期刊论文详细信息
BMC Developmental Biology
Expression of maternally derived KHDC3, NLRP5, OOEP and TLE6 is associated with oocyte developmental competence in the ovine species
Sergio Ledda1  Laura Falchi1  Mauro Fattorini1  Ombretta Murrone1  Laura Masala1  Luisa Bogliolo1  Federica Ariu1  Daniela Bebbere1 
[1] Department of Veterinary Medicine, University of Sassari, via Vienna 2, Sassari, 07100, Italy
关键词: Subcortical Maternal Complex;    Oocyte developmental competence;    Ovine embryo;   
Others  :  1084775
DOI  :  10.1186/s12861-014-0040-y
 received in 2014-04-22, accepted in 2014-11-11,  发布年份 2014
PDF
【 摘 要 】

Background

The sub-cortical maternal complex (SCMC), located in the subcortex of mouse oocytes and preimplantation embryos, is composed of at least four proteins encoded by maternal effect genes: OOEP, NLRP5/MATER, TLE6 and KHDC3/FILIA. The SCMC assembles during oocyte growth and was seen to be essential for murine zygote progression beyond the first embryonic cell divisions; although roles in chromatin reprogramming and embryonic genome activation were hypothesized, the full range of functions of the complex in preimplantation development remains largely unknown.

Results

Here we report the expression of the SCMC genes in ovine oocytes and pre-implantation embryos, describing for the first time its expression in a large mammalian species.

We report sheep-specific patterns of expression and a relationship with the oocyte developmental potential in terms of delayed degradation of maternal SCMC transcripts in pre-implantation embryos derived from developmentally incompetent oocytes.

In addition, by determining OOEP full length cDNA by Rapid Amplification of cDNA Ends (RACE) we identified two different transcript variants (OOEP1 and OOEP2), both expressed in oocytes and early embryos, but with different somatic tissue distributions.

In silico translation showed that 140 aminoacid peptide OOEP1 shares an identity with orthologous proteins ranging from 95% with the bovine to 45% with mouse. Conversely, OOEP2 contains a premature termination codon, thus representing an alternative noncoding transcript and supporting the existence of aberrant splicing during ovine oogenesis.

Conclusions

These findings confirm the existence of the SCMC in sheep and its key role for the oocyte developmental potential, deepening our understanding on the molecular differences underlying cytoplasmic vs nuclear maturation of the oocytes.

Describing differences and overlaps in transcriptome composition between model organisms advance our comprehension of the diversity/uniformity between mammalian species during early embryonic development and provide information on genes that play important regulatory roles in fertility in nonmurine models, including the human.

【 授权许可】

   
2014 Bebbere et al.; licensee BioMed Central Ltd.

【 预 览 】
附件列表
Files Size Format View
20150113164300413.pdf 2056KB PDF download
Figure 5. 22KB Image download
Figure 4. 134KB Image download
Figure 3. 145KB Image download
Figure 2. 76KB Image download
Figure 1. 28KB Image download
【 图 表 】

Figure 1.

Figure 2.

Figure 3.

Figure 4.

Figure 5.

