Skip to main content
Erschienen in: Journal of Assisted Reproduction and Genetics 8/2018

30.06.2018 | Reproductive Physiology and Disease

Oviductal glycoprotein 1 (OVGP1) is expressed by endometrial epithelium that regulates receptivity and trophoblast adhesion

verfasst von: Saniya Laheri, Nancy Ashary, Purvi Bhatt, Deepak Modi

Erschienen in: Journal of Assisted Reproduction and Genetics | Ausgabe 8/2018

Einloggen, um Zugang zu erhalten

Abstract

Purpose

To study the regulation and functions of oviductal glycoprotein 1 (OVGP1) in endometrial epithelial cells.

Methods

Expression of OVGP1 in mouse endometrium during pregnancy and in the endometrial epithelial cell line (Ishikawa) was studied by immunofluorescence, Western blotting, and RT-PCR. Regulation of OVGP1 in response to ovarian steroids and human chorionic gonadotropin (hCG) was studied by real-time RT-PCR. OVGP1 expression was knockdown in Ishikawa cells by shRNA, and expression of receptivity associated genes was studied by real-time RT-PCR. Adhesion of trophoblast cell line (JAr) was studied by in vitro adhesion assays.

Results

OVGP1 was localized exclusively in the luminal epithelial cells of mouse endometrium at the time of embryo implantation. Along with estrogen and progesterone, hCG induced the expression of OVGP1 in Ishikawa cells. Knockdown of OVGP1 in Ishikawa cells reduced mRNA expression of ITGAV, ITGB3, ITGA5, HOXA10, LIF, and IL15; it increased the expression of HOXA11, MMP9, TIMP1, and TIMP3. Supernatants derived from OVGP1 knockdown Ishikawa cells reduced the adhesiveness of JAr cells in vitro. Expression of OVGP1 mRNA was found to be significantly lowered in the endometrium of women with recurrent implantation failure.

