Skip to main content
Top
Published in: Journal of Assisted Reproduction and Genetics 10/2018

01-10-2018 | Review

GDF-9 and BMP-15 direct the follicle symphony

Authors: Alexandra Sanfins, Patrícia Rodrigues, David F. Albertini

Published in: Journal of Assisted Reproduction and Genetics | Issue 10/2018

Login to get access

Abstract

Understanding the physiology underlying the complex dialog between the oocyte and its surrounding somatic cells within the ovarian follicle has been crucial in defining optimal procedures for the development of clinical approaches in ART for women suffering from infertility and ovarian dysfunction. Recent studies have implicated oocyte-secreted factors like growth differentiation factor 9 (GDF-9) and bone morphogenetic protein 15 (BMP-15), members of the transforming growth factor-beta (TGFβ) superfamily, as potent regulators of folliculogenesis and ovulation. These two factors act as biologically active heterodimers or as homodimers in a synergistic cooperation. Through autocrine and paracrine mechanisms, the GDF-9 and BMP-15 system has been shown to regulate growth, differentiation, and function of granulosa and thecal cells during follicular development playing a vital role in oocyte development, ovulation, fertilization, and embryonic competence. The present mini-review provides an overview of recent findings relating GDF-9 and BMP-15 as fundamental factors implicated in the regulation of ovarian function and discusses their potential role as markers of oocyte quality in women.
Literature
1.
go back to reference Richards JS, Fitzpatrick SL, Clemens JW, Morris JK, Alliston T, Sirois J. Ovarian cell differentiation: a cascade of multiple hormones, cellular signals, and regulated genes. Recent Prog Horm Res. 1995;50:223–54.PubMed Richards JS, Fitzpatrick SL, Clemens JW, Morris JK, Alliston T, Sirois J. Ovarian cell differentiation: a cascade of multiple hormones, cellular signals, and regulated genes. Recent Prog Horm Res. 1995;50:223–54.PubMed
2.
go back to reference McGinnis LK, Limback SD, Albertini DF. Signaling modalities during oogenesis in mammals. Curr Top Dev Biol. 2013;102:227–42.PubMedCrossRef McGinnis LK, Limback SD, Albertini DF. Signaling modalities during oogenesis in mammals. Curr Top Dev Biol. 2013;102:227–42.PubMedCrossRef
3.
go back to reference Albertini DF, Barrett SL. Oocyte-somatic cell communication. Reprod Suppl. 2003;61:49–54.PubMed Albertini DF, Barrett SL. Oocyte-somatic cell communication. Reprod Suppl. 2003;61:49–54.PubMed
4.
go back to reference Rodrigues P, Limback D, McGinnis LK, Plancha CE, Albertini DF. Oogenesis: prospects and challenges for the future. J Cell Physiol. 2008;216(2):355–65.PubMedCrossRef Rodrigues P, Limback D, McGinnis LK, Plancha CE, Albertini DF. Oogenesis: prospects and challenges for the future. J Cell Physiol. 2008;216(2):355–65.PubMedCrossRef
5.
go back to reference Carabatsos MJ, Sellitto C, Goodenough DA, Albertini DF. Oocyte-granulosa cell heterologous gap junctions are required for the coordination of nuclear and cytoplasmic meiotic competence. Dev Biol. 2000;226(2):167–79.PubMedCrossRef Carabatsos MJ, Sellitto C, Goodenough DA, Albertini DF. Oocyte-granulosa cell heterologous gap junctions are required for the coordination of nuclear and cytoplasmic meiotic competence. Dev Biol. 2000;226(2):167–79.PubMedCrossRef
6.
go back to reference Gilchrist RB, Ritter LJ, Armstrong DT. Oocyte-somatic cell interactions during follicle development in mammals. Anim Reprod Sci. 2004;82-83:431–46.PubMedCrossRef Gilchrist RB, Ritter LJ, Armstrong DT. Oocyte-somatic cell interactions during follicle development in mammals. Anim Reprod Sci. 2004;82-83:431–46.PubMedCrossRef
7.
go back to reference Gougeon A. Regulation of ovarian follicular development in primates: facts and hypotheses. Endocr Rev. 1996;17(2):121–55.PubMedCrossRef Gougeon A. Regulation of ovarian follicular development in primates: facts and hypotheses. Endocr Rev. 1996;17(2):121–55.PubMedCrossRef
8.
go back to reference Albertini DF. The Mammalian Oocyte. In: The Mammalian Oocyte, in Knobil and Neill's Physiology of Reproduction (Fourth Edition). Editor: A.Z. Tony Plant; 2015. p. 59–97.CrossRef Albertini DF. The Mammalian Oocyte. In: The Mammalian Oocyte, in Knobil and Neill's Physiology of Reproduction (Fourth Edition). Editor: A.Z. Tony Plant; 2015. p. 59–97.CrossRef
9.
go back to reference Monniaux D. Driving folliculogenesis by the oocyte-somatic cell dialog: lessons from genetic models. Theriogenology. 2016;86(1):41–53.PubMedCrossRef Monniaux D. Driving folliculogenesis by the oocyte-somatic cell dialog: lessons from genetic models. Theriogenology. 2016;86(1):41–53.PubMedCrossRef
10.
go back to reference Falck B. Site of production of oestrogen in rat ovary as studied in micro-transplants. Acta Physiol Scand Suppl. 1959;47(163):1–101.PubMed Falck B. Site of production of oestrogen in rat ovary as studied in micro-transplants. Acta Physiol Scand Suppl. 1959;47(163):1–101.PubMed
11.
go back to reference el-Fouly MA, et al. Role of the ovum in follicular luteinization. Endocrinology. 1970;87(2):286–93.PubMedCrossRef el-Fouly MA, et al. Role of the ovum in follicular luteinization. Endocrinology. 1970;87(2):286–93.PubMedCrossRef
12.
go back to reference Nekola MV, Nalbandov AV. Morphological changes of rat follicular cells as influenced by oocytes. Biol Reprod. 1971;4(2):154–60.PubMedCrossRef Nekola MV, Nalbandov AV. Morphological changes of rat follicular cells as influenced by oocytes. Biol Reprod. 1971;4(2):154–60.PubMedCrossRef
13.
go back to reference Hutt KJ, Albertini DF. An oocentric view of folliculogenesis and embryogenesis. Reprod BioMed Online. 2007;14(6):758–64.PubMedCrossRef Hutt KJ, Albertini DF. An oocentric view of folliculogenesis and embryogenesis. Reprod BioMed Online. 2007;14(6):758–64.PubMedCrossRef
14.
go back to reference Erickson GF, Shimasaki S. The role of the oocyte in folliculogenesis. Trends Endocrinol Metab. 2000;11(5):193–8.PubMedCrossRef Erickson GF, Shimasaki S. The role of the oocyte in folliculogenesis. Trends Endocrinol Metab. 2000;11(5):193–8.PubMedCrossRef
15.
