayasena SD. Functional siRNAs and miRNAs exhibit strand bias. Cell 2003; 115: 209–216.
  • O’Carroll D, Schaefer A. General principals of miRNA biogenesis and regulation in the brain. Neuropsychopharmacology 2013; 38: 39–54.
  • Westholm JO, Lai EC. Mirtrons: microRNA biogenesis via splicing. Biochimie 2011; 93: 1897–1904.
  • Tripurani SK, Xiao C, Salem M, Yao J. Cloning and analysis of fetal ovary microRNAs in cattle. Anim Reprod Sci 2010; 120: 16–22.
  • Hossain MM, Ghanem N, Hoelker M, Rings F, Phatsara C, Tholen E, Schellander K, Tesfaye D. Identification and characterization of miRNAs expressed in the bovine ovary. BMC Genet 2009; 10: 443.
  • Huang J, Ju Z, Li Q, Hou Q, Wang C, Li J, Li R, Wang L, Sun T, Hang S et al. Solexa sequencing of novel and differentially expressed microRNAs in testicular and ovarian tissues in Holstein cattle. Int J Biol Sci 2011; 7: 1016–1026.
  • Salilew-Wondim D, Ahmad I, Gebremedhn S, Sahadevan S, Hoelker M, Rings F, Udin J, Tholen E, Looft C, Schellander K et al. Regulatory microRNA enrichment and degradation in granulosa cells during bovine follicular recruitment and dominance. Reprod Fert Develop 2013; 26: 169–170.
  • Gebremedhn S, Ahmad I, Salilew-Wondim D, Sahadevan S, Hoelker M, Rings F, Udin J, Tholen E, Looft C, Schellander K et al. Expression profiling of noncoding microRNAs in bovine granulosa cells of preovulatory dominant follicle using deep sequencing. Reprod Fert Develop 2013; 26: 170–171.
  • Sontakke SD, Mohammed BT, McNeilly AS, Donadeu FX. Characterization of microRNAs differentially expressed during bovine follicle development. Reproduction 2014; 148: 271–283.
  • Sohel MMH, Hoelker M, Noferesti SS, Salilew-Wondim D, Tholen E, Looft C, Rings F, Uddin MJ, Spencer TE, Schellander K et al. Exosomal and non-exosomal transport of extra-cellular microRNAs in follicular fluid: implications for bovine oocyte developmental competence. PLoS One 2013; 8: e78505.
  • da Silveira JC, Veeramachaneni DNR, 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.
  • Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ, Lötvall JO. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 2007; 9: 654–659.
  • Properzi F, Logozzi M, Fais S. Exosomes: the future of biomarkers in medicine. Biomark Med 2013; 7: 769–778.
  • Tesfaye D, Worku D, Rings F, Phatsara C, Tholen E, Schellander K, Hoelker M. Identification and expression profiling of microRNAs during bovine oocyte maturation using heterologous approach. Mol Reprod Dev 2009; 76: 665–677.
  • Abd El Naby WS, Hagos TH, Hossain MM, Salilew-Wondim D, Gad AY, Rings F, Cinar MU, Tholen E, Looft C, Schellander K et al. Expression analysis of regulatory microRNAs in bovine cumulus oocyte complex and preimplantation embryos. Zygote 2013; 21: 31–51.
  • Sinha P, Hossain M, Rings F, Hoelker M, Phatsara C, Tholen E, Schellander K, Tesfaye D. Functional characterization of miR-130 family during bovine pre-implantation embryo development. Reprod Fert Develop 2009; 22: 283.
  • Miles JR, McDaneld TG, Wiedmann RT, Cushman RA, Echternkamp SE, Vallet JL, Smith TP. MicroRNA expression profile in bovine cumulus-oocyte complexes: possible role of let-7 and miR-106a in the development of bovine oocytes. Anim Reprod Sci 2012; 130: 16–26.
  • Richards JAS. Genetics of ovulation. Semin Reprod Med 2007; 25: 235–242.
  • Otsuka M, Zheng M, Hayashi M, Lee JD, Yoshino O, Lin S, Han J. Impaired microRNA processing causes corpus luteum insufficiency and infertility in mice. J Clin Invest 2008; 118: 1944–1954.
  • Carletti MZ, Fiedler SD, Christenson LK. MicroRNA 21 blocks apoptosis in mouse periovulatory granulosa cells. Biol Reprod 2010; 83: 286–295.
  • McBride D, Carré W, Sontakke SD, Hogg CO, Law A, Donadeu FX, Clinton M. Identification of miRNAs associated with the follicular-luteal transition in the ruminant ovary. Reproduction 2012; 144: 221–233.
  • Suh N, Baehner L, Moltzahn F, Melton C, Shenoy A, Chen J, Blelloch R. MicroRNA function is globally suppressed in mouse oocytes and early embryos. Curr Biol 2010; 20: 271– 277.
  • Ma J, Flemr M, Stein P, Berninger P, Malik R, Zavolan M, Svoboda P, Schultz RM. MicroRNA activity is suppressed in mouse oocytes. Curr Biol 2010; 20: 265–270.
  • Bazzini AA, Lee MT, Giraldez AJ. Ribosome profiling shows that miR-430 reduces translation before causing mRNA decay in zebrafish. Science 2012; 336: 233–237.
  • Lund E, Liu M, Hartley RS, Sheets MD, Dahlberg JE. Deadenylation of maternal mRNAs mediated by miR-427 in Xenopus laevis embryos. RNA 2009; 15: 2351–2363.