Conclusion

OVGP1 is specifically induced in the luminal epithelium at the time of embryo implantation where it regulates receptivity-related genes and aids in trophoblast adhesion.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Polanski LT, Baumgarten MN, Quenby S, Brosens J, Campbell BK, Raine-Fenning NJ. What exactly do we mean by ‘recurrent implantation failure’? A systematic review and opinion. Reprod BioMed Online. 2014;28(4):409–23.CrossRefPubMed Polanski LT, Baumgarten MN, Quenby S, Brosens J, Campbell BK, Raine-Fenning NJ. What exactly do we mean by ‘recurrent implantation failure’? A systematic review and opinion. Reprod BioMed Online. 2014;28(4):409–23.CrossRefPubMed
2.
Zurück zum Zitat European IVF-monitoring Consortium. Assisted reproductive technology in Europe, 2013: results generated from European registers by ESHRE. Hum Reprod. 2017;32(10):1957–73.CrossRef European IVF-monitoring Consortium. Assisted reproductive technology in Europe, 2013: results generated from European registers by ESHRE. Hum Reprod. 2017;32(10):1957–73.CrossRef
3.
Zurück zum Zitat Cha J, Sun X, Dey SK. Mechanisms of implantation: strategies for successful pregnancy. Nat Med. 2012;18(12):1754–67.CrossRefPubMed Cha J, Sun X, Dey SK. Mechanisms of implantation: strategies for successful pregnancy. Nat Med. 2012;18(12):1754–67.CrossRefPubMed
4.
Zurück zum Zitat Modi DN, Bhartiya P. Physiology of embryo-endometrial cross talk. Biomed Res J. 2015;2:83–104. Modi DN, Bhartiya P. Physiology of embryo-endometrial cross talk. Biomed Res J. 2015;2:83–104.
5.
Zurück zum Zitat Evans J, Salamonsen LA, Winship A, Menkhorst E, Nie G, Gargett CE, et al. Fertile ground: human endometrial programming and lessons in health and disease. Nat Rev Endocrinol. 2016;12(11):654–67.CrossRefPubMed Evans J, Salamonsen LA, Winship A, Menkhorst E, Nie G, Gargett CE, et al. Fertile ground: human endometrial programming and lessons in health and disease. Nat Rev Endocrinol. 2016;12(11):654–67.CrossRefPubMed
6.
Zurück zum Zitat Dey SK, Lim H, Das SK, Reese J, Paria BC, Daikoku T, et al. Molecular cues to implantation. Endocr Rev. 2004;25(3):341–73.CrossRefPubMed Dey SK, Lim H, Das SK, Reese J, Paria BC, Daikoku T, et al. Molecular cues to implantation. Endocr Rev. 2004;25(3):341–73.CrossRefPubMed
7.
Zurück zum Zitat Wang H, Dey SK. Roadmap to embryo implantation: clues from mouse models. Nat Rev Genet. 2006;7:185–99.CrossRefPubMed Wang H, Dey SK. Roadmap to embryo implantation: clues from mouse models. Nat Rev Genet. 2006;7:185–99.CrossRefPubMed
8.
Zurück zum Zitat Altmäe S, Koel M, Võsa U, Adler P, Suhorutšenko M, Laisk-Podar T, et al. Meta-signature of human endometrial receptivity: a meta-analysis and validation study of transcriptomic biomarkers. Sci Rep. 2017;7(1):10077.CrossRefPubMedPubMedCentral Altmäe S, Koel M, Võsa U, Adler P, Suhorutšenko M, Laisk-Podar T, et al. Meta-signature of human endometrial receptivity: a meta-analysis and validation study of transcriptomic biomarkers. Sci Rep. 2017;7(1):10077.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Díaz-Gimeno P, Ruiz-Alonso M, Sebastian-Leon P, Pellicer A, Valbuena D, Simón C. Window of implantation transcriptomic stratification reveals different endometrial subsignatures associated with live birth and biochemical pregnancy. Fertil Steril. 2017;108(4):703–10.CrossRefPubMed Díaz-Gimeno P, Ruiz-Alonso M, Sebastian-Leon P, Pellicer A, Valbuena D, Simón C. Window of implantation transcriptomic stratification reveals different endometrial subsignatures associated with live birth and biochemical pregnancy. Fertil Steril. 2017;108(4):703–10.CrossRefPubMed
10.
Zurück zum Zitat Teh WT, McBain J, Rogers P. What is the contribution of embryo-endometrial asynchrony to implantation failure? J Assist Reprod Genet. 2016;33(11):1419–30.CrossRefPubMedPubMedCentral Teh WT, McBain J, Rogers P. What is the contribution of embryo-endometrial asynchrony to implantation failure? J Assist Reprod Genet. 2016;33(11):1419–30.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Valdes CT, Schutt A, Simon C. Implantation failure of endometrial origin: it is not pathology, but our failure to synchronize the developing embryo with a receptive endometrium. Fertil Steril. 2017;108(1):15–8.CrossRefPubMed Valdes CT, Schutt A, Simon C. Implantation failure of endometrial origin: it is not pathology, but our failure to synchronize the developing embryo with a receptive endometrium. Fertil Steril. 2017;108(1):15–8.CrossRefPubMed