go back to reference Matzuk MM, Burns KH, Viveiros MM, Eppig JJ. Intercellular communication in the mammalian ovary: oocytes carry the conversation. Science. 2002;296(5576):2178–80.PubMedCrossRef Matzuk MM, Burns KH, Viveiros MM, Eppig JJ. Intercellular communication in the mammalian ovary: oocytes carry the conversation. Science. 2002;296(5576):2178–80.PubMedCrossRef
16.
go back to reference Gilchrist RB, Lane M, Thompson JG. Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Hum Reprod Update. 2008;14(2):159–77.PubMedCrossRef Gilchrist RB, Lane M, Thompson JG. Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Hum Reprod Update. 2008;14(2):159–77.PubMedCrossRef
17.
go back to reference Eppig JJ. Oocyte control of ovarian follicular development and function in mammals. Reproduction. 2001;122(6):829–38.PubMedCrossRef Eppig JJ. Oocyte control of ovarian follicular development and function in mammals. Reproduction. 2001;122(6):829–38.PubMedCrossRef
18.
go back to reference Eppig JJ, Chesnel F, Hirao Y, O'Brien MJ, Pendola FL, Watanabe S, et al. Oocyte control of granulosa cell development: how and why. Hum Reprod. 1997;12(11 Suppl):127–32.PubMed Eppig JJ, Chesnel F, Hirao Y, O'Brien MJ, Pendola FL, Watanabe S, et al. Oocyte control of granulosa cell development: how and why. Hum Reprod. 1997;12(11 Suppl):127–32.PubMed
19.
go back to reference Hussein TS, Thompson JG, Gilchrist RB. Oocyte-secreted factors enhance oocyte developmental competence. Dev Biol. 2006;296(2):514–21.PubMedCrossRef Hussein TS, Thompson JG, Gilchrist RB. Oocyte-secreted factors enhance oocyte developmental competence. Dev Biol. 2006;296(2):514–21.PubMedCrossRef
20.
go back to reference Su YQ, Sugiura K, Eppig JJ. Mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med. 2009;27(1):32–42.PubMedPubMedCentralCrossRef Su YQ, Sugiura K, Eppig JJ. Mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med. 2009;27(1):32–42.PubMedPubMedCentralCrossRef
21.
go back to reference Vanderhyden BC, et al. Evaluation of members of the TGFbeta superfamily as candidates for the oocyte factors that control mouse cumulus expansion and steroidogenesis. Reprod Suppl. 2003;61:55–70.PubMed Vanderhyden BC, et al. Evaluation of members of the TGFbeta superfamily as candidates for the oocyte factors that control mouse cumulus expansion and steroidogenesis. Reprod Suppl. 2003;61:55–70.PubMed
22.
go back to reference Vanderhyden BC, Tonary AM. Differential regulation of progesterone and estradiol production by mouse cumulus and mural granulosa cells by A factor(s) secreted by the oocyte. Biol Reprod. 1995;53(6):1243–50.PubMedCrossRef Vanderhyden BC, Tonary AM. Differential regulation of progesterone and estradiol production by mouse cumulus and mural granulosa cells by A factor(s) secreted by the oocyte. Biol Reprod. 1995;53(6):1243–50.PubMedCrossRef
23.
go back to reference Eppig JJ, Wigglesworth K, Pendola F, Hirao Y. Murine oocytes suppress expression of luteinizing hormone receptor messenger ribonucleic acid by granulosa cells. Biol Reprod. 1997;56(4):976–84.PubMedCrossRef Eppig JJ, Wigglesworth K, Pendola F, Hirao Y. Murine oocytes suppress expression of luteinizing hormone receptor messenger ribonucleic acid by granulosa cells. Biol Reprod. 1997;56(4):976–84.PubMedCrossRef
24.
go back to reference Gilchrist RB, et al. Molecular basis of oocyte-paracrine signalling that promotes granulosa cell proliferation. J Cell Sci. 2006;119(Pt 18):3811–21.PubMedCrossRef Gilchrist RB, et al. Molecular basis of oocyte-paracrine signalling that promotes granulosa cell proliferation. J Cell Sci. 2006;119(Pt 18):3811–21.PubMedCrossRef
25.
go back to reference Eppig JJ, Pendola FL, Wigglesworth K. Mouse oocytes suppress cAMP-induced expression of LH receptor mRNA by granulosa cells in vitro. Mol Reprod Dev. 1998;49(3):327–32.PubMedCrossRef Eppig JJ, Pendola FL, Wigglesworth K. Mouse oocytes suppress cAMP-induced expression of LH receptor mRNA by granulosa cells in vitro. Mol Reprod Dev. 1998;49(3):327–32.PubMedCrossRef
26.
go back to reference Gilchrist RB, Ritter LJ, Armstrong DT. Mouse oocyte mitogenic activity is developmentally coordinated throughout folliculogenesis and meiotic maturation. Dev Biol. 2001;240(1):289–98.PubMedCrossRef Gilchrist RB, Ritter LJ, Armstrong DT. Mouse oocyte mitogenic activity is developmentally coordinated throughout folliculogenesis and meiotic maturation. Dev Biol. 2001;240(1):289–98.PubMedCrossRef
27.
go back to reference Armstrong DT, Xia P, de Gannes G, Tekpetey FR, Khamsi F. Differential effects of insulin-like growth factor-I and follicle-stimulating hormone on proliferation and differentiation of bovine cumulus cells and granulosa cells. Biol Reprod. 1996;54(2):331–8.PubMedCrossRef Armstrong DT, Xia P, de Gannes G, Tekpetey FR, Khamsi F. Differential effects of insulin-like growth factor-I and follicle-stimulating hormone on proliferation and differentiation of bovine cumulus cells and granulosa cells. Biol Reprod. 1996;54(2):331–8.PubMedCrossRef
28.
go back to reference Li R, Norman RJ, Armstrong DT, Gilchrist RB. Oocyte-secreted factor(s) determine functional differences between bovine mural granulosa cells and cumulus cells. Biol Reprod. 2000;63(3):839–45.PubMedCrossRef Li R, Norman RJ, Armstrong DT, Gilchrist RB. Oocyte-secreted factor(s) determine functional differences between bovine mural granulosa cells and cumulus cells. Biol Reprod. 2000;63(3):839–45.PubMedCrossRef
29.
go back to reference Coskun S, Uzumcu M, Lin YC, Friedman CI, Alak BM. Regulation of cumulus cell steroidogenesis by the porcine oocyte and preliminary characterization of oocyte-produced factor(s). Biol Reprod. 1995;53(3):670–5.PubMedCrossRef Coskun S, Uzumcu M, Lin YC, Friedman CI, Alak BM. Regulation of cumulus cell steroidogenesis by the porcine oocyte and preliminary characterization of oocyte-produced factor(s). Biol Reprod. 1995;53(3):670–5.PubMedCrossRef
30.
