Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes

Dankert D, Demond H, Trapphoff T, Heiligentag M, Rademacher K, Eichenlaub-Ritter U, Horsthemke B, Gruemmer R (2014)
PLoS ONE 9(10): e108907.

Zeitschriftenaufsatz | Veröffentlicht | Englisch
 
Download
Es wurden keine Dateien hochgeladen. Nur Publikationsnachweis!
Autor*in
Dankert, Debora; Demond, Hannah; Trapphoff, Tom; Heiligentag, Martyna; Rademacher, Katrin; Eichenlaub-Ritter, UrsulaUniBi; Horsthemke, Bernhard; Gruemmer, Ruth
Abstract / Bemerkung
Maternal effect genes code for oocyte proteins that are important for early embryogenesis. Transcription in oocytes does not take place from the onset of meiotic progression until zygotic genome activation. During this period, protein levels are regulated posttranscriptionally, for example by poly(A) tail length. Posttranscriptional regulation may be impaired in preovulatory and postovulatory aged oocytes, caused by delayed ovulation or delayed fertilization, respectively, and may lead to developmental defects. We investigated transcript levels and poly(A) tail length of ten maternal effect genes in in vivo-and in vitro- (follicle culture) grown oocytes after pre- and postovulatory aging. Quantitative RT-PCR was performed using random hexamer-primed cDNA to determine total transcript levels and oligo(dT)(16)-primed cDNA to analyze poly(A) tail length. Transcript levels of in vivo preovulatory-aged oocytes remained stable except for decreases in Brg1 and Tet3. Most genes investigated showed a tendency towards increased poly(A) content. Polyadenylation of in vitro preovulatory-aged oocytes was also increased, along with transcript level declines of Trim28, Nlrp2, Nlrp14 and Zar1. In contrast to preovulatory aging, postovulatory aging of in vivo-and in vitro-grown oocytes led to a shortening of poly(A) tails. Postovulatory aging of in vivo-grown oocytes resulted in deadenylation of Nlrp5 after 12 h, and deadenylation of 4 further genes (Tet3, Trim28, Dnmt1, Oct4) after 24 h. Similarly, transcripts of in vitro-grown oocytes were deadenylated after 12 h of postovulatory aging (Tet3, Trim28, Zfp57, Dnmt1, Nlrp5, Zar1). This impact of aging on poly(A) tail length may affect the timed translation of maternal effect gene transcripts and thereby contribute to developmental defects.
Erscheinungsjahr
2014
Zeitschriftentitel
PLoS ONE
Band
9
Ausgabe
10
Art.-Nr.
e108907
ISSN
1932-6203
eISSN
1932-6203
Page URI
https://pub.uni-bielefeld.de/record/2707906

Zitieren

Dankert D, Demond H, Trapphoff T, et al. Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes. PLoS ONE. 2014;9(10): e108907.
Dankert, D., Demond, H., Trapphoff, T., Heiligentag, M., Rademacher, K., Eichenlaub-Ritter, U., Horsthemke, B., et al. (2014). Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes. PLoS ONE, 9(10), e108907. doi:10.1371/journal.pone.0108907
Dankert, Debora, Demond, Hannah, Trapphoff, Tom, Heiligentag, Martyna, Rademacher, Katrin, Eichenlaub-Ritter, Ursula, Horsthemke, Bernhard, and Gruemmer, Ruth. 2014. “Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes”. PLoS ONE 9 (10): e108907.
Dankert, D., Demond, H., Trapphoff, T., Heiligentag, M., Rademacher, K., Eichenlaub-Ritter, U., Horsthemke, B., and Gruemmer, R. (2014). Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes. PLoS ONE 9:e108907.
Dankert, D., et al., 2014. Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes. PLoS ONE, 9(10): e108907.
D. Dankert, et al., “Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes”, PLoS ONE, vol. 9, 2014, : e108907.
Dankert, D., Demond, H., Trapphoff, T., Heiligentag, M., Rademacher, K., Eichenlaub-Ritter, U., Horsthemke, B., Gruemmer, R.: Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes. PLoS ONE. 9, : e108907 (2014).
Dankert, Debora, Demond, Hannah, Trapphoff, Tom, Heiligentag, Martyna, Rademacher, Katrin, Eichenlaub-Ritter, Ursula, Horsthemke, Bernhard, and Gruemmer, Ruth. “Pre- and Postovulatory Aging of Murine Oocytes Affect the Transcript Level and Poly(A) Tail Length of Maternal Effect Genes”. PLoS ONE 9.10 (2014): e108907.