  • Giraldez AJ, Mishima Y, Rihel J, Grocock RJ, Van Dongen S, Inoue K, Enright AJ, Schier AF. Zebrafish MiR-430 promotes deadenylation and clearance of maternal mRNAs. Science 2006; 312: 75–79.
  • Ramachandra RK, Salem M, Gahr S, Rexroad CE, Yao J. Cloning and characterization of microRNAs from rainbow trout (Oncorhynchus mykiss): their expression during early embryonic development. BMC Dev Biol 2008; 8: 41.
  • Mondou E, Dufort I, Gohin M, Fournier E, Sirard MA. Analysis of microRNAs and their precursors in bovine early embryonic development. Mol Hum Reprod 2012; 18: 425–434.
  • Goossens K, Mestdagh P, Lefever S, Van Poucke M, Van Zeveren A, Van Soom A, Vandesompele J, Peelman L. Regulatory microRNA network identification in bovine blastocyst development. Stem Cells Dev 2013; 22: 1907–1920.
  • Zheng P, Dean J. Oocyte-specific genes affect folliculogenesis, fertilization, and early development. Semin Reprod Med 2007; 25: 243–251.
  • Sun QY, Liu K, Kikuchi K. Oocyte-specific knockout: a novel in vivo approach for studying gene functions during folliculogenesis, oocyte maturation, fertilization, and embryogenesis. Biol Reprod 2008; 79: 1014–1020.
  • Pangas SA, Rajkovic A. Transcriptional regulation of early oogenesis: in search of masters. Hum Reprod Update 2006; 12: 65–76.
  • Rajkovic A, Pangas SA, Ballow D, Suzumori N, Matzuk MM. NOBOX deficiency disrupts early folliculogenesis and oocyte- specific gene expression. Science 2004; 305: 1157–1159.
  • Soyal SM, Amleh A, Dean J. FIGα, a germ cell-specific transcription factor required for ovarian follicle formation. Development 2000; 127: 4645–4654.
  • Dong J, Albertini DF, Nishimori K, Kumar TR, Lu N, Matzuk MM. Growth differentiation factor-9 is required during early ovarian folliculogenesis. Nature 1996; 383: 531–535.
  • Virant-Klun I, Knez K, Tomazevic T, Skutella T. Gene expression profiling of human oocytes developed and matured in vivo or in vitro. BioMed Res Int 2013; 2013: 879489.
  • Philpott A, Leno GH, Laskey RA. Sperm decondensation in Xenopus egg cytoplasm is mediated by nucleoplasmin. Cell 1991; 65: 569–578.
  • Burns KH, Viveiros MM, Ren Y, Wang P, DeMayo FJ, Frail DE, Eppig JJ, Matzuk MM. Roles of NPM2 in chromatin and nucleolar organization in oocytes and embryos. Science 2003; 300: 633–636.
  • Betthauser JM, Pfister‐Genskow M, Xu H, Golueke PJ, Lacson JC, Koppang RW, Myers C, Liu B, Hoeschele I, Eilertsen KJ et al. Nucleoplasmin facilitates reprogramming and in vivo development of bovine nuclear transfer embryos. Mol Reprod Dev 2006; 73: 977–986.
  • Lingenfelter BM, Tripurani SK, Tejomurtula J, Smith GW, Yao J. Molecular cloning and expression of bovine nucleoplasmin 2 (NPM2): a maternal effect gene regulated by miR-181a. Reprod Biol Endocrin 2011; 9: 40.
  • Gu P, Reid JG, Gao X, Shaw Ca, Creighton C, Tran PL, Zhou X, Drabek RB, Steffen DL, Hoang DM et al. Novel microRNA candidates and miRNA-mRNA pairs in embryonic stem (ES) cells. PLoS One 2008; 3: e2548.
  • Suzumori N, Yan C, Matzuk MM, Rajkovic A. Nobox is a homeobox-encoding gene preferentially expressed in primordial and growing oocytes. Mech Develop 2002; 111: 137–141.
  • Tripurani SK, Wee G, Lee KB, Smith GW, Wang L, Yao J. MicroRNA-212 post-transcriptionally regulates oocyte- specific basic-helix-loop-helix transcription factor, factor in the germline alpha (FIGLA), during bovine early embryogenesis. PLoS One 2013; 8: e76114.
  • Otis KO, Thompson KR, Martin KC. Importin-mediated nuclear transport in neurons. Curr Opin Neurobiol 2006; 16: 329–335.
  • Chook YM, Blobel G. Karyopherins and nuclear import. Curr Opin Struc Biol 2001; 11: 703–715.
  • Tejomurtula J, Lee KB, Tripurani SK, Smith GW, Yao J. Role of importin α8, a new member of the importin α family of nuclear transport proteins, in early embryonic development in cattle. Biol Reprod 2009; 81: 333–342.
  • Hu J, Wang F, Yuan Y, Zhu X, Wang Y, Zhang Y, Kou Z, Wang S, Gao S. Novel importin-α family member Kpna7 is required for normal fertility and fecundity in the mouse. J Biol Chem 2010; 285: 33113–33122.
  • Wang X, Park KE, Koser S, Liu S, Magnani L, Cabot RA. KPNA7, an oocyte- and embryo-specific karyopherin α subtype, is required for porcine embryo development. Reprod Fert Develop 2012; 24: 382–391.
  • Wang L, Smith GW, Yao J. MicroRNA-1296 regulates bovine oocyte-specific karyropherin alpha 7 expression during maternal-zygotic transition. In: Society for the Study of Reproduction 46th Annual Meeting; Montreal, Quebec; 2013.
Turkish Journal of Veterinary and Animal Sciences-Cover
  • ISSN: 1300-0128
  • Yayın Aralığı: Yılda 6 Sayı
  • Yayıncı: TÜBİTAK
Sayıdaki Diğer Makaleler

