Skip to main content
Top
Published in: Journal of Assisted Reproduction and Genetics 8/2017

01-08-2017 | Gamete Biology

Transporting cumulus complexes using novel meiotic arresting conditions permits maintenance of oocyte developmental competence

Authors: Nicolas W. Santiquet, Jason R. Herrick, Angelica Giraldo, Jennifer P. Barfield, William B. Schoolcraft, Rebecca L. Krisher

Published in: Journal of Assisted Reproduction and Genetics | Issue 8/2017

Login to get access

Abstract

Purpose

The aim of this study is to evaluate the effect of a novel bovine cumulus oocyte complex (COC) shipping media designed to arrest meiotic resumption during transport on meiotic arrest, as well as meiotic resumption, subsequent embryonic development, and embryo quality.

Methods

Bovine cumulus oocyte complexes were transported overnight from the collection facility to the laboratory. COCs were placed in control in vitro maturation (IVM) or in shipping arrest medium (SAM) containing multiple meiotic inhibitors, and then shipped to our laboratory. Upon arrival, meiotic status was assessed, control COCs were inseminated, and arrested COCs were matured and inseminated the next day. Embryonic development and quality were analyzed.

Results

When bovine COC arrived at the laboratory after overnight shipment (21 h) in SAM, the majority of oocytes remained at the GV stage (75.6 ± 2.9% GV). Arrested oocytes successfully resumed and completed meiosis during IVM after removal from SAM (96.8 ± 0.5% metaphase II compared to control 88.3 ± 5.0%). Moreover, the development of blastocysts per COC was not different from control (22.3 ± 2.4% for control and 18.7 ± 2.1% for SAM), nor was any difference detected in blastocyst quality as determined by cell number and allocation.