11 Zitationen in Europe PMC

Daten bereitgestellt von Europe PubMed Central.

Genetic diagnosis of subfertility: the impact of meiosis and maternal effects.
Gheldof A, Mackay DJG, Cheong Y, Verpoest W., J Med Genet 56(5), 2019
PMID: 30728173
The possible involvement of oxidative stress in the oocyte ageing process in goldfish Carassius auratus (Linnaeus, 1758).
Samarin AM, Samarin AM, Østbye TK, Ruyter B, Sampels S, Burkina V, Blecha M, Policar T., Sci Rep 9(1), 2019
PMID: 31320670
mRNA abundance changes during in vitro oocyte ageing in African catfish Clarias gariepinus (Burchell, 1822)
Samarin AM, Sabine Sampels, Tomas Policar, Marek Rodina, Nima Hematyar, Azin Mohagheghi Samarin., Aquac Res 49(2), 2018
PMID: IND605885002
Maternal variants in NLRP and other maternal effect proteins are associated with multilocus imprinting disturbance in offspring.
Begemann M, Rezwan FI, Beygo J, Docherty LE, Kolarova J, Schroeder C, Buiting K, Chokkalingam K, Degenhardt F, Wakeling EL, Kleinle S, González Fassrainer D, Oehl-Jaschkowitz B, Turner CLS, Patalan M, Gizewska M, Binder G, Bich Ngoc CT, Chi Dung V, Mehta SG, Baynam G, Hamilton-Shield JP, Aljareh S, Lokulo-Sodipe O, Horton R, Siebert R, Elbracht M, Temple IK, Eggermann T, Mackay DJG., J Med Genet 55(7), 2018
PMID: 29574422
Maternal RNA regulates Aurora C kinase during mouse oocyte maturation in a translation-independent fashion.
Balboula AZ, Blengini CS, Gentilello AS, Takahashi M, Schindler K., Biol Reprod 96(6), 2017
PMID: 28575288
Postovulatory aging affects dynamics of mRNA, expression and localization of maternal effect proteins, spindle integrity and pericentromeric proteins in mouse oocytes.
Trapphoff T, Heiligentag M, Dankert D, Demond H, Deutsch D, Fröhlich T, Arnold GJ, Grümmer R, Horsthemke B, Eichenlaub-Ritter U., Hum Reprod 31(1), 2016
PMID: 26577303
Cloning of Porcine Pituitary Tumor Transforming Gene 1 and Its Expression in Porcine Oocytes and Embryos.
Xie B, Qin Z, Liu S, Nong S, Ma Q, Chen B, Liu M, Pan T, Liao DJ., PLoS One 11(4), 2016
PMID: 27058238
The subcortical maternal complex: multiple functions for one biological structure?
Bebbere D, Masala L, Albertini DF, Ledda S., J Assist Reprod Genet 33(11), 2016
PMID: 27525657
Preovulatory Aging In Vivo and In Vitro Affects Maturation Rates, Abundance of Selected Proteins, Histone Methylation Pattern and Spindle Integrity in Murine Oocytes.
Demond H, Trapphoff T, Dankert D, Heiligentag M, Grümmer R, Horsthemke B, Eichenlaub-Ritter U., PLoS One 11(9), 2016
PMID: 27611906
Mutations in NLRP5 are associated with reproductive wastage and multilocus imprinting disorders in humans.
Docherty LE, Rezwan FI, Poole RL, Turner CL, Kivuva E, Maher ER, Smithson SF, Hamilton-Shield JP, Patalan M, Gizewska M, Peregud-Pogorzelski J, Beygo J, Buiting K, Horsthemke B, Soellner L, Begemann M, Eggermann T, Baple E, Mansour S, Temple IK, Mackay DJ., Nat Commun 6(), 2015
PMID: 26323243