【 参考文献 】
  • [1]Li L, Baibakov B, Dean J: A subcortical maternal complex essential for preimplantation mouse embryogenesis. Dev Cell 2008, 15:416-425.
  • [2]Tong ZB, Gold L, Pfeifer KE, Dorward H, Lee E, Bondy CA, Dean J, Nelson LM: Mater, a maternal effect gene required for early embryonic development in mice. Nat Genet 2000, 26:267-268.
  • [3]Tashiro F, Kanai-Azuma M, Miyazaki S, Kato M, Tanaka T, Toyoda S, Yamato E, Kawakami H, Miyazaki T, Miyazaki J: Maternal-effect gene Ces5/Ooep/Moep19/Floped is essential for oocyte cytoplasmic lattice formation and embryonic development at the maternal-zygotic stage transition. Genes Cells 2010, 15:813-828.
  • [4]Zheng P, Dean J: Role of Filia, a maternal effect gene, in maintaining euploidy during cleavage-stage mouse embryogenesis. Proc Natl Acad Sci U S A 2009, 106:7473-7478.
  • [5]Dean J: Oocyte-specific genes regulate follicle formation, fertility and early mouse development. J Reprod Immunol 2002, 53:171-180.
  • [6]Zheng P, Dean J: Oocyte-specific genes affect folliculogenesis, fertilization, and early development. Semin Reprod Med 2007, 25:243-251.
  • [7]Wu X, Viveiros MM, Eppig JJ, Bai Y, Fitzpatrick SL, Matzuk MM: Zygote arrest 1 (Zar1) is a novel maternal-effect gene critical for the oocyte-to-embryo transition. Nat Genet 2003, 33:187-191.
  • [8]Tong ZB, Nelson LM: A mouse gene encoding an oocyte antigen associated with autoimmune premature ovarian failure. Endocrinology 1999, 140:3720-3726.
  • [9]Gilbert SF: Developmental biology. 6th edition. Sinauer Associates, Sunderland MA; 2000.
  • [10]Evsikov AV, Graber JH, Brockman JM, Hampl A, Holbrook AE, Singh P, Eppig JJ, Solter D, Knowles BB: Cracking the egg: molecular dynamics and evolutionary aspects of the transition from the fully grown oocyte to embryo. Genes Dev 2006, 20:2713-2727.
  • [11]Vallee M, Robert C, Methot S, Palin MF, Sirard MA: Cross-species hybridizations on a multi-species cDNA microarray to identify evolutionarily conserved genes expressed in oocytes. BMC Genomics 2006, 7:113. BioMed Central Full Text
  • [12]Vallee M, Aiba K, Piao Y, Palin MF, Ko MS, Sirard MA: Comparative analysis of oocyte transcript profiles reveals a high degree of conservation among species. Reproduction 2008, 135:439-448.
  • [13]Telford NA, Watson AJ, Schultz GA: Transition from maternal to embryonic control in early mammalian development: a comparison of several species. Mol Reprod Dev 1999, 26:90-100.
  • [14]Bettegowda A, Yao J, Sen A, Li Q, Lee KB, Kobayashi Y, Patel OV, Coussens PM, Ireland JJ, Smith GW: JY-1, an oocyte-specific gene, regulates granulosa cell function and early embryonic development in cattle. Proc Natl Acad Sci U S A 2007, 104:17602-17607.
  • [15]Hanrahan JP, Gregan SM, Mulsant P, Mullen M, Davis GH, Powell R, Galloway SM: Mutations in the genes for oocyte-derived growth factors GDF9 and BMP15 are associated with both increased ovulation rate and sterility in Cambridge and Belclare sheep (Ovis aries). Biol Reprod 2004, 70:900-909.
  • [16]Moore RK, Erickson GF, Shimasaki S: Are BMP-15 and GDF-9 primary determinants of ovulation quota in mammals? Trends Endocrinol Metab 2004, 15:356-361.
  • [17]Galloway SM, McNatty KP, Cambridge LM, Laitinen MP, Juengel JL, Jokiranta TS, McLaren RJ, Luiro K, Dodds KG, Montgomery GW, Beattie AE, Davis GH, Ritvos O: Mutations in an oocyte-derived growth factor gene (BMP15) cause increased ovulation rate and infertility in a dosage-sensitive manner. Nat Genet 2000, 25:279-283.
  • [18]Galloway SM, Gregan SM, Wilson T, McNatty KP, Juengel JL, Ritvos O, Davis GH: Bmp15 mutations and ovarian function. Mol Cell Endocrinol 2002, 191:15-18.