Conclusions

Our study demonstrates that a physiological system incorporating cyclic adenosine monophosphate and cyclic guanosine monophosphate modulators can be used to maintain meiotic arrest followed by successful nuclear maturation and pre-implantation embryo development equal to control IVM-derived embryos. Our results offer promising insights for the development of pre-IVM media that may improve oocyte developmental competence in vitro.
Literature
1.
go back to reference Pincus G, Enzmann EV. The comparative behavior of mammalian eggs in vivo and in vitro: I. The activation of ovarian eggs. J Exp Med. 1935;62(5):665–75.CrossRefPubMedPubMedCentral Pincus G, Enzmann EV. The comparative behavior of mammalian eggs in vivo and in vitro: I. The activation of ovarian eggs. J Exp Med. 1935;62(5):665–75.CrossRefPubMedPubMedCentral
2.
go back to reference ASRM. In vitro maturation: a committee opinion. Fertil Steril. 2013;99(3):663–6.CrossRef ASRM. In vitro maturation: a committee opinion. Fertil Steril. 2013;99(3):663–6.CrossRef
3.
go back to reference Gremeau AS, Andreadis N, Fatum M, Craig J, Turner K, McVeigh E, et al. In vitro maturation or in vitro fertilization for women with polycystic ovaries? A case-control study of 194 treatment cycles. Fertil Steril. 2012;98(2):355–60.CrossRefPubMed Gremeau AS, Andreadis N, Fatum M, Craig J, Turner K, McVeigh E, et al. In vitro maturation or in vitro fertilization for women with polycystic ovaries? A case-control study of 194 treatment cycles. Fertil Steril. 2012;98(2):355–60.CrossRefPubMed
4.
go back to reference Wrenzycki C, Stinshoff H. Maturation environment and impact on subsequent developmental competence of bovine oocytes. Reproduction in domestic animals = Zuchthygiene. 2013;48(Suppl 1):38–43.CrossRefPubMed Wrenzycki C, Stinshoff H. Maturation environment and impact on subsequent developmental competence of bovine oocytes. Reproduction in domestic animals = Zuchthygiene. 2013;48(Suppl 1):38–43.CrossRefPubMed
5.
go back to reference Gilchrist RB, Thompson JG. Oocyte maturation: emerging concepts and technologies to improve developmental potential in vitro. Theriogenology. 2007;67(1):6–15.CrossRefPubMed Gilchrist RB, Thompson JG. Oocyte maturation: emerging concepts and technologies to improve developmental potential in vitro. Theriogenology. 2007;67(1):6–15.CrossRefPubMed
6.
go back to reference Eppig JJ, O’Brien MJ, Wigglesworth K, Nicholson A, Zhang W, King BA. Effect of in vitro maturation of mouse oocytes on the health and lifespan of adult offspring. Hum Reprod. 2009;24(4):922–8.CrossRefPubMedPubMedCentral Eppig JJ, O’Brien MJ, Wigglesworth K, Nicholson A, Zhang W, King BA. Effect of in vitro maturation of mouse oocytes on the health and lifespan of adult offspring. Hum Reprod. 2009;24(4):922–8.CrossRefPubMedPubMedCentral
7.
go back to reference Gilchrist RB. Recent insights into oocyte-follicle cell interactions provide opportunities for the development of new approaches to in vitro maturation. Reprod Fertil Dev. 2011;23(1):23–31.CrossRefPubMed Gilchrist RB. Recent insights into oocyte-follicle cell interactions provide opportunities for the development of new approaches to in vitro maturation. Reprod Fertil Dev. 2011;23(1):23–31.CrossRefPubMed
8.
go back to reference Edwards RG. Are minimal stimulation IVF and IVM set to replace routine IVF? Reprod BioMed Online. 2007;14(2):267–70.CrossRefPubMed Edwards RG. Are minimal stimulation IVF and IVM set to replace routine IVF? Reprod BioMed Online. 2007;14(2):267–70.CrossRefPubMed
9.
go back to reference Rose BI, Laky D, Miller B. The case for in vitro maturation lower cost and more patient friendly. J Reprod Med. 2014;59(11–12):571–8.PubMed Rose BI, Laky D, Miller B. The case for in vitro maturation lower cost and more patient friendly. J Reprod Med. 2014;59(11–12):571–8.PubMed
10.