65 References

Daten bereitgestellt von Europe PubMed Central.


AUTHOR UNKNOWN, 0
Translational control by changes in poly(A) tail length: recycling mRNAs.
Weill L, Belloc E, Bava FA, Mendez R., Nat. Struct. Mol. Biol. 19(6), 2012
PMID: 22664985
Epigenetic reprogramming in early mammalian development and following somatic nuclear transfer.
Dean W, Santos F, Reik W., Semin. Cell Dev. Biol. 14(1), 2003
PMID: 12524012
The role of Tet3 DNA dioxygenase in epigenetic reprogramming by oocytes.
Gu TP, Guo F, Yang H, Wu HP, Xu GF, Liu W, Xie ZG, Shi L, He X, Jin SG, Iqbal K, Shi YG, Deng Z, Szabo PE, Pfeifer GP, Li J, Xu GL., Nature 477(7366), 2011
PMID: 21892189
Maternal BRG1 regulates zygotic genome activation in the mouse.
Bultman SJ, Gebuhr TC, Pan H, Svoboda P, Schultz RM, Magnuson T., Genes Dev. 20(13), 2006
PMID: 16818606
Trim28 is required for epigenetic stability during mouse oocyte to embryo transition.
Messerschmidt DM, de Vries W, Ito M, Solter D, Ferguson-Smith A, Knowles BB., Science 335(6075), 2012
PMID: 22442485
Selective degradation of transcripts during meiotic maturation of mouse oocytes.
Su YQ, Sugiura K, Woo Y, Wigglesworth K, Kamdar S, Affourtit J, Eppig JJ., Dev. Biol. 302(1), 2006
PMID: 17022963
Polyadenylated RNA of mouse ova and loss of maternal RNA in early development.
Bachvarova R, De Leon V., Dev. Biol. 74(1), 1980
PMID: 7350004
Maternal control of early mouse development.
Li L, Zheng P, Dean J., Development 137(6), 2010
PMID: 20179092
Preovulatory overripeness of the oocyte as a cause of ovarian dysfunction in the human female.
Smits LJ, Jongbloet PH, Zielhuis GA., Med. Hypotheses 45(5), 1995
PMID: 8748083
Hormone-induced delayed ovulation affects early embryonic development.
Bittner AK, Horsthemke B, Winterhager E, Grummer R., Fertil. Steril. 95(7), 2011
PMID: 21474126
Versuche der Befruchtung überreifer Eier
AUTHOR UNKNOWN, 1882
The effects of delayed fertilization on the development of the guinea pig ovum
AUTHOR UNKNOWN, 1939
The effect of delayed fertilization on the development of the rat ovum
AUTHOR UNKNOWN, 1941
OVUM AGE AND THE COURSE OF GESTATION IN THE GUINEA PIG.
Patek AJ Jr, Taylor FH., Science 84(2177), 1936
PMID: 17742899
An experimental analysis of female sterility in the rabbit.
CHANG MC., Fertil. Steril. 3(3), 1952
PMID: 14945538
Long-term effects of postovulatory aging of mouse oocytes on offspring: a two-generational study.
Tarin JJ, Perez-Albala S, Aguilar A, Minarro J, Hermenegildo C, Cano A., Biol. Reprod. 61(5), 1999
PMID: 10529284
Contrast in levels of metabolic enzymes in human and mouse ova.
Chi MM, Manchester JK, Yang VC, Curato AD, Strickler RC, Lowry OH., Biol. Reprod. 39(2), 1988
PMID: 2902884
Loss of methylation imprint of Snrpn in postovulatory aging mouse oocyte.
Liang XW, Zhu JQ, Miao YL, Liu JH, Wei L, Lu SS, Hou Y, Schatten H, Lu KH, Sun QY., Biochem. Biophys. Res. Commun. 371(1), 2008
PMID: 18381202
Oocyte aging: cellular and molecular changes, developmental potential and reversal possibility.
Miao YL, Kikuchi K, Sun QY, Schatten H., Hum. Reprod. Update 15(5), 2009
PMID: 19429634
Association between nondisjunction and maternal age in meiosis-II human oocytes.
Dailey T, Dale B, Cohen J, Munne S., Am. J. Hum. Genet. 59(1), 1996
PMID: 8659524

