Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Reproductive performance: at the cross-road of genetics, technologies and environment

Daniel Vaiman
+ Author Affiliations
- Author Affiliations

Institut Cochin, U1016 INSERM, UMR8104 CNRS, 24, rue du Faubourg St Jacques, 75014, Paris, France. Email: daniel.vaiman@inserm.fr

Reproduction, Fertility and Development 27(1) 1-13 https://doi.org/10.1071/RD14316
Published: 4 December 2014

Abstract

Sexual reproduction depends on a negotiation between the sexes at the level of the cells (gametes), tissue (trophectoderm of the blastocyst and endometrium in the uterus) and organisms (to allow sexual intercourse). This review evaluates new questions linked to sexual reproduction in the biosphere in the context of the 21st century, in light of current knowledge in genetics and epigenetics. It presents the challenge of ‘forcing reproductive efficiency’ using ineffective gametes, or despite other fertility problems, through medically assisted reproduction and presents the reproductive challenge of high production farm animals, which are in a situation of chronically negative energy balance. It also analyses the situation created by the release of endocrine disruptors into the environment and discusses the possible transgenerational consequences of environmental modifications linked to these compounds.

Additional keywords: endocrine disrupters, epigenetics, fertility, high-producing dairy cows, medically assisted reproduction, QSAR modelling.


References

Adams, I. R., and McLaren, A. (2002). Sexually dimorphic development of mouse primordial germ cells: switching from oogenesis to spermatogenesis. Development 129, 1155–1164.
| 1:CAS:528:DC%2BD38XisVWrur0%3D&md5=263eceb3f9e6c96c7c87279a6c952355CAS | 11874911PubMed |

Anttonen, M., Pihlajoki, M., Andersson, N., Georges, A., L’Hote, D., Vattulainen, S., Farkkila, A., Unkila-Kallio, L., Veitia, R. A., and Heikinheimo, M. (2014). FOXL2, GATA4, and SMAD3 co-operatively modulate gene expression, cell viability and apoptosis in ovarian granulosa cell tumor cells. PLoS ONE 9, e85545.
FOXL2, GATA4, and SMAD3 co-operatively modulate gene expression, cell viability and apoptosis in ovarian granulosa cell tumor cells.Crossref | GoogleScholarGoogle Scholar | 24416423PubMed |

Anway, M. D., Cupp, A. S., Uzumcu, M., and Skinner, M. K. (2005). Epigenetic transgenerational actions of endocrine disruptors and male fertility. Science 308, 1466–1469.
Epigenetic transgenerational actions of endocrine disruptors and male fertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXks1eqsbY%3D&md5=05d42e23f3872a7670bad89c3f6eb436CAS | 15933200PubMed |

ASRM Office of Public Affairs (2013). Five million babies born with help of assisted reproductive technologies. Available at http://www.asrm.org/Five_Million_Babies_Born_with_Help_of_Assisted_Reproductive_Technologies/ [Verified 29 August 2014]

Auger, J., Kunstmann, J. M., Czyglik, F., and Jouannet, P. (1995). Decline in semen quality among fertile men in Paris during the past 20 years. N. Engl. J. Med. 332, 281–285.
Decline in semen quality among fertile men in Paris during the past 20 years.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2M7hsFOitg%3D%3D&md5=994b2c179a8aaa0d11e386881ef99f13CAS | 7816062PubMed |

Auger, J., Eustache, F., Maceiras, P., Broussard, C., Chafey, P., Lesaffre, C., Vaiman, D., Camoin, L., and Auer, J. (2010). Modified expression of several sperm proteins after chronic exposure to the antiandrogenic compound vinclozolin. Toxicol. Sci. 117, 475–484.
Modified expression of several sperm proteins after chronic exposure to the antiandrogenic compound vinclozolin.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhtFyltb3K&md5=a64ddf792c2b96dc9100e78433bef7c1CAS | 20616205PubMed |

Auger, J., Le Denmat, D., Berges, R., Doridot, L., Salmon, B., Canivenc-Lavier, M. C., and Eustache, F. (2013). Environmental levels of oestrogenic and antiandrogenic compounds feminize digit ratios in male rats and their unexposed male progeny. Proc. Biol. Sci. 280, 20131532.
Environmental levels of oestrogenic and antiandrogenic compounds feminize digit ratios in male rats and their unexposed male progeny.Crossref | GoogleScholarGoogle Scholar | 23926155PubMed |