go back to reference Smitz JE, Thompson JG, Gilchrist RB. The promise of in vitro maturation in assisted reproduction and fertility preservation. Semin Reprod Med. 2011;29(1):24–37.CrossRefPubMed Smitz JE, Thompson JG, Gilchrist RB. The promise of in vitro maturation in assisted reproduction and fertility preservation. Semin Reprod Med. 2011;29(1):24–37.CrossRefPubMed
11.
go back to reference Walls M, Junk S, Ryan JP, Hart R. IVF versus ICSI for the fertilization of in-vitro matured human oocytes. Reprod BioMed Online. 2012;25(6):603–7.CrossRefPubMed Walls M, Junk S, Ryan JP, Hart R. IVF versus ICSI for the fertilization of in-vitro matured human oocytes. Reprod BioMed Online. 2012;25(6):603–7.CrossRefPubMed
12.
go back to reference Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC, Brambillasca F, Turchi D, et al. Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update. 2015;21(4):427–54.CrossRefPubMed Coticchio G, Dal Canto M, Mignini Renzini M, Guglielmo MC, Brambillasca F, Turchi D, et al. Oocyte maturation: gamete-somatic cells interactions, meiotic resumption, cytoskeletal dynamics and cytoplasmic reorganization. Hum Reprod Update. 2015;21(4):427–54.CrossRefPubMed
13.
go back to reference Li R, Albertini DF. The road to maturation: somatic cell interaction and self-organization of the mammalian oocyte. Nat Rev Mol Cell Biol. 2013;14(3):141–52.CrossRefPubMed Li R, Albertini DF. The road to maturation: somatic cell interaction and self-organization of the mammalian oocyte. Nat Rev Mol Cell Biol. 2013;14(3):141–52.CrossRefPubMed
14.
go back to reference Albuz FK, Sasseville M, Lane M, Armstrong DT, Thompson JG, Gilchrist RB. Simulated physiological oocyte maturation (SPOM): a novel in vitro maturation system that substantially improves embryo yield and pregnancy outcomes. Hum Reprod. 2010;25(12):2999–3011.CrossRefPubMed Albuz FK, Sasseville M, Lane M, Armstrong DT, Thompson JG, Gilchrist RB. Simulated physiological oocyte maturation (SPOM): a novel in vitro maturation system that substantially improves embryo yield and pregnancy outcomes. Hum Reprod. 2010;25(12):2999–3011.CrossRefPubMed
15.
go back to reference Downs SM, Schroeder AC, Eppig JJ. Developmental capacity of mouse oocytes following maintenance of meiotic arrest in vitro. Gamete research. 1986;15(4):305–16.CrossRef Downs SM, Schroeder AC, Eppig JJ. Developmental capacity of mouse oocytes following maintenance of meiotic arrest in vitro. Gamete research. 1986;15(4):305–16.CrossRef
16.
go back to reference Lonergan P, Khatir H, Carolan C, Mermillod P. Bovine blastocyst production in vitro after inhibition of oocyte meiotic resumption for 24 h. J Reprod Fertil. 1997;109(2):355–65.CrossRefPubMed Lonergan P, Khatir H, Carolan C, Mermillod P. Bovine blastocyst production in vitro after inhibition of oocyte meiotic resumption for 24 h. J Reprod Fertil. 1997;109(2):355–65.CrossRefPubMed
17.
go back to reference Luciano AM, Pocar P, Milanesi E, Modina S, Rieger D, Lauria A, et al. Effect of different levels of intracellular cAMP on the in vitro maturation of cattle oocytes and their subsequent development following in vitro fertilization. Mol Reprod Dev. 1999;54(1):86–91.CrossRefPubMed Luciano AM, Pocar P, Milanesi E, Modina S, Rieger D, Lauria A, et al. Effect of different levels of intracellular cAMP on the in vitro maturation of cattle oocytes and their subsequent development following in vitro fertilization. Mol Reprod Dev. 1999;54(1):86–91.CrossRefPubMed
18.
go back to reference Mermillod P, Tomanek M, Marchal R, Meijer L. High developmental competence of cattle oocytes maintained at the germinal vesicle stage for 24 hours in culture by specific inhibition of MPF kinase activity. Mol Reprod Dev. 2000;55(1):89–95.CrossRefPubMed Mermillod P, Tomanek M, Marchal R, Meijer L. High developmental competence of cattle oocytes maintained at the germinal vesicle stage for 24 hours in culture by specific inhibition of MPF kinase activity. Mol Reprod Dev. 2000;55(1):89–95.CrossRefPubMed
19.