12.
Zurück zum Zitat Rosario GX, Modi DN, Sachdeva G, Manjramkar DD, Puri CP. Morphological events in the primate endometrium in the presence of a preimplantation embryo, detected by the serum preimplantation factor bioassay. Hum Reprod. 2005;20:61–71.CrossRefPubMed Rosario GX, Modi DN, Sachdeva G, Manjramkar DD, Puri CP. Morphological events in the primate endometrium in the presence of a preimplantation embryo, detected by the serum preimplantation factor bioassay. Hum Reprod. 2005;20:61–71.CrossRefPubMed
13.
Zurück zum Zitat Godbole GB, Modi DN, Puri CP. Regulation of homeobox A10 expression in the primate endometrium by progesterone and embryonic stimuli. Reproduction. 2007;134(3):513–23.CrossRefPubMed Godbole GB, Modi DN, Puri CP. Regulation of homeobox A10 expression in the primate endometrium by progesterone and embryonic stimuli. Reproduction. 2007;134(3):513–23.CrossRefPubMed
14.
Zurück zum Zitat Nimbkar-Joshi S, Katkam RR, Chaudhari UK, Jacob S, Manjramkar DD, Metkari SM, et al. Endometrial epithelial cell modifications in response to embryonic signals in bonnet monkeys (Macaca radiata). Histochem Cell Biol. 2012;138:289–304.CrossRefPubMed Nimbkar-Joshi S, Katkam RR, Chaudhari UK, Jacob S, Manjramkar DD, Metkari SM, et al. Endometrial epithelial cell modifications in response to embryonic signals in bonnet monkeys (Macaca radiata). Histochem Cell Biol. 2012;138:289–304.CrossRefPubMed
15.
Zurück zum Zitat Modi DN, Godbole G, Suman P, Gupta SK. Endometrial biology during trophoblast invasion. Front Biosci (Schol Ed). 2012;4:1151–71. Modi DN, Godbole G, Suman P, Gupta SK. Endometrial biology during trophoblast invasion. Front Biosci (Schol Ed). 2012;4:1151–71.
16.
Zurück zum Zitat Fazleabas AT, Donnelly KM, Srinivasan S, Fortman JD, Miller JB. Modulation of the baboon (Papio anubis) uterine endometrium by chorionic gonadotrophin during the period of uterine receptivity. Proc Natl Acad Sci U S A. 1999;96:2543–8.CrossRefPubMedPubMedCentral Fazleabas AT, Donnelly KM, Srinivasan S, Fortman JD, Miller JB. Modulation of the baboon (Papio anubis) uterine endometrium by chorionic gonadotrophin during the period of uterine receptivity. Proc Natl Acad Sci U S A. 1999;96:2543–8.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Strakova Z, Mavrogianis P, Meng X, Hastings JM, Jackson KS, Cameo P, et al. In vivo infusion of interleukin-1β and chorionic gonadotropin induces endometrial changes that mimic early pregnancy events in the baboon. Endocrinol. 2005;146:4097–104.CrossRef Strakova Z, Mavrogianis P, Meng X, Hastings JM, Jackson KS, Cameo P, et al. In vivo infusion of interleukin-1β and chorionic gonadotropin induces endometrial changes that mimic early pregnancy events in the baboon. Endocrinol. 2005;146:4097–104.CrossRef
18.
Zurück zum Zitat Koot YE, Van Hooff SR, Boomsma CM, Van Leenen D, Koerkamp MJ, Goddijn M, et al. An endometrial gene expression signature accurately predicts recurrent implantation failure after IVF. Sci Rep. 2016;6:19411.CrossRefPubMedPubMedCentral Koot YE, Van Hooff SR, Boomsma CM, Van Leenen D, Koerkamp MJ, Goddijn M, et al. An endometrial gene expression signature accurately predicts recurrent implantation failure after IVF. Sci Rep. 2016;6:19411.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Ashary N, Tiwari A, Modi D. Embryo implantation: war in times of love. Endocrin. 2018;159:1188–98.CrossRef Ashary N, Tiwari A, Modi D. Embryo implantation: war in times of love. Endocrin. 2018;159:1188–98.CrossRef
21.
Zurück zum Zitat Tu Z, Ran H, Zhang S, Xia G, Wang B, Wang H. Molecular determinants of uterine receptivity. Int J Dev Biol. 2014;58:147–54.CrossRefPubMed Tu Z, Ran H, Zhang S, Xia G, Wang B, Wang H. Molecular determinants of uterine receptivity. Int J Dev Biol. 2014;58:147–54.CrossRefPubMed
22.