go back to reference Hussein TS, et al. Oocytes prevent cumulus cell apoptosis by maintaining a morphogenic paracrine gradient of bone morphogenetic proteins. J Cell Sci. 2005;118(Pt 22):5257–68.PubMedCrossRef Hussein TS, et al. Oocytes prevent cumulus cell apoptosis by maintaining a morphogenic paracrine gradient of bone morphogenetic proteins. J Cell Sci. 2005;118(Pt 22):5257–68.PubMedCrossRef
31.
go back to reference Knight PG, Glister C. TGF-beta superfamily members and ovarian follicle development. Reproduction. 2006;132(2):191–206.PubMedCrossRef Knight PG, Glister C. TGF-beta superfamily members and ovarian follicle development. Reproduction. 2006;132(2):191–206.PubMedCrossRef
32.
go back to reference Chang H, Brown CW, Matzuk MM. Genetic analysis of the mammalian transforming growth factor-beta superfamily. Endocr Rev. 2002;23(6):787–823.PubMedCrossRef Chang H, Brown CW, Matzuk MM. Genetic analysis of the mammalian transforming growth factor-beta superfamily. Endocr Rev. 2002;23(6):787–823.PubMedCrossRef
33.
go back to reference Juengel JL, McNatty KP. The role of proteins of the transforming growth factor-beta superfamily in the intraovarian regulation of follicular development. Hum Reprod Update. 2005;11(2):143–60.PubMedCrossRef Juengel JL, McNatty KP. The role of proteins of the transforming growth factor-beta superfamily in the intraovarian regulation of follicular development. Hum Reprod Update. 2005;11(2):143–60.PubMedCrossRef
34.
go back to reference Chang HM, Qiao J, Leung PC. Oocyte-somatic cell interactions in the human ovary-novel role of bone morphogenetic proteins and growth differentiation factors. Hum Reprod Update. 2016;23(1):1–18.PubMedPubMedCentralCrossRef Chang HM, Qiao J, Leung PC. Oocyte-somatic cell interactions in the human ovary-novel role of bone morphogenetic proteins and growth differentiation factors. Hum Reprod Update. 2016;23(1):1–18.PubMedPubMedCentralCrossRef
35.
go back to reference de Castro FC, Cruz MH, Leal CL. Role of growth differentiation factor 9 and bone morphogenetic protein 15 in ovarian function and their importance in mammalian female fertility - a review. Asian-Australas J Anim Sci. 2016;29(8):1065–74.PubMedCrossRef de Castro FC, Cruz MH, Leal CL. Role of growth differentiation factor 9 and bone morphogenetic protein 15 in ovarian function and their importance in mammalian female fertility - a review. Asian-Australas J Anim Sci. 2016;29(8):1065–74.PubMedCrossRef
36.
go back to reference Persani L, Rossetti R, di Pasquale E, Cacciatore C, Fabre S. The fundamental role of bone morphogenetic protein 15 in ovarian function and its involvement in female fertility disorders. Hum Reprod Update. 2014;20(6):869–83.PubMedCrossRef Persani L, Rossetti R, di Pasquale E, Cacciatore C, Fabre S. The fundamental role of bone morphogenetic protein 15 in ovarian function and its involvement in female fertility disorders. Hum Reprod Update. 2014;20(6):869–83.PubMedCrossRef
38.
go back to reference Shimasaki S, Zachow RJ, Li D, Kim H, Iemura SI, Ueno N, et al. A functional bone morphogenetic protein system in the ovary. Proc Natl Acad Sci U S A. 1999;96(13):7282–7.PubMedPubMedCentralCrossRef Shimasaki S, Zachow RJ, Li D, Kim H, Iemura SI, Ueno N, et al. A functional bone morphogenetic protein system in the ovary. Proc Natl Acad Sci U S A. 1999;96(13):7282–7.PubMedPubMedCentralCrossRef
39.
go back to reference Berisha B, Schams D, Kosmann M, Amselgruber W, Einspanier R. Expression and localisation of vascular endothelial growth factor and basic fibroblast growth factor during the final growth of bovine ovarian follicles. J Endocrinol. 2000;167(3):371–82.PubMedCrossRef Berisha B, Schams D, Kosmann M, Amselgruber W, Einspanier R. Expression and localisation of vascular endothelial growth factor and basic fibroblast growth factor during the final growth of bovine ovarian follicles. J Endocrinol. 2000;167(3):371–82.PubMedCrossRef
40.
go back to reference Shimasaki S, Moore RK, Otsuka F, Erickson GF. The bone morphogenetic protein system in mammalian reproduction. Endocr Rev. 2004;25(1):72–101.PubMedCrossRef Shimasaki S, Moore RK, Otsuka F, Erickson GF. The bone morphogenetic protein system in mammalian reproduction. Endocr Rev. 2004;25(1):72–101.PubMedCrossRef
41.
go back to reference McPherron AC, Lee SJ. GDF-3 and GDF-9: two new members of the transforming growth factor-beta superfamily containing a novel pattern of cysteines. J Biol Chem. 1993;268(5):3444–9.PubMed McPherron AC, Lee SJ. GDF-3 and GDF-9: two new members of the transforming growth factor-beta superfamily containing a novel pattern of cysteines. J Biol Chem. 1993;268(5):3444–9.PubMed
42.
go back to reference McGrath SA, Esquela AF, Lee SJ. Oocyte-specific expression of growth/differentiation factor-9. Mol Endocrinol. 1995;9(1):131–6.PubMed McGrath SA, Esquela AF, Lee SJ. Oocyte-specific expression of growth/differentiation factor-9. Mol Endocrinol. 1995;9(1):131–6.PubMed
43.
go back to reference Laitinen M, Vuojolainen K, Jaatinen R, Ketola I, Aaltonen J, Lehtonen E, et al. A novel growth differentiation factor-9 (GDF-9) related factor is co-expressed with GDF-9 in mouse oocytes during folliculogenesis. Mech Dev. 1998;78(1–2):135–40.PubMedCrossRef Laitinen M, Vuojolainen K, Jaatinen R, Ketola I, Aaltonen J, Lehtonen E, et al. A novel growth differentiation factor-9 (GDF-9) related factor is co-expressed with GDF-9 in mouse oocytes during folliculogenesis. Mech Dev. 1998;78(1–2):135–40.PubMedCrossRef
44.
go back to reference Dube JL, Wang P, Elvin J, Lyons KM, Celeste AJ, Matzuk MM. The bone morphogenetic protein 15 gene is X-linked and expressed in oocytes. Mol Endocrinol. 1998;12(12):1809–17.PubMedCrossRef Dube JL, Wang P, Elvin J, Lyons KM, Celeste AJ, Matzuk MM. The bone morphogenetic protein 15 gene is X-linked and expressed in oocytes. Mol Endocrinol. 1998;12(12):1809–17.PubMedCrossRef
45.