go back to reference Nogueira D, Ron-El R, Friedler S, Schachter M, Raziel A, Cortvrindt R, et al. Meiotic arrest in vitro by phosphodiesterase 3-inhibitor enhances maturation capacity of human oocytes and allows subsequent embryonic development. Biol Reprod. 2006;74(1):177–84.CrossRefPubMed Nogueira D, Ron-El R, Friedler S, Schachter M, Raziel A, Cortvrindt R, et al. Meiotic arrest in vitro by phosphodiesterase 3-inhibitor enhances maturation capacity of human oocytes and allows subsequent embryonic development. Biol Reprod. 2006;74(1):177–84.CrossRefPubMed
20.
go back to reference Richani D, Wang X, Zeng HT, Smitz J, Thompson JG, Gilchrist RB. Pre-maturation with cAMP modulators in conjunction with EGF-like peptides during in vitro maturation enhances mouse oocyte developmental competence. Mol Reprod Dev. 2014;81(5):422–35.CrossRefPubMed Richani D, Wang X, Zeng HT, Smitz J, Thompson JG, Gilchrist RB. Pre-maturation with cAMP modulators in conjunction with EGF-like peptides during in vitro maturation enhances mouse oocyte developmental competence. Mol Reprod Dev. 2014;81(5):422–35.CrossRefPubMed
21.
go back to reference Sirard MA, First NL. In vitro inhibition of oocyte nuclear maturation in the bovine. Biol Reprod. 1988;39(2):229–34.CrossRefPubMed Sirard MA, First NL. In vitro inhibition of oocyte nuclear maturation in the bovine. Biol Reprod. 1988;39(2):229–34.CrossRefPubMed
22.
go back to reference Vanhoutte L, De Sutter P, Nogueira D, Gerris J, Dhont M, Van der Elst J. Nuclear and cytoplasmic maturation of in vitro matured human oocytes after temporary nuclear arrest by phosphodiesterase 3-inhibitor. Hum Reprod. 2007;22(5):1239–46.CrossRefPubMed Vanhoutte L, De Sutter P, Nogueira D, Gerris J, Dhont M, Van der Elst J. Nuclear and cytoplasmic maturation of in vitro matured human oocytes after temporary nuclear arrest by phosphodiesterase 3-inhibitor. Hum Reprod. 2007;22(5):1239–46.CrossRefPubMed
23.
go back to reference Wu GM, Sun QY, Mao J, Lai L, McCauley TC, Park KW, et al. High developmental competence of pig oocytes after meiotic inhibition with a specific M-phase promoting factor kinase inhibitor, butyrolactone I. Biol Reprod. 2002;67(1):170–7.CrossRefPubMed Wu GM, Sun QY, Mao J, Lai L, McCauley TC, Park KW, et al. High developmental competence of pig oocytes after meiotic inhibition with a specific M-phase promoting factor kinase inhibitor, butyrolactone I. Biol Reprod. 2002;67(1):170–7.CrossRefPubMed
24.
go back to reference Zeng HT, Richani D, Sutton-McDowall ML, Ren Z, Smitz JE, Stokes Y, et al. Prematuration with cyclic adenosine monophosphate modulators alters cumulus cell and oocyte metabolism and enhances developmental competence of in vitro-matured mouse oocytes. Biol Reprod. 2014;91(2):47.CrossRefPubMed Zeng HT, Richani D, Sutton-McDowall ML, Ren Z, Smitz JE, Stokes Y, et al. Prematuration with cyclic adenosine monophosphate modulators alters cumulus cell and oocyte metabolism and enhances developmental competence of in vitro-matured mouse oocytes. Biol Reprod. 2014;91(2):47.CrossRefPubMed
25.
go back to reference Guimaraes AL, Pereira SA, Leme LO, Dode MA. Evaluation of the simulated physiological oocyte maturation system for improving bovine in vitro embryo production. Theriogenology. 2015;83(1):52–7.CrossRefPubMed Guimaraes AL, Pereira SA, Leme LO, Dode MA. Evaluation of the simulated physiological oocyte maturation system for improving bovine in vitro embryo production. Theriogenology. 2015;83(1):52–7.CrossRefPubMed
26.
go back to reference Gilchrist RB, Zeng HT, Wang X, Richani D, Smitz J, Thompson JG. “Re-evaluation and evolution of the simulated physiological oocyte maturation (SPOM) system.” Theriogenology. 2015;84(4):656–7. Gilchrist RB, Zeng HT, Wang X, Richani D, Smitz J, Thompson JG. “Re-evaluation and evolution of the simulated physiological oocyte maturation (SPOM) system.” Theriogenology. 2015;84(4):656–7.
27.
go back to reference Li HJ, Sutton-McDowall ML, Wang X, Sugimura S, Thompson JG, Gilchrist RB. “Extending prematuration with cAMP modulators enhances the cumulus contribution to oocyte antioxidant defence and oocyte quality via gap junctions.” Hum Reprod. 2016;31(4):810-21. Li HJ, Sutton-McDowall ML, Wang X, Sugimura S, Thompson JG, Gilchrist RB. “Extending prematuration with cAMP modulators enhances the cumulus contribution to oocyte antioxidant defence and oocyte quality via gap junctions.” Hum Reprod. 2016;31(4):810-21.