Zurück zum Zitat Lee CL, Lam KK, Vijayan M, Koistinen H, Seppala M, Ng EH, et al. The pleiotropic effect of Glycodelin-A in early pregnancy. Am J Reprod Immunol. 2016;75:290–7.CrossRefPubMed Lee CL, Lam KK, Vijayan M, Koistinen H, Seppala M, Ng EH, et al. The pleiotropic effect of Glycodelin-A in early pregnancy. Am J Reprod Immunol. 2016;75:290–7.CrossRefPubMed
23.
Zurück zum Zitat Bastu E, Mutlu MF, Yasa C, Dural O, Aytan AN, Celik C, et al. Role of Mucin 1 and Glycodelin A in recurrent implantation failure. Fertilit Steril. 2015;103:1059–64.CrossRef Bastu E, Mutlu MF, Yasa C, Dural O, Aytan AN, Celik C, et al. Role of Mucin 1 and Glycodelin A in recurrent implantation failure. Fertilit Steril. 2015;103:1059–64.CrossRef
24.
Zurück zum Zitat Focarelli R, Luddi A, De Leo V, Capaldo A, Stendardi A, Pavone V, Benincasa L, Belmonte G, Petraglia F, Piomboni P. Dysregulation of GdA expression in endometrium of women with endometriosis: implication for endometrial receptivity. Reprod Sci 2017;1933719117718276. Focarelli R, Luddi A, De Leo V, Capaldo A, Stendardi A, Pavone V, Benincasa L, Belmonte G, Petraglia F, Piomboni P. Dysregulation of GdA expression in endometrium of women with endometriosis: implication for endometrial receptivity. Reprod Sci 2017;1933719117718276.
25.
26.
Zurück zum Zitat Feng Y, Ma X, Deng L, Yao B, Xiong Y, Wu Y, et al. Role of selectins and their ligands in human implantation stage. Glycobiology. 2017;27(5):385–91.PubMed Feng Y, Ma X, Deng L, Yao B, Xiong Y, Wu Y, et al. Role of selectins and their ligands in human implantation stage. Glycobiology. 2017;27(5):385–91.PubMed
27.
Zurück zum Zitat Natraj U, Bhatt P, Vanage G, Moodbidri SB. Overexpression of monkey oviductal protein: purification and characterization of recombinant protein and its antibodies. Biol Reprod. 2002;67:1897–906.CrossRefPubMed Natraj U, Bhatt P, Vanage G, Moodbidri SB. Overexpression of monkey oviductal protein: purification and characterization of recombinant protein and its antibodies. Biol Reprod. 2002;67:1897–906.CrossRefPubMed
28.
Zurück zum Zitat Bhatt P, Kadam K, Saxena A, Natraj U. Fertilization, embryonic development and oviductal environment: role of estrogen induced oviductal glycoprotein. Indian J Exp Biol. 2004;42:1043–55.PubMed Bhatt P, Kadam K, Saxena A, Natraj U. Fertilization, embryonic development and oviductal environment: role of estrogen induced oviductal glycoprotein. Indian J Exp Biol. 2004;42:1043–55.PubMed
29.
Zurück zum Zitat Buhi WC. Characterization and biological roles of oviduct-specific, oestrogen-dependent glycoprotein. Reproduction. 2002;123:355–62.CrossRefPubMed Buhi WC. Characterization and biological roles of oviduct-specific, oestrogen-dependent glycoprotein. Reproduction. 2002;123:355–62.CrossRefPubMed
30.
Zurück zum Zitat Choudhary S, Kumaresan A, Kumar M, Chhillar S, Malik H, Kumar S, et al. Effect of recombinant and native buffalo OVGP1 on sperm functions and in vitro embryo development: a comparative study. J Anim Sci Biotechnol. 2017;8:69.CrossRefPubMedPubMedCentral Choudhary S, Kumaresan A, Kumar M, Chhillar S, Malik H, Kumar S, et al. Effect of recombinant and native buffalo OVGP1 on sperm functions and in vitro embryo development: a comparative study. J Anim Sci Biotechnol. 2017;8:69.CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Kobayashi A, Behringer RR. Developmental genetics of the female reproductive tract in mammals. Nat Rev Genet. 2003;4:969–80.CrossRefPubMed Kobayashi A, Behringer RR. Developmental genetics of the female reproductive tract in mammals. Nat Rev Genet. 2003;4:969–80.CrossRefPubMed
32.
Zurück zum Zitat Laheri S, Modi D, Bhatt P. Extra-oviductal expression of oviductal glycoprotein 1 in mouse: detection in testis, epididymis and ovary. J Biosci. 2017;42:69–80.CrossRefPubMed Laheri S, Modi D, Bhatt P. Extra-oviductal expression of oviductal glycoprotein 1 in mouse: detection in testis, epididymis and ovary. J Biosci. 2017;42:69–80.CrossRefPubMed
33.
Zurück zum Zitat Roux E, Bleau G, Kan FW. Fate of hamster oviductin in the oviduct and uterus during early gestation. Molecular Reproduction and Development Mol Reprod Dev. 1997;46:306–17.CrossRefPubMed Roux E, Bleau G, Kan FW. Fate of hamster oviductin in the oviduct and uterus during early gestation. Molecular Reproduction and Development Mol Reprod Dev. 1997;46:306–17.CrossRefPubMed