AUTHOR UNKNOWN, 0
Aging of Xenopus tropicalis eggs leads to deadenylation of a specific set of maternal mRNAs and loss of developmental potential.
Kosubek A, Klein-Hitpass L, Rademacher K, Horsthemke B, Ryffel GU., PLoS ONE 5(10), 2010
PMID: 21042572
Effects of low O2 and ageing on spindles and chromosomes in mouse oocytes from pre-antral follicle culture.
Hu Y, Betzendahl I, Cortvrindt R, Smitz J, Eichenlaub-Ritter U., Hum. Reprod. 16(4), 2001
PMID: 11278227
Acquisition and loss of oocyte meiotic and developmental competence during in vitro antral follicle growth in mouse.
Segers I, Adriaenssens T, Ozturk E, Smitz J., Fertil. Steril. 93(8), 2010
PMID: 20056201
Bidirectional communication between oocytes and ovarian follicular somatic cells is required for meiotic arrest of mammalian oocytes.
Wigglesworth K, Lee KB, O'Brien MJ, Peng J, Matzuk MM, Eppig JJ., Proc. Natl. Acad. Sci. U.S.A. 110(39), 2013
PMID: 23980176
Proposal for a classification of oocytes and follicles in the mouse ovary.
Pedersen T, Peters H., J. Reprod. Fertil. 17(3), 1968
PMID: 5715685
Differential regulation of abundance and deadenylation of maternal transcripts during bovine oocyte maturation in vitro and in vivo.
Thelie A, Papillier P, Pennetier S, Perreau C, Traverso JM, Uzbekova S, Mermillod P, Joly C, Humblot P, Dalbies-Tran R., BMC Dev. Biol. 7(), 2007
PMID: 17988387
ePAT: a simple method to tag adenylated RNA to measure poly(A)-tail length and other 3' RACE applications.
Janicke A, Vancuylenberg J, Boag PR, Traven A, Beilharz TH., RNA 18(6), 2012
PMID: 22543866
A maternal-zygotic effect gene, Zfp57, maintains both maternal and paternal imprints.
Li X, Ito M, Zhou F, Youngson N, Zuo X, Leder P, Ferguson-Smith AC., Dev. Cell 15(4), 2008
PMID: 18854139
Evolution and functional divergence of NLRP genes in mammalian reproductive systems.
Tian X, Pascal G, Monget P., BMC Evol. Biol. 9(), 2009
PMID: 19682372
Nlrp2, a maternal effect gene required for early embryonic development in the mouse.
Peng H, Chang B, Lu C, Su J, Wu Y, Lv P, Wang Y, Liu J, Zhang B, Quan F, Guo Z, Zhang Y., PLoS ONE 7(1), 2012
PMID: 22295082
Germline mutation in NLRP2 (NALP2) in a familial imprinting disorder (Beckwith-Wiedemann Syndrome).
Meyer E, Lim D, Pasha S, Tee LJ, Rahman F, Yates JR, Woods CG, Reik W, Maher ER., PLoS Genet. 5(3), 2009
PMID: 19300480
Mutations in NALP7 cause recurrent hydatidiform moles and reproductive wastage in humans.
Murdoch S, Djuric U, Mazhar B, Seoud M, Khan R, Kuick R, Bagga R, Kircheisen R, Ao A, Ratti B, Hanash S, Rouleau GA, Slim R., Nat. Genet. 38(3), 2006