28.
go back to reference Campen KA, Clark ZL, Olds MA, McNatty KP, Pitman JL. The in-vitro effects of cAMP and cGMP modulators on inter-cellular dye transfer and gene expression levels in rat cumulus cell-oocyte complexes. Mol Cell Endocrinol. 2015;420:46–56.CrossRefPubMed Campen KA, Clark ZL, Olds MA, McNatty KP, Pitman JL. The in-vitro effects of cAMP and cGMP modulators on inter-cellular dye transfer and gene expression levels in rat cumulus cell-oocyte complexes. Mol Cell Endocrinol. 2015;420:46–56.CrossRefPubMed
29.
go back to reference Zhang J, Wei Q, Cai J, Zhao X, Ma B. Effect of C-type natriuretic peptide on maturation and developmental competence of goat oocytes matured in vitro. PLoS One. 2015;10(7):e0132318.CrossRefPubMedPubMedCentral Zhang J, Wei Q, Cai J, Zhao X, Ma B. Effect of C-type natriuretic peptide on maturation and developmental competence of goat oocytes matured in vitro. PLoS One. 2015;10(7):e0132318.CrossRefPubMedPubMedCentral
30.
go back to reference Zhang M, Su YQ, Sugiura K, Wigglesworth K, Xia G, Eppig JJ. Estradiol promotes and maintains cumulus cell expression of natriuretic peptide receptor 2 (NPR2) and meiotic arrest in mouse oocytes in vitro. Endocrinology. 2011;152(11):4377–85.CrossRefPubMedPubMedCentral Zhang M, Su YQ, Sugiura K, Wigglesworth K, Xia G, Eppig JJ. Estradiol promotes and maintains cumulus cell expression of natriuretic peptide receptor 2 (NPR2) and meiotic arrest in mouse oocytes in vitro. Endocrinology. 2011;152(11):4377–85.CrossRefPubMedPubMedCentral
31.
go back to reference Franciosi F, Coticchio G, Lodde V, Tessaro I, Modina SC, Fadini R, et al. Natriuretic peptide precursor C delays meiotic resumption and sustains gap junction-mediated communication in bovine cumulus-enclosed oocytes. Biol Reprod. 2014;91(3):61.CrossRefPubMed Franciosi F, Coticchio G, Lodde V, Tessaro I, Modina SC, Fadini R, et al. Natriuretic peptide precursor C delays meiotic resumption and sustains gap junction-mediated communication in bovine cumulus-enclosed oocytes. Biol Reprod. 2014;91(3):61.CrossRefPubMed
32.
go back to reference Sugimura S, Ritter LJ, Sutton-McDowall ML, Mottershead DG, Thompson JG, Gilchrist RB. Amphiregulin co-operates with bone morphogenetic protein 15 to increase bovine oocyte developmental competence: effects on gap junction-mediated metabolite supply. Mol Hum Reprod. 2014;20(6):499–513.CrossRefPubMed Sugimura S, Ritter LJ, Sutton-McDowall ML, Mottershead DG, Thompson JG, Gilchrist RB. Amphiregulin co-operates with bone morphogenetic protein 15 to increase bovine oocyte developmental competence: effects on gap junction-mediated metabolite supply. Mol Hum Reprod. 2014;20(6):499–513.CrossRefPubMed
33.
go back to reference Winterhager E, Kidder GM. Gap junction connexins in female reproductive organs: implications for women’s reproductive health. Hum Reprod Update. 2015;21(3):340–52.CrossRefPubMed Winterhager E, Kidder GM. Gap junction connexins in female reproductive organs: implications for women’s reproductive health. Hum Reprod Update. 2015;21(3):340–52.CrossRefPubMed
34.
go back to reference Downs SM, Coleman DL, Ward-Bailey PF, Eppig JJ. Hypoxanthine is the principal inhibitor of murine oocyte maturation in a low molecular weight fraction of porcine follicular fluid. Proc Natl Acad Sci U S A. 1985;82(2):454–8.CrossRefPubMedPubMedCentral Downs SM, Coleman DL, Ward-Bailey PF, Eppig JJ. Hypoxanthine is the principal inhibitor of murine oocyte maturation in a low molecular weight fraction of porcine follicular fluid. Proc Natl Acad Sci U S A. 1985;82(2):454–8.CrossRefPubMedPubMedCentral
35.