34.
Zurück zum Zitat Uchida H, Maruyama T, Nishikawa-Uchida S, Oda H, Miyazaki K, Yamasaki A, et al. Studies using an in vitro model show evidence of involvement of epithelial-mesenchymal transition of human endometrial epithelial cells in human embryo implantation. J Biol Chem. 2012;287:4441–50.CrossRefPubMed Uchida H, Maruyama T, Nishikawa-Uchida S, Oda H, Miyazaki K, Yamasaki A, et al. Studies using an in vitro model show evidence of involvement of epithelial-mesenchymal transition of human endometrial epithelial cells in human embryo implantation. J Biol Chem. 2012;287:4441–50.CrossRefPubMed
35.
Zurück zum Zitat Ruane PT, Berneau SC, Koeck R, Watts J, Kimber SJ, Brison DR, et al. Apposition to endometrial epithelial cells activates mouse blastocysts for implantation. Mol Hum Reprod. 2017;23:617–27.CrossRefPubMed Ruane PT, Berneau SC, Koeck R, Watts J, Kimber SJ, Brison DR, et al. Apposition to endometrial epithelial cells activates mouse blastocysts for implantation. Mol Hum Reprod. 2017;23:617–27.CrossRefPubMed
36.
Zurück zum Zitat Godbole G, Modi D. Regulation of decidualization, interleukin-11 and interleukin-15 by homeobox A 10 in endometrial stromal cells. J Reprod Immunol. 2010;85:130–9.CrossRefPubMed Godbole G, Modi D. Regulation of decidualization, interleukin-11 and interleukin-15 by homeobox A 10 in endometrial stromal cells. J Reprod Immunol. 2010;85:130–9.CrossRefPubMed
37.
Zurück zum Zitat Godbole G, Suman P, Malik A, Galvankar M, Joshi N, Fazleabas A, et al. Decrease in expression of HOXA10 in the decidua after embryo implantation promotes trophoblast invasion. Endocrin. 2017;158:2618–33.CrossRef Godbole G, Suman P, Malik A, Galvankar M, Joshi N, Fazleabas A, et al. Decrease in expression of HOXA10 in the decidua after embryo implantation promotes trophoblast invasion. Endocrin. 2017;158:2618–33.CrossRef
38.
39.
Zurück zum Zitat Germeyer A, Savaris RF, Jauckus J, Lessey B. Endometrial beta3 integrin profile reflects endometrial receptivity defects in women with unexplained recurrent pregnancy loss. Reprod Biol Endocrinol. 2014;12(1):53.CrossRefPubMedPubMedCentral Germeyer A, Savaris RF, Jauckus J, Lessey B. Endometrial beta3 integrin profile reflects endometrial receptivity defects in women with unexplained recurrent pregnancy loss. Reprod Biol Endocrinol. 2014;12(1):53.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Elnaggar A, Farag AH, Gaber ME, Hafeez MA, Ali MS, Atef AM. AlphaVBeta3 integrin expression within uterine endometrium in unexplained infertility: a prospective cohort study. BMC Womens Health. 2017;17:90.CrossRefPubMedPubMedCentral Elnaggar A, Farag AH, Gaber ME, Hafeez MA, Ali MS, Atef AM. AlphaVBeta3 integrin expression within uterine endometrium in unexplained infertility: a prospective cohort study. BMC Womens Health. 2017;17:90.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Modi D, Godbole G. HOXA10 signals on the highway through pregnancy. J Reprod Immunol. 2009;83:72–8.CrossRefPubMed Modi D, Godbole G. HOXA10 signals on the highway through pregnancy. J Reprod Immunol. 2009;83:72–8.CrossRefPubMed
42.
Zurück zum Zitat Du H, Taylor HS. The role of Hox genes in female reproductive tract development, adult function, and fertility. Cold Spring Harb Perspect Med. 2016;6(1):a023002.CrossRefPubMedCentral Du H, Taylor HS. The role of Hox genes in female reproductive tract development, adult function, and fertility. Cold Spring Harb Perspect Med. 2016;6(1):a023002.CrossRefPubMedCentral
43.
Zurück zum Zitat Bagot CN, Kliman HJ, Taylor HS. Maternal Hoxa10 is required for pinopod formation in the development of mouse uterine receptivity to embryo implantation. Dev Dyn. 2001;222:538–44.CrossRefPubMed Bagot CN, Kliman HJ, Taylor HS. Maternal Hoxa10 is required for pinopod formation in the development of mouse uterine receptivity to embryo implantation. Dev Dyn. 2001;222:538–44.CrossRefPubMed
44.
Zurück zum Zitat Daftary GS, Troy PJ, Bagot CN, Young SL, Taylor HS. Direct regulation of β3-integrin subunit gene expression by HOXA10 in endometrial cells. Mol Endocrinol. 2002;16(3):571–9.PubMed Daftary GS, Troy PJ, Bagot CN, Young SL, Taylor HS. Direct regulation of β3-integrin subunit gene expression by HOXA10 in endometrial cells. Mol Endocrinol. 2002;16(3):571–9.PubMed