Avenarius, M. R., Hildebrand, M. S., Zhang, Y., Meyer, N. C., Smith, L. L., Kahrizi, K., Najmabadi, H., and Smith, R. J. (2009). Human male infertility caused by mutations in the CATSPER1 channel protein. Am. J. Hum. Genet. 84, 505–510.
Human male infertility caused by mutations in the CATSPER1 channel protein.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXmvVahsLo%3D&md5=2b38bc817852581702bca054c4f1745aCAS | 19344877PubMed |

Barbaux, S., Gascoin-Lachambre, G., Buffat, C., Monnier, P., Mondon, F., Tonanny, M. B., Pinard, A., Auer, J., Bessieres, B., Barlier, A., Jacques, S., Simeoni, U., Dandolo, L., Letourneur, F., Jammes, H., and Vaiman, D. (2012). A genome-wide approach reveals novel imprinted genes expressed in the human placenta. Epigenetics 7, 1079–1090.
A genome-wide approach reveals novel imprinted genes expressed in the human placenta.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhs1aqs7Y%3D&md5=54d2e8ce64d8ccfa795d7be588615848CAS | 22894909PubMed |

Batista, F., Vaiman, D., Dausset, J., Fellous, M., and Veitia, R. A. (2007). Potential targets of FOXL2, a transcription factor involved in craniofacial and follicular development, identified by transcriptomics. Proc. Natl Acad. Sci. USA 104, 3330–3335.
Potential targets of FOXL2, a transcription factor involved in craniofacial and follicular development, identified by transcriptomics.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXjtVWlt74%3D&md5=eb4aaa97089fbf15c3cf447a615b20b3CAS | 17360647PubMed |

Baumann, C. A., Brady, M. J., and Saltiel, A. R. (2001). Activation of glycogen synthase by insulin in 3T3-L1 adipocytes involves c-Cbl-associating protein (CAP)-dependent and CAP-independent signaling pathways. J. Biol. Chem. 276, 6065–6068.
Activation of glycogen synthase by insulin in 3T3-L1 adipocytes involves c-Cbl-associating protein (CAP)-dependent and CAP-independent signaling pathways.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXhslShs7Y%3D&md5=338912405b48b647afe93e12c0fed01aCAS | 11222622PubMed |

Benayoun, B. A., Georges, A. B., L’Hote, D., Andersson, N., Dipietromaria, A., Todeschini, A. L., Caburet, S., Bazin, C., Anttonen, M., and Veitia, R. A. (2011). Transcription factor FOXL2 protects granulosa cells from stress and delays cell cycle: role of its regulation by the SIRT1 deacetylase. Hum. Mol. Genet. 20, 1673–1686.
Transcription factor FOXL2 protects granulosa cells from stress and delays cell cycle: role of its regulation by the SIRT1 deacetylase.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXksFSrsr8%3D&md5=d5f5df3248229344e489530144ab48a6CAS | 21289058PubMed |

Berta, P., Hawkins, J. R., Sinclair, A. H., Taylor, A., Griffiths, B. L., Goodfellow, P. N., and Fellous, M. (1990). Genetic evidence equating SRY and the testis-determining factor. Nature 348, 448–450.
Genetic evidence equating SRY and the testis-determining factor.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXhvVOnsg%3D%3D&md5=81fd199c80963b2e8f042af035090471CAS | 2247149PubMed |

Boissonnas, C. C., Abdalaoui, H. E., Haelewyn, V., Fauque, P., Dupont, J. M., Gut, I., Vaiman, D., Jouannet, P., Tost, J., and Jammes, H. (2010). Specific epigenetic alterations of IGF2–H19 locus in spermatozoa from infertile men. Eur. J. Hum. Genet. 18, 73–80.
Specific epigenetic alterations of IGF2–H19 locus in spermatozoa from infertile men.Crossref | GoogleScholarGoogle Scholar | 19584898PubMed |

Borghese, B., Santulli, P., Hequet, D., Pierre, G., de Ziegler, D., Vaiman, D., and Chapron, C. (2012). Genetic polymorphisms of DNMT3L involved in hypermethylation of chromosomal ends are associated with greater risk of developing ovarian endometriosis. Am. J. Pathol. 180, 1781–1786.
Genetic polymorphisms of DNMT3L involved in hypermethylation of chromosomal ends are associated with greater risk of developing ovarian endometriosis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XnsF2ks7s%3D&md5=0d6688244f7ec3b0c69f83f0027e5a22CAS | 22401780PubMed |