go back to reference Eppig JJ, Ward-Bailey PF, Coleman DL. Hypoxanthine and adenosine in murine ovarian follicular fluid: concentrations and activity in maintaining oocyte meiotic arrest. Biol Reprod. 1985;33(5):1041–9.CrossRefPubMed Eppig JJ, Ward-Bailey PF, Coleman DL. Hypoxanthine and adenosine in murine ovarian follicular fluid: concentrations and activity in maintaining oocyte meiotic arrest. Biol Reprod. 1985;33(5):1041–9.CrossRefPubMed
36.
go back to reference Laforest MF, Pouliot E, Gueguen L, Richard FJ. Fundamental significance of specific phosphodiesterases in the control of spontaneous meiotic resumption in porcine oocytes. Mol Reprod Dev. 2005;70(3):361–72.CrossRefPubMed Laforest MF, Pouliot E, Gueguen L, Richard FJ. Fundamental significance of specific phosphodiesterases in the control of spontaneous meiotic resumption in porcine oocytes. Mol Reprod Dev. 2005;70(3):361–72.CrossRefPubMed
37.
go back to reference Bakhtari A, Ross PJ. DPPA3 prevents cytosine hydroxymethylation of the maternal pronucleus and is required for normal development in bovine embryos. Epigenetics. 2014;9(9):1271–9.CrossRefPubMedPubMedCentral Bakhtari A, Ross PJ. DPPA3 prevents cytosine hydroxymethylation of the maternal pronucleus and is required for normal development in bovine embryos. Epigenetics. 2014;9(9):1271–9.CrossRefPubMedPubMedCentral
38.
go back to reference Funahashi H, Cantley TC, Day BN. Synchronization of meiosis in porcine oocytes by exposure to dibutyryl cyclic adenosine monophosphate improves developmental competence following in vitro fertilization. Biol Reprod. 1997;57(1):49–53.CrossRefPubMed Funahashi H, Cantley TC, Day BN. Synchronization of meiosis in porcine oocytes by exposure to dibutyryl cyclic adenosine monophosphate improves developmental competence following in vitro fertilization. Biol Reprod. 1997;57(1):49–53.CrossRefPubMed
39.
go back to reference Luciano AM, Modina S, Vassena R, Milanesi E, Lauria A, Gandolfi F. Role of intracellular cyclic adenosine 3′,5′-monophosphate concentration and oocyte-cumulus cells communications on the acquisition of the developmental competence during in vitro maturation of bovine oocyte. Biol Reprod. 2004;70(2):465–72.CrossRefPubMed Luciano AM, Modina S, Vassena R, Milanesi E, Lauria A, Gandolfi F. Role of intracellular cyclic adenosine 3′,5′-monophosphate concentration and oocyte-cumulus cells communications on the acquisition of the developmental competence during in vitro maturation of bovine oocyte. Biol Reprod. 2004;70(2):465–72.CrossRefPubMed
40.
go back to reference Nogueira D, Albano C, Adriaenssens T, Cortvrindt R, Bourgain C, Devroey P, et al. Human oocytes reversibly arrested in prophase I by phosphodiesterase type 3 inhibitor in vitro. Biol Reprod. 2003a;69(3):1042–52.CrossRefPubMed Nogueira D, Albano C, Adriaenssens T, Cortvrindt R, Bourgain C, Devroey P, et al. Human oocytes reversibly arrested in prophase I by phosphodiesterase type 3 inhibitor in vitro. Biol Reprod. 2003a;69(3):1042–52.CrossRefPubMed
41.
go back to reference Nogueira D, Cortvrindt R, De Matos DG, Vanhoutte L, Smitz J. Effect of phosphodiesterase type 3 inhibitor on developmental competence of immature mouse oocytes in vitro. Biol Reprod. 2003b;69(6):2045–52.CrossRefPubMed Nogueira D, Cortvrindt R, De Matos DG, Vanhoutte L, Smitz J. Effect of phosphodiesterase type 3 inhibitor on developmental competence of immature mouse oocytes in vitro. Biol Reprod. 2003b;69(6):2045–52.CrossRefPubMed
42.
go back to reference Shu YM, Zeng HT, Ren Z, Zhuang GL, Liang XY, Shen HW, et al. Effects of cilostamide and forskolin on the meiotic resumption and embryonic development of immature human oocytes. Hum Reprod. 2008;23(3):504–13.CrossRefPubMed Shu YM, Zeng HT, Ren Z, Zhuang GL, Liang XY, Shen HW, et al. Effects of cilostamide and forskolin on the meiotic resumption and embryonic development of immature human oocytes. Hum Reprod. 2008;23(3):504–13.CrossRefPubMed
43.