go back to reference Incerti B, Dong J, Borsani G, Matzuk MM. Structure of the mouse growth/differentiation factor 9 gene. Biochim Biophys Acta. 1994;1222(1):125–8.PubMedCrossRef Incerti B, Dong J, Borsani G, Matzuk MM. Structure of the mouse growth/differentiation factor 9 gene. Biochim Biophys Acta. 1994;1222(1):125–8.PubMedCrossRef
46.
go back to reference Ahmad HI, et al. Maximum-likelihood approaches reveal signatures of positive selection in BMP-15 and GDF-9 genes modulating ovarian function in mammalian female fertility, in Ecol Evol. 2017. p. 8895–902.PubMedPubMedCentralCrossRef Ahmad HI, et al. Maximum-likelihood approaches reveal signatures of positive selection in BMP-15 and GDF-9 genes modulating ovarian function in mammalian female fertility, in Ecol Evol. 2017. p. 8895–902.PubMedPubMedCentralCrossRef
47.
go back to reference Ahmad HI, Ahmad MJ, Adeel MM, Asif AR, du X. Positive selection drives the evolution of endocrine regulatory bone morphogenetic protein system in mammals. Oncotarget. 2018;9(26):18435–45.PubMedPubMedCentralCrossRef Ahmad HI, Ahmad MJ, Adeel MM, Asif AR, du X. Positive selection drives the evolution of endocrine regulatory bone morphogenetic protein system in mammals. Oncotarget. 2018;9(26):18435–45.PubMedPubMedCentralCrossRef
48.
go back to reference Mottershead DG, Pulkki MM, Muggalla P, Pasternack A, Tolonen M, Myllymaa S, et al. Characterization of recombinant human growth differentiation factor-9 signaling in ovarian granulosa cells. Mol Cell Endocrinol. 2008;283(1–2):58–67.PubMedCrossRef Mottershead DG, Pulkki MM, Muggalla P, Pasternack A, Tolonen M, Myllymaa S, et al. Characterization of recombinant human growth differentiation factor-9 signaling in ovarian granulosa cells. Mol Cell Endocrinol. 2008;283(1–2):58–67.PubMedCrossRef
49.
go back to reference Paulini F, Melo EO. The role of oocyte-secreted factors GDF-9 and BMP-15 in follicular development and oogenesis. Reprod Domest Anim. 2011;46(2):354–61.PubMedCrossRef Paulini F, Melo EO. The role of oocyte-secreted factors GDF-9 and BMP-15 in follicular development and oogenesis. Reprod Domest Anim. 2011;46(2):354–61.PubMedCrossRef
50.
go back to reference McIntosh CJ, Lun S, Lawrence S, Western AH, McNatty KP, Juengel JL. The proregion of mouse BMP-15 regulates the cooperative interactions of BMP-15 and GDF-9. Biol Reprod. 2008;79(5):889–96.PubMedCrossRef McIntosh CJ, Lun S, Lawrence S, Western AH, McNatty KP, Juengel JL. The proregion of mouse BMP-15 regulates the cooperative interactions of BMP-15 and GDF-9. Biol Reprod. 2008;79(5):889–96.PubMedCrossRef
51.
go back to reference Yan C, Wang P, DeMayo J, DeMayo FJ, Elvin JA, Carino C, et al. Synergistic roles of bone morphogenetic protein 15 and growth differentiation factor 9 in ovarian function. Mol Endocrinol. 2001;15(6):854–66.PubMedCrossRef Yan C, Wang P, DeMayo J, DeMayo FJ, Elvin JA, Carino C, et al. Synergistic roles of bone morphogenetic protein 15 and growth differentiation factor 9 in ovarian function. Mol Endocrinol. 2001;15(6):854–66.PubMedCrossRef
52.
go back to reference Peng J, Li Q, Wigglesworth K, Rangarajan A, Kattamuri C, Peterson RT, et al. Growth differentiation factor 9: bone morphogenetic protein 15 heterodimers are potent regulators of ovarian functions. Proc Natl Acad Sci U S A. 2013;110(8):E776–85.PubMedPubMedCentralCrossRef Peng J, Li Q, Wigglesworth K, Rangarajan A, Kattamuri C, Peterson RT, et al. Growth differentiation factor 9: bone morphogenetic protein 15 heterodimers are potent regulators of ovarian functions. Proc Natl Acad Sci U S A. 2013;110(8):E776–85.PubMedPubMedCentralCrossRef
53.
go back to reference Mottershead DG, Sugimura S, al-Musawi SL, Li JJ, Richani D, White MA, et al. Cumulin, an oocyte-secreted heterodimer of the transforming growth factor-beta family, is a potent activator of granulosa cells and improves oocyte quality. J Biol Chem. 2015;290(39):24007–20.PubMedPubMedCentralCrossRef Mottershead DG, Sugimura S, al-Musawi SL, Li JJ, Richani D, White MA, et al. Cumulin, an oocyte-secreted heterodimer of the transforming growth factor-beta family, is a potent activator of granulosa cells and improves oocyte quality. J Biol Chem. 2015;290(39):24007–20.PubMedPubMedCentralCrossRef
54.
go back to reference Su YQ, Sugiura K, Wigglesworth K, O'Brien MJ, Affourtit JP, Pangas SA, et al. Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP-15 and GDF-9 control cholesterol biosynthesis in cumulus cells. Development. 2008;135(1):111–21.CrossRefPubMed Su YQ, Sugiura K, Wigglesworth K, O'Brien MJ, Affourtit JP, Pangas SA, et al. Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP-15 and GDF-9 control cholesterol biosynthesis in cumulus cells. Development. 2008;135(1):111–21.CrossRefPubMed
55.
go back to reference Sugiura K, Su YQ, Diaz FJ, Pangas SA, Sharma S, Wigglesworth K, et al. Oocyte-derived BMP-15 and FGFs cooperate to promote glycolysis in cumulus cells. Development. 2007;134(14):2593–603.PubMedCrossRef Sugiura K, Su YQ, Diaz FJ, Pangas SA, Sharma S, Wigglesworth K, et al. Oocyte-derived BMP-15 and FGFs cooperate to promote glycolysis in cumulus cells. Development. 2007;134(14):2593–603.PubMedCrossRef
56.
go back to reference Diaz FJ, Wigglesworth K, Eppig JJ. Oocytes are required for the preantral granulosa cell to cumulus cell transition in mice. Dev Biol. 2007;305(1):300–11.PubMedPubMedCentralCrossRef Diaz FJ, Wigglesworth K, Eppig JJ. Oocytes are required for the preantral granulosa cell to cumulus cell transition in mice. Dev Biol. 2007;305(1):300–11.PubMedPubMedCentralCrossRef
57.
go back to reference Hashimoto O, Moore RK, Shimasaki S. Posttranslational processing of mouse and human BMP-15: potential implication in the determination of ovulation quota. Proc Natl Acad Sci U S A. 2005;102(15):5426–31.PubMedPubMedCentralCrossRef Hashimoto O, Moore RK, Shimasaki S. Posttranslational processing of mouse and human BMP-15: potential implication in the determination of ovulation quota. Proc Natl Acad Sci U S A. 2005;102(15):5426–31.PubMedPubMedCentralCrossRef
58.