PMID: 16462743
Zygote arrest 1 (Zar1) is a novel maternal-effect gene critical for the oocyte-to-embryo transition.
Wu X, Viveiros MM, Eppig JJ, Bai Y, Fitzpatrick SL, Matzuk MM., Nat. Genet. 33(2), 2003
PMID: 12539046
Development potential of bovine oocytes matured in vitro or in vivo.
Leibfried-Rutledge ML, Critser ES, Eyestone WH, Northey DL, First NL., Biol. Reprod. 36(2), 1987
PMID: 3580458
Estrogen is a critical determinant that specifies the duration of the window of uterine receptivity for implantation.
Ma WG, Song H, Das SK, Paria BC, Dey SK., Proc. Natl. Acad. Sci. U.S.A. 100(5), 2003
PMID: 12601161
Cumulus cells accelerate aging of mouse oocytes.
Miao YL, Liu XY, Qiao TW, Miao DQ, Luo MJ, Tan JH., Biol. Reprod. 73(5), 2005
PMID: 15987819
Cumulus cells accelerate aging of mouse oocytes by secreting a soluble factor(s).
Qiao TW, Liu N, Miao DQ, Zhang X, Han D, Ge L, Tan JH., Mol. Reprod. Dev. 75(3), 2008
PMID: 17886273
Pyruvate prevents aging of mouse oocytes.
Liu N, Wu YG, Lan GC, Sui HS, Ge L, Wang JZ, Liu Y, Qiao TW, Tan JH., Reproduction 138(2), 2009
PMID: 19465488
Mater, a maternal effect gene required for early embryonic development in mice.
Tong ZB, Gold L, Pfeifer KE, Dorward H, Lee E, Bondy CA, Dean J, Nelson LM., Nat. Genet. 26(3), 2000
PMID: 11062459
NLRP5 mediates mitochondrial function in mouse oocytes and embryos.
Fernandes R, Tsuda C, Perumalsamy AL, Naranian T, Chong J, Acton BM, Tong ZB, Nelson LM, Jurisicova A., Biol. Reprod. 86(5), 2012
PMID: 22357545
The role of MATER in endoplasmic reticulum distribution and calcium homeostasis in mouse oocytes.
Kim B, Zhang X, Kan R, Cohen R, Mukai C, Travis AJ, Coonrod SA., Dev. Biol. 386(2), 2013
PMID: 24374158
Genomic imprinting disrupted by a maternal effect mutation in the Dnmt1 gene.
Howell CY, Bestor TH, Ding F, Latham KE, Mertineit C, Trasler JM, Chaillet JR., Cell 104(6), 2001
PMID: 11290321
Maternal and zygotic Dnmt1 are necessary and sufficient for the maintenance of DNA methylation imprints during preimplantation development.
Hirasawa R, Chiba H, Kaneda M, Tajima S, Li E, Jaenisch R, Sasaki H., Genes Dev. 22(12), 2008
PMID: 18559477
Postovulatory aging of oocytes decreases reproductive fitness and longevity of offspring.
Tarin JJ, Perez-Albala S, Perez-Hoyos S, Cano A., Biol. Reprod. 66(2), 2002
PMID: 11804967
Export

Markieren/ Markierung löschen
Markierte Publikationen

Open Data PUB

Web of Science

Dieser Datensatz im Web of Science®
Quellen

PMID: 25271735
PubMed | Europe PMC

Suchen in

Google Scholar