go back to reference Thomas RE, Armstrong DT, Gilchrist RB. Bovine cumulus cell-oocyte gap junctional communication during in vitro maturation in response to manipulation of cell-specific cyclic adenosine 3′,5′-monophosophate levels. Biol Reprod. 2004a;70(3):548–56.CrossRefPubMed Thomas RE, Armstrong DT, Gilchrist RB. Bovine cumulus cell-oocyte gap junctional communication during in vitro maturation in response to manipulation of cell-specific cyclic adenosine 3′,5′-monophosophate levels. Biol Reprod. 2004a;70(3):548–56.CrossRefPubMed
44.
go back to reference Thomas RE, Thompson JG, Armstrong DT, Gilchrist RB. Effect of specific phosphodiesterase isoenzyme inhibitors during in vitro maturation of bovine oocytes on meiotic and developmental capacity. Biol Reprod. 2004b;71(4):1142–9.CrossRefPubMed Thomas RE, Thompson JG, Armstrong DT, Gilchrist RB. Effect of specific phosphodiesterase isoenzyme inhibitors during in vitro maturation of bovine oocytes on meiotic and developmental capacity. Biol Reprod. 2004b;71(4):1142–9.CrossRefPubMed
45.
go back to reference Vanhoutte L, Nogueira D, De Sutter P. Prematuration of human denuded oocytes in a three-dimensional co-culture system: effects on meiosis progression and developmental competence. Hum Reprod. 2009a;24(3):658–69.CrossRefPubMed Vanhoutte L, Nogueira D, De Sutter P. Prematuration of human denuded oocytes in a three-dimensional co-culture system: effects on meiosis progression and developmental competence. Hum Reprod. 2009a;24(3):658–69.CrossRefPubMed
46.
go back to reference Vanhoutte L, Nogueira D, Dumortier F, De Sutter P. Assessment of a new in vitro maturation system for mouse and human cumulus-enclosed oocytes: three-dimensional prematuration culture in the presence of a phosphodiesterase 3-inhibitor. Hum Reprod. 2009b;24(8):1946–59.CrossRefPubMed Vanhoutte L, Nogueira D, Dumortier F, De Sutter P. Assessment of a new in vitro maturation system for mouse and human cumulus-enclosed oocytes: three-dimensional prematuration culture in the presence of a phosphodiesterase 3-inhibitor. Hum Reprod. 2009b;24(8):1946–59.CrossRefPubMed
47.
go back to reference Zeng HT, Ren Z, Guzman L, Wang X, Sutton-McDowall ML, Ritter LJ, et al. Heparin and cAMP modulators interact during pre-in vitro maturation to affect mouse and human oocyte meiosis and developmental competence. Hum Reprod. 2013;28(6):1536–45.CrossRefPubMed Zeng HT, Ren Z, Guzman L, Wang X, Sutton-McDowall ML, Ritter LJ, et al. Heparin and cAMP modulators interact during pre-in vitro maturation to affect mouse and human oocyte meiosis and developmental competence. Hum Reprod. 2013;28(6):1536–45.CrossRefPubMed
48.
go back to reference Kawamura K, Cheng Y, Kawamura N, Takae S, Okada A, Kawagoe Y, et al. Pre-ovulatory LH/hCG surge decreases C-type natriuretic peptide secretion by ovarian granulosa cells to promote meiotic resumption of pre-ovulatory oocytes. Hum Reprod. 2011;26(11):3094–101.CrossRefPubMed Kawamura K, Cheng Y, Kawamura N, Takae S, Okada A, Kawagoe Y, et al. Pre-ovulatory LH/hCG surge decreases C-type natriuretic peptide secretion by ovarian granulosa cells to promote meiotic resumption of pre-ovulatory oocytes. Hum Reprod. 2011;26(11):3094–101.CrossRefPubMed
49.
go back to reference Kiyosu C, Tsuji T, Yamada K, Kajita S, Kunieda T. NPPC/NPR2 signaling is essential for oocyte meiotic arrest and cumulus oophorus formation during follicular development in the mouse ovary. Reproduction. 2012;144(2):187–93.CrossRefPubMed Kiyosu C, Tsuji T, Yamada K, Kajita S, Kunieda T. NPPC/NPR2 signaling is essential for oocyte meiotic arrest and cumulus oophorus formation during follicular development in the mouse ovary. Reproduction. 2012;144(2):187–93.CrossRefPubMed
50.
go back to reference Robinson JW, Zhang M, Shuhaibar LC, Norris RP, Geerts A, Wunder F, et al. Luteinizing hormone reduces the activity of the NPR2 guanylyl cyclase in mouse ovarian follicles, contributing to the cyclic GMP decrease that promotes resumption of meiosis in oocytes. Dev Biol. 2012;366(2):308–16.CrossRefPubMedPubMedCentral Robinson JW, Zhang M, Shuhaibar LC, Norris RP, Geerts A, Wunder F, et al. Luteinizing hormone reduces the activity of the NPR2 guanylyl cyclase in mouse ovarian follicles, contributing to the cyclic GMP decrease that promotes resumption of meiosis in oocytes. Dev Biol. 2012;366(2):308–16.CrossRefPubMedPubMedCentral