Boulanger, L., Pannetier, M., Gall, L., Allais-Bonnet, A., Elzaiat, M., Le Bourhis, D., Daniel, N., Richard, C., Cotinot, C., Ghyselinck, N. B., and Pailhoux, E. (2014). FOXL2 is a female sex-determining gene in the goat. Curr. Biol. 24, 404–408.
FOXL2 is a female sex-determining gene in the goat.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhs1ags74%3D&md5=02d655666fa799d1c9127326b930f1b6CAS | 24485832PubMed |

Caburet, S., Arboleda, V. A., Llano, E., Overbeek, P. A., Barbero, J. L., Oka, K., Harrison, W., Vaiman, D., Ben-Neriah, Z., Garcia-Tunon, I., Fellous, M., Pendas, A. M., Veitia, R. A., and Vilain, E. (2014). Mutant cohesin in premature ovarian failure. N. Engl. J. Med. 370, 943–949.
Mutant cohesin in premature ovarian failure.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXkt1ehsb8%3D&md5=5169b4315a381426131196289bceb18cCAS | 24597867PubMed |

Carone, B. R., Fauquier, L., Habib, N., Shea, J. M., Hart, C. E., Li, R., Bock, C., Li, C., Gu, H., Zamore, P. D., Meissner, A., Weng, Z., Hofmann, H. A., Friedman, N., and Rando, O. J. (2010). Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143, 1084–1096.
Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhs1agtrbE&md5=3f6304745f8d786be525e7342b7d3547CAS | 21183072PubMed |

Ceelen, M., van Weissenbruch, M. M., Vermeiden, J. P., van Leeuwen, F. E., and Delemarre-van de Waal, H. A. (2008). Cardiometabolic differences in children born after in vitro fertilization: follow-up study. J. Clin. Endocrinol. Metab. 93, 1682–1688.
Cardiometabolic differences in children born after in vitro fertilization: follow-up study.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXlvFWgs78%3D&md5=6e00b523509eb2e509a1be8f2b598dd8CAS | 18285409PubMed |

Ceelen, M., van Weissenbruch, M. M., Prein, J., Smit, J. J., Vermeiden, J. P., Spreeuwenberg, M., van Leeuwen, F. E., and Delemarre-van de Waal, H. A. (2009). Growth during infancy and early childhood in relation to blood pressure and body fat measures at age 8–18 years of IVF children and spontaneously conceived controls born to subfertile parents. Hum. Reprod. 24, 2788–2795.
Growth during infancy and early childhood in relation to blood pressure and body fat measures at age 8–18 years of IVF children and spontaneously conceived controls born to subfertile parents.Crossref | GoogleScholarGoogle Scholar | 19648588PubMed |

Celton-Morizur, S., Merlen, G., Couton, D., Margall-Ducos, G., and Desdouets, C. (2009). The insulin/Akt pathway controls a specific cell division program that leads to generation of binucleated tetraploid liver cells in rodents. J. Clin. Invest. 119, 1880–1887.
| 1:CAS:528:DC%2BD1MXosVCku7c%3D&md5=d456f4959c33461f0d85cf5c6517e917CAS | 19603546PubMed |

Chagas, L. M., Bass, J. J., Blache, D., Burke, C. R., Kay, J. K., Lindsay, D. R., Lucy, M. C., Martin, G. B., Meier, S., Rhodes, F. M., Roche, J. R., Thatcher, W. W., and Webb, R. (2007). Invited review: new perspectives on the roles of nutrition and metabolic priorities in the subfertility of high-producing dairy cows. J. Dairy Sci. 90, 4022–4032.
Invited review: new perspectives on the roles of nutrition and metabolic priorities in the subfertility of high-producing dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXpsleqtb0%3D&md5=f750a15cbfb912fa9956938158ffbecaCAS | 17699018PubMed |

Chaouat, G., Zourbas, S., Ostojic, S., Lappree-Delage, G., Dubanchet, S., Ledee, N., and Martal, J. (2002). A brief review of recent data on some cytokine expressions at the materno–foetal interface which might challenge the classical Th1/Th2 dichotomy. J. Reprod. Immunol. 53, 241–256.
A brief review of recent data on some cytokine expressions at the materno–foetal interface which might challenge the classical Th1/Th2 dichotomy.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXos1Kjt78%3D&md5=6894062eb567eb49437d95cb8904853bCAS | 11730920PubMed |

Chassot, A. A., Ranc, F., Gregoire, E. P., Roepers-Gajadien, H. L., Taketo, M. M., Camerino, G., de Rooij, D. G., Schedl, A., and Chaboissier, M. C. (2008). Activation of beta-catenin signaling by Rspo1 controls differentiation of the mammalian ovary. Hum. Mol. Genet. 17, 1264–1277.
Activation of beta-catenin signaling by Rspo1 controls differentiation of the mammalian ovary.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXkvVOhurw%3D&md5=e1bff11f720fe3d43b94fb0a92f7d584CAS | 18250098PubMed |