  • [19]Yan C, Wang P, DeMayo J, DeMayo FJ, Elvin JA, Carino C, Prasad SV, Skinner SS, Dunbar BS, Dube JL, Celeste AJ, Matzuk MM: Synergistic roles of bone morphogenetic protein 15 and growth differentiation factor 9 in ovarian function. Mol Endocrinol 2001, 15:854-866.
  • [20]Dade S, Callebaut I, Mermillod P, Monget P: Identification of a new expanding family of genes characterized by atypical LRR domains: localization of a cluster preferentially expressed in oocyte. FEBS Lett 2003, 555:533-538.
  • [21]Dade S, Callebaut I, Paillisson A, Bontoux M, Dalbies-Tran R, Monget P: In silico identification and structural features of six new genes similar to MATER specifically expressed in the oocyte. Biochem Biophys Res Commun 2004, 324:547-553.
  • [22]Paillisson A, Dade S, Callebaut I, Bontoux M, Dalbies-Tran R, Vaiman D, Monget P: Identification, characterization and metagenome analysis of oocyte-specific genes organized in clusters in the mouse genome. BMC Genomics 2005, 6:76. BioMed Central Full Text
  • [23]Swanson WJ, Vacquier VD: The rapid evolution of reproductive proteins. Nat Rev Genet 2002, 3:137-144.
  • [24]Bebbere D, Bogliolo L, Ariu F, Fois S, Leoni GG, Tore S, Succu S, Berlinguer F, Naitana S, Ledda S: Expression pattern of zygote arrest 1 (ZAR1), maternal antigen that embryo requires (MATER), growth differentiation factor 9 (GDF9) and bone morphogenetic protein 15 (BMP15) genes in ovine oocytes and in vitro-produced preimplantation embryos. Reprod Fertil Dev 2008, 20:908-915.
  • [25]Pisani LF, Ramelli P, Lazzari B, Braglia S, Ceciliani F, Mariani P: Characterization of maternal antigen that embryos require (MATER/NLRP5) gene and protein in pig somatic tissues and germ cells. J Reprod Dev 2010, 56:41-48.
  • [26]Zhang P, Dixon M, Zucchelli M, Hambiliki F, Levkov L, Hovatta O, Kere J: Expression analysis of the NLRP gene family suggests a role in human preimplantation development. PLoS One 2008, 3:e2755.
  • [27]Pierre A, Gautier M, Callebaut I, Bontoux M, Jeanpierre E, Pontarotti P, Monget P: Atypical structure and phylogenomic evolution of the new eutherian oocyte- and embryo-expressed KHDC1/DPPA5/ECAT1/OOEP gene family. Genomics 2007, 90:583-594.
  • [28]Tervit HR, Whittingham DG, Rowson LE: Successful culture in vitro of sheep and cattle ova. J Reprod Fertil 1972, 30:493-497.
  • [29]Walker SK, Hill JL, Kleemann DO, Nancarrow CD: Development of ovine embryos in synthetic oviductal fluid containing amino acids at oviductal fluid concentrations. Biol Reprod 1996, 55:703-708.
  • [30]Bogliolo L, Ariu F, Leoni G, Uccheddu S, Bebbere D: High hydrostatic pressure treatment improves the quality of in vitro-produced ovine blastocysts. Reprod Fertil Dev 2011, 23:809-817.
  • [31]Evsikov AV, Marín de Evsikova C: Gene expression during the oocyte-to-embryo transition in mammals. Mol Reprod Dev 2009, 76:805-818.
  • [32]Su YQ, Sugiura K, Woo Y, Wigglesworth K, Kamdar S, Affourtit J, Eppig JJ: Selective degradation of transcripts during meiotic maturation of mouse oocytes. Dev Biol 2007, 302:104-117.
  • [33]Ohsugi M, Zheng P, Baibakov B, Li L, Dean J: Maternally derived FILIA-MATER complex localizes asymmetrically in cleavage-stage mouse embryos. Development 2008, 135:259-269.
  • [34]Armstrong DT, Holm P, Irvine B, Petersen BA, Stubbings RB, McLean D, Stevens G, Seamark RF: Pregnancies and live birth from in vitro fertilization of calf oocytes collected by laparoscopic follicular aspiration. Theriogenology 1992, 38:667-678.
  • [35]Armstrong DT, Kotaras PJ, Earl CR: Advances in production of embryos in vitro from juvenile and prepubertal oocytes from the calf and lamb. Reprod Fertil Dev 1997, 9:333-339.