go back to reference Pangas SA, Matzuk MM. The art and artifact of GDF-9 activity: cumulus expansion and the cumulus expansion-enabling factor. Biol Reprod. 2005;73(4):582–5.PubMedCrossRef Pangas SA, Matzuk MM. The art and artifact of GDF-9 activity: cumulus expansion and the cumulus expansion-enabling factor. Biol Reprod. 2005;73(4):582–5.PubMedCrossRef
59.
go back to reference Diaz FJ, Wigglesworth K, Eppig JJ. Oocytes determine cumulus cell lineage in mouse ovarian follicles. J Cell Sci. 2007;120(Pt 8):1330–40.PubMedCrossRef Diaz FJ, Wigglesworth K, Eppig JJ. Oocytes determine cumulus cell lineage in mouse ovarian follicles. J Cell Sci. 2007;120(Pt 8):1330–40.PubMedCrossRef
60.
go back to reference Ernst EH, Franks S, Hardy K, Villesen P, Lykke-Hartmann K. Granulosa cells from human primordial and primary follicles show differential global gene expression profiles. Hum Reprod. 2018;33(4):666–79.PubMedCrossRef Ernst EH, Franks S, Hardy K, Villesen P, Lykke-Hartmann K. Granulosa cells from human primordial and primary follicles show differential global gene expression profiles. Hum Reprod. 2018;33(4):666–79.PubMedCrossRef
61.
go back to reference Chang HM, Cheng JC, Leung PC. Theca-derived BMP4 and BMP7 down-regulate connexin43 expression and decrease gap junction intercellular communication activity in immortalized human granulosa cells. J Clin Endocrinol Metab. 2013;98(3):E437–45.PubMedCrossRef Chang HM, Cheng JC, Leung PC. Theca-derived BMP4 and BMP7 down-regulate connexin43 expression and decrease gap junction intercellular communication activity in immortalized human granulosa cells. J Clin Endocrinol Metab. 2013;98(3):E437–45.PubMedCrossRef
62.
go back to reference Chang HM, Cheng JC, Taylor E, Leung PCK. Oocyte-derived BMP-15 but not GDF-9 down-regulates connexin43 expression and decreases gap junction intercellular communication activity in immortalized human granulosa cells. Mol Hum Reprod. 2014;20(5):373–83.PubMedPubMedCentralCrossRef Chang HM, Cheng JC, Taylor E, Leung PCK. Oocyte-derived BMP-15 but not GDF-9 down-regulates connexin43 expression and decreases gap junction intercellular communication activity in immortalized human granulosa cells. Mol Hum Reprod. 2014;20(5):373–83.PubMedPubMedCentralCrossRef
63.
go back to reference 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(6600):531–5.PubMedCrossRef 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(6600):531–5.PubMedCrossRef
64.
go back to reference Galloway SM, McNatty KP, Cambridge LM, Laitinen MPE, Juengel JL, Jokiranta TS, et al. Mutations in an oocyte-derived growth factor gene (BMP-15) cause increased ovulation rate and infertility in a dosage-sensitive manner. Nat Genet. 2000;25(3):279–83.PubMedCrossRef Galloway SM, McNatty KP, Cambridge LM, Laitinen MPE, Juengel JL, Jokiranta TS, et al. Mutations in an oocyte-derived growth factor gene (BMP-15) cause increased ovulation rate and infertility in a dosage-sensitive manner. Nat Genet. 2000;25(3):279–83.PubMedCrossRef
65.
go back to reference Gueripel X, Brun V, Gougeon A. Oocyte bone morphogenetic protein 15, but not growth differentiation factor 9, is increased during gonadotropin-induced follicular development in the immature mouse and is associated with cumulus oophorus expansion. Biol Reprod. 2006;75(6):836–43.PubMedCrossRef Gueripel X, Brun V, Gougeon A. Oocyte bone morphogenetic protein 15, but not growth differentiation factor 9, is increased during gonadotropin-induced follicular development in the immature mouse and is associated with cumulus oophorus expansion. Biol Reprod. 2006;75(6):836–43.PubMedCrossRef
66.
go back to reference Elvin JA, Yan C, Wang P, Nishimori K, Matzuk MM. Molecular characterization of the follicle defects in the growth differentiation factor 9-deficient ovary. Mol Endocrinol. 1999;13(6):1018–34.PubMedCrossRef Elvin JA, Yan C, Wang P, Nishimori K, Matzuk MM. Molecular characterization of the follicle defects in the growth differentiation factor 9-deficient ovary. Mol Endocrinol. 1999;13(6):1018–34.PubMedCrossRef
67.
go back to reference Li JJ, et al. Modifications of human growth differentiation factor 9 to improve the generation of embryos from low competence oocytes, in Mol Endocrinol. 2015. p. 40–52.PubMedCrossRef Li JJ, et al. Modifications of human growth differentiation factor 9 to improve the generation of embryos from low competence oocytes, in Mol Endocrinol. 2015. p. 40–52.PubMedCrossRef
68.
go back to reference Hennet ML, Combelles CM. The antral follicle: a microenvironment for oocyte differentiation. Int J Dev Biol. 2012;56(10–12):819–31.PubMedCrossRef Hennet ML, Combelles CM. The antral follicle: a microenvironment for oocyte differentiation. Int J Dev Biol. 2012;56(10–12):819–31.PubMedCrossRef
69.
go back to reference de Caestecker M. The transforming growth factor-beta superfamily of receptors. Cytokine Growth Factor Rev. 2004;15(1):1–11.PubMedCrossRef de Caestecker M. The transforming growth factor-beta superfamily of receptors. Cytokine Growth Factor Rev. 2004;15(1):1–11.PubMedCrossRef
70.
go back to reference Moore RK, Otsuka F, Shimasaki S. Molecular basis of bone morphogenetic protein-15 signaling in granulosa cells. J Biol Chem. 2003;278(1):304–10.PubMedCrossRef Moore RK, Otsuka F, Shimasaki S. Molecular basis of bone morphogenetic protein-15 signaling in granulosa cells. J Biol Chem. 2003;278(1):304–10.PubMedCrossRef
71.
go back to reference Mazerbourg S, Klein C, Roh J, Kaivo-Oja N, Mottershead DG, Korchynskyi O, et al. Growth differentiation factor-9 signaling is mediated by the type I receptor, activin receptor-like kinase 5. Mol Endocrinol. 2004;18(3):653–65.PubMedCrossRef Mazerbourg S, Klein C, Roh J, Kaivo-Oja N, Mottershead DG, Korchynskyi O, et al. Growth differentiation factor-9 signaling is mediated by the type I receptor, activin receptor-like kinase 5. Mol Endocrinol. 2004;18(3):653–65.PubMedCrossRef
72.