Clark, D. A., Chaouat, G., Guenet, J. L., and Kiger, N. (1987). Local active suppression and successful vaccination against spontaneous abortion in CBA/J mice. J. Reprod. Immunol. 10, 79–85.
Local active suppression and successful vaccination against spontaneous abortion in CBA/J mice.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL2s3isF2huw%3D%3D&md5=2b714d7a9d109dea4e9644ae033c8cfdCAS | 2953893PubMed |

Cocquet, J., Ellis, P. J., Yamauchi, Y., Mahadevaiah, S. K., Affara, N. A., Ward, M. A., and Burgoyne, P. S. (2009). The multicopy gene Sly represses the sex chromosomes in the male mouse germline after meiosis. PLoS Biol. 7, e1000244.
The multicopy gene Sly represses the sex chromosomes in the male mouse germline after meiosis.Crossref | GoogleScholarGoogle Scholar | 19918361PubMed |

Cocquet, J., Ellis, P. J., Mahadevaiah, S. K., Affara, N. A., Vaiman, D., and Burgoyne, P. S. (2012). A genetic basis for a postmeiotic X versus Y chromosome intragenomic conflict in the mouse. PLoS Genet. 8, e1002900.
A genetic basis for a postmeiotic X versus Y chromosome intragenomic conflict in the mouse.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhsVSjtLrF&md5=5f92e05d3dcd664c2a45ffc462138d8eCAS | 23028340PubMed |

Colucci-Guyon, E., Portier, M. M., Dunia, I., Paulin, D., Pournin, S., and Babinet, C. (1994). Mice lacking vimentin develop and reproduce without an obvious phenotype. Cell 79, 679–694.
Mice lacking vimentin develop and reproduce without an obvious phenotype.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXit1Gksrk%3D&md5=e2937292baa43d015b716d212000802dCAS | 7954832PubMed |

Combes, R., Scott, K., Dillon, D., Meredith, C., McAdam, K., and Proctor, C. (2012). The effect of a novel tobacco process on the in vitro cytotoxicity and genotoxicity of cigarette smoke particulate matter. Toxicol. In Vitro 26, 1022–1029.
The effect of a novel tobacco process on the in vitro cytotoxicity and genotoxicity of cigarette smoke particulate matter.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XmvFCgt7g%3D&md5=1dce9e9f9c81f4a2af998f757569f9f9CAS | 22542757PubMed |

Constância, M., Kelsey, G., and Reik, W. (2004). Resourceful imprinting. Nature 432, 53–57.
Resourceful imprinting.Crossref | GoogleScholarGoogle Scholar | 15525980PubMed |

Cooper, T. G., Noonan, E., von Eckardstein, S., Auger, J., Baker, H. W., Behre, H. M., Haugen, T. B., Kruger, T., Wang, C., Mbizvo, M. T., and Vogelsong, K. M. (2010). World Health Organization reference values for human semen characteristics. Hum. Reprod. Update 16, 231–245.
World Health Organization reference values for human semen characteristics.Crossref | GoogleScholarGoogle Scholar | 19934213PubMed |

Crisponi, L., Deiana, M., Loi, A., Chiappe, F., Uda, M., Amati, P., Bisceglia, L., Zelante, L., Nagaraja, R., Porcu, S., Ristaldi, M. S., Marzella, R., Rocchi, M., Nicolino, M., Lienhardt-Roussie, A., Nivelon, A., Verloes, A., Schlessinger, D., Gasparini, P., Bonneau, D., Cao, A., and Pilia, G. (2001). The putative forkhead transcription factor FOXL2 is mutated in blepharophimosis/ptosis/epicanthus inversus syndrome. Nat. Genet. 27, 159–166.
The putative forkhead transcription factor FOXL2 is mutated in blepharophimosis/ptosis/epicanthus inversus syndrome.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXhtFGktLs%3D&md5=be63752760be0d10beeb8531bfdb41a6CAS | 11175783PubMed |

Crosier, A. E., Comizzoli, P., Baker, T., Davidson, A., Munson, L., Howard, J., Marker, L. L., and Wildt, D. E. (2011). Increasing age influences uterine integrity, but not ovarian function or oocyte quality, in the cheetah (Acinonyx jubatus). Biol. Reprod. 85, 243–253.
Increasing age influences uterine integrity, but not ovarian function or oocyte quality, in the cheetah (Acinonyx jubatus).Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXpslOntbs%3D&md5=7a73aac49cacf1ee3db097dd33c6edcdCAS | 21565998PubMed |