45.
Zurück zum Zitat Singh M, Chaudhry P, Asselin E. Bridging endometrial receptivity and implantation: network of hormones, cytokines, and growth factors. J Endocrinol. 2011;210(1):5–14.CrossRefPubMed Singh M, Chaudhry P, Asselin E. Bridging endometrial receptivity and implantation: network of hormones, cytokines, and growth factors. J Endocrinol. 2011;210(1):5–14.CrossRefPubMed
46.
Zurück zum Zitat Sharma S, Godbole G, Modi D. Decidual control of trophoblast invasion. Am J Reprod Immunol. 2016;75:341–50.CrossRefPubMed Sharma S, Godbole G, Modi D. Decidual control of trophoblast invasion. Am J Reprod Immunol. 2016;75:341–50.CrossRefPubMed
47.
Zurück zum Zitat Dimitriadis E, Menkhorst E, Salamonsen LA, Paiva PLIF. IL11 in trophoblast-endometrial interactions during the establishment of pregnancy. Placenta. 2010;31:99–104.CrossRef Dimitriadis E, Menkhorst E, Salamonsen LA, Paiva PLIF. IL11 in trophoblast-endometrial interactions during the establishment of pregnancy. Placenta. 2010;31:99–104.CrossRef
48.
Zurück zum Zitat Shuya LL, Menkhorst EM, Yap J, Li P, Lane N, Dimitriadis E. Leukemia inhibitory factor enhances endometrial stromal cell decidualization in humans and mice. PLoS One. 2011;6:e25288.CrossRefPubMedPubMedCentral Shuya LL, Menkhorst EM, Yap J, Li P, Lane N, Dimitriadis E. Leukemia inhibitory factor enhances endometrial stromal cell decidualization in humans and mice. PLoS One. 2011;6:e25288.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Gellersen B, Brosens JJ. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocr Rev. 2014;35:851–905.CrossRefPubMed Gellersen B, Brosens JJ. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocr Rev. 2014;35:851–905.CrossRefPubMed
50.
Zurück zum Zitat Gupta SK, Malhotra SS, Malik A, Verma S, Chaudhary P. Cell signaling pathways involved during invasion and syncytialization of trophoblast cells. Am J Reprod Immunol. 2016;75:361–71.CrossRefPubMed Gupta SK, Malhotra SS, Malik A, Verma S, Chaudhary P. Cell signaling pathways involved during invasion and syncytialization of trophoblast cells. Am J Reprod Immunol. 2016;75:361–71.CrossRefPubMed
51.
Zurück zum Zitat Salamonsen LA, Evans J, Nguyen HPT, Edgell TA. The microenvironment of human implantation: determinant of reproductive success. Am J Reprod Immunol. 2016;75:218–25.CrossRefPubMed Salamonsen LA, Evans J, Nguyen HPT, Edgell TA. The microenvironment of human implantation: determinant of reproductive success. Am J Reprod Immunol. 2016;75:218–25.CrossRefPubMed
52.
Zurück zum Zitat Alexander CM, Hansell EJ, Behrendtsen O, Flannery ML, Kishnani NS, Hawkes SP, et al. Expression and function of matrix metalloproteinases and their inhibitors at the maternal-embryonic boundary during mouse embryo implantation. Development. 1996;122:1723–36.PubMed Alexander CM, Hansell EJ, Behrendtsen O, Flannery ML, Kishnani NS, Hawkes SP, et al. Expression and function of matrix metalloproteinases and their inhibitors at the maternal-embryonic boundary during mouse embryo implantation. Development. 1996;122:1723–36.PubMed
53.
Zurück zum Zitat Araki Y, Nohara M, Yoshida-Komiya H, Kuramochi T, Mamoru IT, Hoshi H, et al. Effect of a null mutation of the oviduct-specific glycoprotein gene on mouse fertilization. Biochem J. 2003;374:551–7.CrossRefPubMedPubMedCentral Araki Y, Nohara M, Yoshida-Komiya H, Kuramochi T, Mamoru IT, Hoshi H, et al. Effect of a null mutation of the oviduct-specific glycoprotein gene on mouse fertilization. Biochem J. 2003;374:551–7.CrossRefPubMedPubMedCentral
Metadaten
Titel
Oviductal glycoprotein 1 (OVGP1) is expressed by endometrial epithelium that regulates receptivity and trophoblast adhesion
verfasst von
Saniya Laheri
Nancy Ashary
Purvi Bhatt
Deepak Modi
Publikationsdatum
30.06.2018
Verlag
Springer US
Erschienen in
Journal of Assisted Reproduction and Genetics / Ausgabe 8/2018
Print ISSN: 1058-0468
Elektronische ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-018-1231-4

Weitere Artikel der Ausgabe 8/2018

Journal of Assisted Reproduction and Genetics 8/2018 Zur Ausgabe

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.