Polledness intersex syndrome in goats – molecular and histological aspects

İwona SZATKOWSKA, Daniel ZABORSKI, Witold Stanislaw PROSKURA, Sara TABOR

Molecular, cellular, and physiological determinants of bull fertility

Alexis Marie PARISI, Shannon Kate THOMPSON, Abdullah KAYA, Erdoğan MEMİLİ

Study of insulin-like growth factor 1 effects on bovine type A spermatogonia proliferation and viability

Babak QASEMI PANAHI, Parviz TAJIK, Mansoureh MOVAHEDIN, Gholamali MOGHADDAM, Mohammad Hossein GERANMAYEH

Recent advances in the immunology and uterine microbiology of healthy cows and cows that develop uterine disease

Klibs N. GALVAO, Jose Eduardo P. SANTOS

Cholesterol efux from sperm: approaches and applications

Melih AKSOY, Ejaz AHMAD, Zahid NASEER

New understanding of an old phenomenon: uncontrolled factors and misconceptions that cast a shadow over studies of the male efect' on reproduction in small ruminants

Trina JORRE DE ST JORRE, Graeme Bruce MARTIN, Penny Alison Rhian HAWKEN

Application of dietary fish oil in dairy cow reproduction

Anna ZWYRZYKOWSKA, Robert KUPCZYNSKI

Radoslava VLCKOVA, Jan POSIVAK, İgor VALOCKY, Zuzana KRAVCOVA, Andrea EIBENOVA, Drahomíra SOPKOVA

New understanding of an old phenomenon: uncontrolled factors and misconceptions that cast a shadow over studies of the ‘male effect’ on reproduction in small ruminants

Trina Jorre De St JORRE, Penny Alison Rhian HAWKEN, Graeme Bruce MARTIN

Investigation of the effcacy of Tarantula cubensis extract (Teranekron D6) in the treatment of subclinical and clinical mastitis in dairy cows

Esra CANOOĞLU, Murat ABAY, Kutlay GÜRBULAK, Aytaç AKÇAY, Tayfur BEKYÜREK, Birgit SIST, Simone STEINER, Kadir Semih GÜMÜŞSOY