  • [36]Ledda S, Bogliolo L, Leoni G, Naitana S: Production and lambing rate of blastocysts derived from in vitro matured oocytes after gonadotrophin treatment of prepubertal ewes. J Anim Sci 1999, 77:2234-2239.
  • [37]Eppig JJ: Coordination of nuclear and cytoplasmic oocyte maturation in eutherian mammals. Reprod Fertil Dev 1996, 8:485-489.
  • [38]Leoni GG, Bebbere D, Succu S, Berlinguer F, Mossa F, Galioto M, Bogliolo L, Ledda S, Naitana S: Relations between relative mRNA abundance and developmental competence of ovine oocytes. Mol Reprod Dev 2007, 74:249-257.
  • [39]Walser CB, Lipshitz HD: Transcript clearance during the maternal-to-zygotic transition. Curr Opin Genet Dev 2011, 21:431-443.
  • [40]Magen A, Ast G: The importance of being divisible by three in alternative splicing. Nucleic Acids Res 2005, 33:5574-5582.
  • [41]Maquat LE: Nonsense-mediated mRNA decay: splicing, translation and mRNP dynamics. Nat Rev Mol Cell Biol 2004, 5:89-99.
  • [42]Lewis BP, Green RE, Brenner SE: Evidence for the widespread coupling of alternative splicing and nonsense-mediated mRNA decay in humans. Proc Natl Acad Sci U S A 2003, 100:189-192.
  • [43]Lareau LF, Green RE, Bhatnagar RS, Brenner SE: The evolving roles of alternative splicing. Curr Opin Struct Biol 2004, 14:273-282.
  • [44]Hillman RT, Green RE, Brenner SE: An unappreciated role for RNA surveillance. Genome Biol 2004, 5:R8. BioMed Central Full Text
  • [45]Wang J, Xu M, Zhu K, Li L, Liu X: The N-terminus of FILIA forms an atypical KH domain with a unique extension involved in interaction with RNA. PLoS One 2012, 7:e30209.
  • [46]Van den Veyver IB, Al-Hussaini TK: Biparental hydatidiform moles: a maternal effect mutation affecting imprinting in the offspring. Hum Reprod Update 2006, 12:233-242.
  • [47]Parry DA, Logan CV, Hayward BE, Shires M, Landolsi H, Diggle C, Carr I, Rittore C, Touitou I, Philibert L, Fisher RA, Fallahian M, Huntriss JD, Picton HM, Malik S, Taylor GR, Johnson CA, Bonthron DT, Sheridan EG: Mutations causing familial biparental hydatidiform mole implicate C6orf221 as a possible regulator of genomic imprinting in the human oocyte. Am J Hum Genet 2011, 89:451-458.
  • [48]Fallahian M, Sebire NJ, Savage PM, Seckl MJ, Fisher RA: Mutations in NLRP7 and KHDC3L confer a complete hydatidiform mole phenotype on digynic triploid conceptions. Hum Mutat 2013, 34:301-308.
  • [49]Reddy R, Akoury E, Phuong Nguyen NM, Abdul-Rahman OA, Dery C, Gupta N, Daley WP, Ao A, Landolsi H, Ann Fisher R, Touitou I, Slim R: Report of four new patients with protein-truncating mutations in C6orf221/KHDC3L and colocalization with NLRP7. Eur J Hum Genet 2013, 21:957-964.
  • [50]Andreasen L, Bolund L, Niemann I, Hansen ES, Sunde L: Mosaic moles and non-familial biparental moles are not caused by mutations in NLRP7, NLRP2 or C6orf221. Mol Hum Reprod 2012, 18:593-598.
  • [51]Manokhina I, Hanna CW, Stephenson MD, McFadden DE, Robinson WP: Maternal NLRP7 and C6orf221 variants are not a common risk factor for androgenetic moles, triploidy and recurrent miscarriage. Mol Hum Reprod 2013, 19:539-544.
  • [52]Pennetier S, Uzbekova S, Perreau C, Papillier P, Mermillod P, Dalbiès-Tran R: Spatio-temporal expression of the germ cell marker genes MATER, ZAR1, GDF9, BMP15, and VASA in adult bovine tissues, oocytes, and preimplantation embryos. Biol Reprod 2004, 71:1359-1366.
  • [53]Leader B, Lim H, Carabatsos MJ, Harrington A, Ecsedy J, Pellman D, Maas R, Leder P: Formin-2, polyploidy, hypofertility and positioning of the meiotic spindle in mouse oocytes. Nat Cell Biol 2002, 4:921-928.
  • [54]Leader B, Leder P: Formin-2, a novel formin homology protein of the cappuccino subfamily, is highly expressed in the developing and adult central nervous system. Mech Dev 2000, 93:221-231.
  文献评价指标  
  下载次数:0次 浏览次数:2次