go back to reference Vitt UA, Mazerbourg S, Klein C, Hsueh AJW. Bone morphogenetic protein receptor type II is a receptor for growth differentiation factor-9. Biol Reprod. 2002;67(2):473–80.PubMedCrossRef Vitt UA, Mazerbourg S, Klein C, Hsueh AJW. Bone morphogenetic protein receptor type II is a receptor for growth differentiation factor-9. Biol Reprod. 2002;67(2):473–80.PubMedCrossRef
73.
go back to reference Mazerbourg S, Hsueh AJ. Genomic analyses facilitate identification of receptors and signalling pathways for growth differentiation factor 9 and related orphan bone morphogenetic protein/growth differentiation factor ligands. Hum Reprod Update. 2006;12(4):373–83.PubMedCrossRef Mazerbourg S, Hsueh AJ. Genomic analyses facilitate identification of receptors and signalling pathways for growth differentiation factor 9 and related orphan bone morphogenetic protein/growth differentiation factor ligands. Hum Reprod Update. 2006;12(4):373–83.PubMedCrossRef
74.
go back to reference Franzen P, et al. Cloning of a TGF beta type I receptor that forms a heteromeric complex with the TGF beta type II receptor. Cell. 1993;75(4):681–92.PubMedCrossRef Franzen P, et al. Cloning of a TGF beta type I receptor that forms a heteromeric complex with the TGF beta type II receptor. Cell. 1993;75(4):681–92.PubMedCrossRef
75.
go back to reference Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature. 1997;390(6659):465–71.PubMedCrossRef Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from cell membrane to nucleus through SMAD proteins. Nature. 1997;390(6659):465–71.PubMedCrossRef
76.
go back to reference Miyazawa K, Shinozaki M, Hara T, Furuya T, Miyazono K. Two major Smad pathways in TGF-beta superfamily signalling. Genes Cells. 2002;7(12):1191–204.PubMedCrossRef Miyazawa K, Shinozaki M, Hara T, Furuya T, Miyazono K. Two major Smad pathways in TGF-beta superfamily signalling. Genes Cells. 2002;7(12):1191–204.PubMedCrossRef
77.
go back to reference Reader KL, Heath DA, Lun S, McIntosh CJ, Western AH, Littlejohn RP, et al. Signalling pathways involved in the cooperative effects of ovine and murine GDF-9+BMP-15-stimulated thymidine uptake by rat granulosa cells. Reproduction. 2011;142(1):123–31.PubMedCrossRef Reader KL, Heath DA, Lun S, McIntosh CJ, Western AH, Littlejohn RP, et al. Signalling pathways involved in the cooperative effects of ovine and murine GDF-9+BMP-15-stimulated thymidine uptake by rat granulosa cells. Reproduction. 2011;142(1):123–31.PubMedCrossRef
78.
go back to reference Mottershead DG, Ritter LJ, Gilchrist RB. Signalling pathways mediating specific synergistic interactions between GDF-9 and BMP-15. Mol Hum Reprod. 2012;18(3):121–8.PubMedCrossRef Mottershead DG, Ritter LJ, Gilchrist RB. Signalling pathways mediating specific synergistic interactions between GDF-9 and BMP-15. Mol Hum Reprod. 2012;18(3):121–8.PubMedCrossRef
79.
go back to reference Reader KL, Mottershead DG, Martin GA, Gilchrist RB, Heath DA, McNatty KP, et al. Signalling pathways involved in the synergistic effects of human growth differentiation factor 9 and bone morphogenetic protein 15. Reprod Fertil Dev. 2016;28(4):491–8.PubMedCrossRef Reader KL, Mottershead DG, Martin GA, Gilchrist RB, Heath DA, McNatty KP, et al. Signalling pathways involved in the synergistic effects of human growth differentiation factor 9 and bone morphogenetic protein 15. Reprod Fertil Dev. 2016;28(4):491–8.PubMedCrossRef
80.
go back to reference Chen H, Liu C, Jiang H, Gao Y, Xu M, Wang J, et al. Regulatory role of miRNA-375 in expression of BMP-15/GDF-9 receptors and its effect on proliferation and apoptosis of bovine cumulus cells. Cell Physiol Biochem. 2017;41(2):439–50.PubMedCrossRef Chen H, Liu C, Jiang H, Gao Y, Xu M, Wang J, et al. Regulatory role of miRNA-375 in expression of BMP-15/GDF-9 receptors and its effect on proliferation and apoptosis of bovine cumulus cells. Cell Physiol Biochem. 2017;41(2):439–50.PubMedCrossRef
81.
go back to reference Liu C, Yuan B, Chen H, Xu M, Sun X, Xu JJ, et al. Effects of MiR-375-BMPR2 as a key factor downstream of BMP-15/GDF-9 on the Smad1/5/8 and Smad2/3 signaling pathways. Cell Physiol Biochem. 2018;46(1):213–25.PubMedCrossRef Liu C, Yuan B, Chen H, Xu M, Sun X, Xu JJ, et al. Effects of MiR-375-BMPR2 as a key factor downstream of BMP-15/GDF-9 on the Smad1/5/8 and Smad2/3 signaling pathways. Cell Physiol Biochem. 2018;46(1):213–25.PubMedCrossRef
82.
go back to reference Gilchrist RB, Ritter LJ. Differences in the participation of TGFB superfamily signalling pathways mediating porcine and murine cumulus cell expansion. Reproduction. 2011;142(5):647–57.PubMedCrossRef Gilchrist RB, Ritter LJ. Differences in the participation of TGFB superfamily signalling pathways mediating porcine and murine cumulus cell expansion. Reproduction. 2011;142(5):647–57.PubMedCrossRef
83.
go back to reference Chang HM, Cheng JC, Klausen C, Leung PCK. BMP-15 suppresses progesterone production by down-regulating StAR via ALK3 in human granulosa cells. Mol Endocrinol. 2013;27(12):2093–104.PubMedPubMedCentralCrossRef Chang HM, Cheng JC, Klausen C, Leung PCK. BMP-15 suppresses progesterone production by down-regulating StAR via ALK3 in human granulosa cells. Mol Endocrinol. 2013;27(12):2093–104.PubMedPubMedCentralCrossRef
84.
go back to reference Albertini DF, et al. Cellular basis for paracrine regulation of ovarian follicle development. Reproduction. 2001;121(5):647–53.PubMedCrossRef Albertini DF, et al. Cellular basis for paracrine regulation of ovarian follicle development. Reproduction. 2001;121(5):647–53.PubMedCrossRef
85.
go back to reference Grondahl ML, et al. Anti-Mullerian hormone remains highly expressed in human cumulus cells during the final stages of folliculogenesis. Reprod BioMed Online. 2011;22(4):389–98.PubMedCrossRef Grondahl ML, et al. Anti-Mullerian hormone remains highly expressed in human cumulus cells during the final stages of folliculogenesis. Reprod BioMed Online. 2011;22(4):389–98.PubMedCrossRef
86.