51.
go back to reference Santiquet N, Papillon-Dion E, Djender N, Guillemette C, Richard FJ. New elements in the C-type natriuretic peptide signaling pathway inhibiting swine in vitro oocyte meiotic resumption. Biol Reprod. 2014;91(1):16.CrossRefPubMed Santiquet N, Papillon-Dion E, Djender N, Guillemette C, Richard FJ. New elements in the C-type natriuretic peptide signaling pathway inhibiting swine in vitro oocyte meiotic resumption. Biol Reprod. 2014;91(1):16.CrossRefPubMed
52.
go back to reference Zhang M, Su YQ, Sugiura K, Xia G, Eppig JJ. Granulosa cell ligand NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes. Science. 2010;330(6002):366–9.CrossRefPubMedPubMedCentral Zhang M, Su YQ, Sugiura K, Xia G, Eppig JJ. Granulosa cell ligand NPPC and its receptor NPR2 maintain meiotic arrest in mouse oocytes. Science. 2010;330(6002):366–9.CrossRefPubMedPubMedCentral
53.
go back to reference Cho WK, Stern S, Biggers JD. Inhibitory effect of dibutyryl cAMP on mouse oocyte maturation in vitro. J Exp Zool. 1974;187(3):383–6.CrossRefPubMed Cho WK, Stern S, Biggers JD. Inhibitory effect of dibutyryl cAMP on mouse oocyte maturation in vitro. J Exp Zool. 1974;187(3):383–6.CrossRefPubMed
54.
55.
go back to reference Schultz RM, Montgomery RR, Belanoff JR. Regulation of mouse oocyte meiotic maturation: implication of a decrease in oocyte cAMP and protein dephosphorylation in commitment to resume meiosis. Dev Biol. 1983;97(2):264–73.CrossRefPubMed Schultz RM, Montgomery RR, Belanoff JR. Regulation of mouse oocyte meiotic maturation: implication of a decrease in oocyte cAMP and protein dephosphorylation in commitment to resume meiosis. Dev Biol. 1983;97(2):264–73.CrossRefPubMed
56.
go back to reference Labrecque R, Lodde V, Dieci C, Tessaro I, Luciano AM, Sirard MA. Chromatin remodelling and histone m RNA accumulation in bovine germinal vesicle oocytes. Mol Reprod Dev. 2015;82(6):450–62.CrossRefPubMed Labrecque R, Lodde V, Dieci C, Tessaro I, Luciano AM, Sirard MA. Chromatin remodelling and histone m RNA accumulation in bovine germinal vesicle oocytes. Mol Reprod Dev. 2015;82(6):450–62.CrossRefPubMed
57.
go back to reference Lodde V, Franciosi F, Tessaro I, Modina SC, Luciano AM. Role of gap junction-mediated communications in regulating large-scale chromatin configuration remodeling and embryonic developmental competence acquisition in fully grown bovine oocyte. J Assist Reprod Genet. 2013;30(9):1219–26.CrossRefPubMedPubMedCentral Lodde V, Franciosi F, Tessaro I, Modina SC, Luciano AM. Role of gap junction-mediated communications in regulating large-scale chromatin configuration remodeling and embryonic developmental competence acquisition in fully grown bovine oocyte. J Assist Reprod Genet. 2013;30(9):1219–26.CrossRefPubMedPubMedCentral
58.
go back to reference Koyama K, Kang SS, Huang W, Yanagawa Y, Takahashi Y, Nagano M. Aging-related changes in in vitro-matured bovine oocytes: oxidative stress, mitochondrial activity and ATP content after nuclear maturation. J Reprod Dev. 2014;60(2):136–42.CrossRefPubMedPubMedCentral Koyama K, Kang SS, Huang W, Yanagawa Y, Takahashi Y, Nagano M. Aging-related changes in in vitro-matured bovine oocytes: oxidative stress, mitochondrial activity and ATP content after nuclear maturation. J Reprod Dev. 2014;60(2):136–42.CrossRefPubMedPubMedCentral
Metadata
Title
Transporting cumulus complexes using novel meiotic arresting conditions permits maintenance of oocyte developmental competence
Authors
Nicolas W. Santiquet
Jason R. Herrick
Angelica Giraldo
Jennifer P. Barfield
William B. Schoolcraft
Rebecca L. Krisher
Publication date
01-08-2017
Publisher
Springer US
Published in
Journal of Assisted Reproduction and Genetics / Issue 8/2017
Print ISSN: 1058-0468
Electronic ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-017-0958-7

Other articles of this Issue 8/2017

Journal of Assisted Reproduction and Genetics 8/2017 Go to the issue