go back to reference Dekel N, Kraicer PF. Induction in vitro of mucification of rat cumulus oophorus by gonadotrophins and adenosine 3′,5′-monophosphate. Endocrinology. 1978;102(6):1797–802.PubMedCrossRef Dekel N, Kraicer PF. Induction in vitro of mucification of rat cumulus oophorus by gonadotrophins and adenosine 3′,5′-monophosphate. Endocrinology. 1978;102(6):1797–802.PubMedCrossRef
87.
go back to reference Eppig JJ. Regulation of cumulus oophorus expansion by gonadotropins in vivo and in vitro. Biol Reprod. 1980;23(3):545–52.PubMedCrossRef Eppig JJ. Regulation of cumulus oophorus expansion by gonadotropins in vivo and in vitro. Biol Reprod. 1980;23(3):545–52.PubMedCrossRef
88.
go back to reference Russell DL, Robker RL. Molecular mechanisms of ovulation: co-ordination through the cumulus complex. Hum Reprod Update. 2007;13(3):289–312.PubMedCrossRef Russell DL, Robker RL. Molecular mechanisms of ovulation: co-ordination through the cumulus complex. Hum Reprod Update. 2007;13(3):289–312.PubMedCrossRef
89.
go back to reference Fang L, Cheng JC, Chang HM, Sun YP, Leung PCK. EGF-like growth factors induce COX-2-derived PGE2 production through ERK1/2 in human granulosa cells. J Clin Endocrinol Metab. 2013;98(12):4932–41.PubMedCrossRef Fang L, Cheng JC, Chang HM, Sun YP, Leung PCK. EGF-like growth factors induce COX-2-derived PGE2 production through ERK1/2 in human granulosa cells. J Clin Endocrinol Metab. 2013;98(12):4932–41.PubMedCrossRef
90.
go back to reference Russell DL, Salustri A. Extracellular matrix of the cumulus-oocyte complex. Semin Reprod Med. 2006;24(4):217–27.PubMedCrossRef Russell DL, Salustri A. Extracellular matrix of the cumulus-oocyte complex. Semin Reprod Med. 2006;24(4):217–27.PubMedCrossRef
91.
go back to reference Baranova NS, Inforzato A, Briggs DC, Tilakaratna V, Enghild JJ, Thakar D, et al. Incorporation of pentraxin 3 into hyaluronan matrices is tightly regulated and promotes matrix cross-linking. J Biol Chem. 2014;289(44):30481–98.PubMedPubMedCentralCrossRef Baranova NS, Inforzato A, Briggs DC, Tilakaratna V, Enghild JJ, Thakar D, et al. Incorporation of pentraxin 3 into hyaluronan matrices is tightly regulated and promotes matrix cross-linking. J Biol Chem. 2014;289(44):30481–98.PubMedPubMedCentralCrossRef
92.
go back to reference Zhang H, Tian S, Klausen C, Zhu H, Liu R, Leung PCK. Differential activation of noncanonical SMAD2/SMAD3 signaling by bone morphogenetic proteins causes disproportionate induction of hyaluronan production in immortalized human granulosa cells. Mol Cell Endocrinol. 2016;428:17–27.PubMedCrossRef Zhang H, Tian S, Klausen C, Zhu H, Liu R, Leung PCK. Differential activation of noncanonical SMAD2/SMAD3 signaling by bone morphogenetic proteins causes disproportionate induction of hyaluronan production in immortalized human granulosa cells. Mol Cell Endocrinol. 2016;428:17–27.PubMedCrossRef
93.
go back to reference Hanrahan JP, Gregan SM, Mulsant P, Mullen M, Davis GH, Powell R, et al. Mutations in the genes for oocyte-derived growth factors GDF-9 and BMP-15 are associated with both increased ovulation rate and sterility in Cambridge and Belclare sheep (Ovis aries). Biol Reprod. 2004;70(4):900–9.PubMedCrossRef Hanrahan JP, Gregan SM, Mulsant P, Mullen M, Davis GH, Powell R, et al. Mutations in the genes for oocyte-derived growth factors GDF-9 and BMP-15 are associated with both increased ovulation rate and sterility in Cambridge and Belclare sheep (Ovis aries). Biol Reprod. 2004;70(4):900–9.PubMedCrossRef
94.
go back to reference Wu YT, et al. High bone morphogenetic protein-15 level in follicular fluid is associated with high quality oocyte and subsequent embryonic development. Hum Reprod. 2007;22(6):1526–31.PubMedCrossRef Wu YT, et al. High bone morphogenetic protein-15 level in follicular fluid is associated with high quality oocyte and subsequent embryonic development. Hum Reprod. 2007;22(6):1526–31.PubMedCrossRef
95.
go back to reference Gode F, et al. Influence of follicular fluid GDF-9 and BMP-15 on embryo quality. Fertil Steril. 2011;95(7):2274–8.PubMedCrossRef Gode F, et al. Influence of follicular fluid GDF-9 and BMP-15 on embryo quality. Fertil Steril. 2011;95(7):2274–8.PubMedCrossRef
96.
97.
98.
go back to reference Takebayashi K, et al. Mutation analysis of the growth differentiation factor-9 and -9B genes in patients with premature ovarian failure and polycystic ovary syndrome. Fertil Steril. 2000;74(5):976–9.PubMedCrossRef Takebayashi K, et al. Mutation analysis of the growth differentiation factor-9 and -9B genes in patients with premature ovarian failure and polycystic ovary syndrome. Fertil Steril. 2000;74(5):976–9.PubMedCrossRef
99.
go back to reference Crispi S, et al. Transcriptional profiling of endometriosis tissues identifies genes related to organogenesis defects. J Cell Physiol. 2013;228(9):1927–34.PubMedCrossRef Crispi S, et al. Transcriptional profiling of endometriosis tissues identifies genes related to organogenesis defects. J Cell Physiol. 2013;228(9):1927–34.PubMedCrossRef
101.
go back to reference Tong S, Short RV. Dizygotic twinning as a measure of human fertility. Hum Reprod. 1998;13(1):95–8.PubMedCrossRef Tong S, Short RV. Dizygotic twinning as a measure of human fertility. Hum Reprod. 1998;13(1):95–8.PubMedCrossRef
102.
go back to reference Zhao SY, et al. Expression of growth differentiation factor-9 and bone morphogenetic protein-15 in oocytes and cumulus granulosa cells of patients with polycystic ovary syndrome. Fertil Steril. 2010;94(1):261–7.PubMedCrossRef Zhao SY, et al. Expression of growth differentiation factor-9 and bone morphogenetic protein-15 in oocytes and cumulus granulosa cells of patients with polycystic ovary syndrome. Fertil Steril. 2010;94(1):261–7.PubMedCrossRef
103.
go back to reference Dey SR, et al. Coculturing denuded oocytes during the in vitro maturation of bovine cumulus oocyte complexes exerts a synergistic effect on embryo development. Theriogenology. 2012;77(6):1064–77.PubMedCrossRef Dey SR, et al. Coculturing denuded oocytes during the in vitro maturation of bovine cumulus oocyte complexes exerts a synergistic effect on embryo development. Theriogenology. 2012;77(6):1064–77.PubMedCrossRef
104.
go back to reference Oocyte-secreted factors in oocyte maturation media enhance subsequent development of bovine cloned embryos - Su - 2014 - Mol Reprod Dev - Wiley Online Library. 2018. Oocyte-secreted factors in oocyte maturation media enhance subsequent development of bovine cloned embryos - Su - 2014 - Mol Reprod Dev - Wiley Online Library. 2018.
105.
go back to reference Sudiman J, et al. Effects of differing oocyte-secreted factors during mouse in vitro maturation on subsequent embryo and fetal development. J Assist Reprod Genet. 2014;31(3):295–306.PubMedPubMedCentralCrossRef Sudiman J, et al. Effects of differing oocyte-secreted factors during mouse in vitro maturation on subsequent embryo and fetal development. J Assist Reprod Genet. 2014;31(3):295–306.PubMedPubMedCentralCrossRef
106.
go back to reference Yeo CX, et al. Exogenous growth differentiation factor 9 in oocyte maturation media enhances subsequent embryo development and fetal viability in mice. Hum Reprod. 2008;23(1):67–73.PubMedCrossRef Yeo CX, et al. Exogenous growth differentiation factor 9 in oocyte maturation media enhances subsequent embryo development and fetal viability in mice. Hum Reprod. 2008;23(1):67–73.PubMedCrossRef
107.
go back to reference Romaguera R, et al. Oocyte secreted factors improve embryo developmental competence of COCs from small follicles in prepubertal goats. Theriogenology. 2010;74(6):1050–9.PubMedCrossRef Romaguera R, et al. Oocyte secreted factors improve embryo developmental competence of COCs from small follicles in prepubertal goats. Theriogenology. 2010;74(6):1050–9.PubMedCrossRef
108.
go back to reference Gomez MN, et al. Effect of oocyte-secreted factors on porcine in vitro maturation, cumulus expansion and developmental competence of parthenotes. Zygote. 2012;20(2):135–45.PubMedCrossRef Gomez MN, et al. Effect of oocyte-secreted factors on porcine in vitro maturation, cumulus expansion and developmental competence of parthenotes. Zygote. 2012;20(2):135–45.PubMedCrossRef
109.
go back to reference Ferrarini E, et al. Clinical characteristics and genetic analysis in women with premature ovarian insufficiency. Maturitas. 2013;74(1):61–7.PubMedCrossRef Ferrarini E, et al. Clinical characteristics and genetic analysis in women with premature ovarian insufficiency. Maturitas. 2013;74(1):61–7.PubMedCrossRef
110.
go back to reference Persani L, Rossetti R, Cacciatore C. Genes involved in human premature ovarian failure. J Mol Endocrinol. 2010;45(5):257–79.PubMedCrossRef Persani L, Rossetti R, Cacciatore C. Genes involved in human premature ovarian failure. J Mol Endocrinol. 2010;45(5):257–79.PubMedCrossRef
111.
go back to reference Tiotiu D, et al. Variants of the BMP-15 gene in a cohort of patients with premature ovarian failure. Hum Reprod. 2010;25(6):1581–7.PubMedCrossRef Tiotiu D, et al. Variants of the BMP-15 gene in a cohort of patients with premature ovarian failure. Hum Reprod. 2010;25(6):1581–7.PubMedCrossRef
112.
go back to reference Auclair S, et al. Positive selection in bone morphogenetic protein 15 targets a natural mutation associated with primary ovarian insufficiency in human. PLoS One. 2013;8(10):e78199.PubMedPubMedCentralCrossRef Auclair S, et al. Positive selection in bone morphogenetic protein 15 targets a natural mutation associated with primary ovarian insufficiency in human. PLoS One. 2013;8(10):e78199.PubMedPubMedCentralCrossRef
113.
go back to reference Di Pasquale E, et al. Identification of new variants of human BMP-15 gene in a large cohort of women with premature ovarian failure. J Clin Endocrinol Metab. 2006;91(5):1976–9.PubMedCrossRef Di Pasquale E, et al. Identification of new variants of human BMP-15 gene in a large cohort of women with premature ovarian failure. J Clin Endocrinol Metab. 2006;91(5):1976–9.PubMedCrossRef
114.
115.
go back to reference Patino LC, et al. BMP-15 mutations associated with primary ovarian insufficiency reduce expression, activity, or synergy with GDF-9. J Clin Endocrinol Metab. 2017;102(3):1009–19.PubMed Patino LC, et al. BMP-15 mutations associated with primary ovarian insufficiency reduce expression, activity, or synergy with GDF-9. J Clin Endocrinol Metab. 2017;102(3):1009–19.PubMed
116.
go back to reference Regan SL, et al. Dysregulation of granulosal bone morphogenetic protein receptor 1B density is associated with reduced ovarian reserve and the age-related decline in human fertility. Mol Cell Endocrinol. 2016;425:84–93.PubMedCrossRef Regan SL, et al. Dysregulation of granulosal bone morphogenetic protein receptor 1B density is associated with reduced ovarian reserve and the age-related decline in human fertility. Mol Cell Endocrinol. 2016;425:84–93.PubMedCrossRef
117.
go back to reference Salehnia M, Pajokh M, Ghorbanmehr N. Short term organ culture of mouse ovary in the medium supplemented with bone morphogenetic protein 15 and follicle stimulating hormone: a morphological, hormonal and molecular study. J Reprod Infertil. 2016;17(4):199–207.PubMedPubMedCentral Salehnia M, Pajokh M, Ghorbanmehr N. Short term organ culture of mouse ovary in the medium supplemented with bone morphogenetic protein 15 and follicle stimulating hormone: a morphological, hormonal and molecular study. J Reprod Infertil. 2016;17(4):199–207.PubMedPubMedCentral
118.
go back to reference Devine PJ, Perreault SD, Luderer U. Roles of reactive oxygen species and antioxidants in ovarian toxicity. Biol Reprod. 2012;86(2):27.PubMedCrossRef Devine PJ, Perreault SD, Luderer U. Roles of reactive oxygen species and antioxidants in ovarian toxicity. Biol Reprod. 2012;86(2):27.PubMedCrossRef
Metadata
Title
GDF-9 and BMP-15 direct the follicle symphony
Authors
Alexandra Sanfins
Patrícia Rodrigues
David F. Albertini
Publication date
01-10-2018
Publisher
Springer US
Published in
Journal of Assisted Reproduction and Genetics / Issue 10/2018
Print ISSN: 1058-0468
Electronic ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-018-1268-4

Other articles of this Issue 10/2018

Journal of Assisted Reproduction and Genetics 10/2018 Go to the issue