Dam, A. H., Koscinski, I., Kremer, J. A., Moutou, C., Jaeger, A. S., Oudakker, A. R., Tournaye, H., Charlet, N., Lagier-Tourenne, C., van Bokhoven, H., and Viville, S. (2007). Homozygous mutation in SPATA16 is associated with male infertility in human globozoospermia. Am. J. Hum. Genet. 81, 813–820.
Homozygous mutation in SPATA16 is associated with male infertility in human globozoospermia.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtFSktrjO&md5=219815b92aed07e028f576719f406c88CAS | 17847006PubMed |

De Baere, E., Beysen, D., Oley, C., Lorenz, B., Cocquet, J., De Sutter, P., Devriendt, K., Dixon, M., Fellous, M., Fryns, J. P., Garza, A., Jonsrud, C., Koivisto, P. A., Krause, A., Leroy, B. P., Meire, F., Plomp, A., Van Maldergem, L., De Paepe, A., Veitia, R., and Messiaen, L. (2003). FOXL2 and BPES: mutational hotspots, phenotypic variability, and revision of the genotype–phenotype correlation. Am. J. Hum. Genet. 72, 478–487.
FOXL2 and BPES: mutational hotspots, phenotypic variability, and revision of the genotype–phenotype correlation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXht12rtLs%3D&md5=818889a200be826a7855facc402c894bCAS | 12529855PubMed |

de Vries, M. J., and Veerkamp, R. F. (2000). Energy balance of dairy cattle in relation to milk production variables and fertility. J. Dairy Sci. 83, 62–69.
Energy balance of dairy cattle in relation to milk production variables and fertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXotVOntw%3D%3D&md5=65ce526be8dc623a42d5b8aea4c7e0c7CAS | 10659965PubMed |

de Ziegler, D., Borghese, B., and Chapron, C. (2010). Endometriosis and infertility: pathophysiology and management. Lancet 376, 730–738.
Endometriosis and infertility: pathophysiology and management.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhtVymt7zI&md5=5095bd861a1e70163e351aee8a01a8abCAS | 20801404PubMed |

Dieterich, K., Soto Rifo, R., Faure, A. K., Hennebicq, S., Ben Amar, B., Zahi, M., Perrin, J., Martinez, D., Sele, B., Jouk, P. S., Ohlmann, T., Rousseaux, S., Lunardi, J., and Ray, P. F. (2007). Homozygous mutation of AURKC yields large-headed polyploid spermatozoa and causes male infertility. Nat. Genet. 39, 661–665.
Homozygous mutation of AURKC yields large-headed polyploid spermatozoa and causes male infertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXksFersb0%3D&md5=1f15d8ec6d1549207d02ec7f0b637d22CAS | 17435757PubMed |

Dieterich, K., Zouari, R., Harbuz, R., Vialard, F., Martinez, D., Bellayou, H., Prisant, N., Zoghmar, A., Guichaoua, M. R., Koscinski, I., Kharouf, M., Noruzinia, M., Nadifi, S., Sefiani, A., Lornage, J., Zahi, M., Viville, S., Sele, B., Jouk, P. S., Jacob, M. C., Escalier, D., Nikas, Y., Hennebicq, S., Lunardi, J., and Ray, P. F. (2009). The aurora kinase C c.144delC mutation causes meiosis I arrest in men and is frequent in the North African population. Hum. Mol. Genet. 18, 1301–1309.
The aurora kinase C c.144delC mutation causes meiosis I arrest in men and is frequent in the North African population.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXjsFSju78%3D&md5=b8b20cfa5c19c7b9e01c3cf5f5a91680CAS | 19147683PubMed |

Dirami, T., Rode, B., Jollivet, M., Da Silva, N., Escalier, D., Gaitch, N., Norez, C., Tuffery, P., Wolf, J. P., Becq, F., Ray, P. F., Dulioust, E., Gacon, G., Bienvenu, T., and Toure, A. (2013). Missense mutations in SLC26A8, encoding a sperm-specific activator of CFTR, are associated with human asthenozoospermia. Am. J. Hum. Genet. 92, 760–766.
Missense mutations in SLC26A8, encoding a sperm-specific activator of CFTR, are associated with human asthenozoospermia.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXlvVCrsbo%3D&md5=cfc68ab3cda03e52ba94b04f53568ccdCAS | 23582645PubMed |

Ellison, C. M., Enoch, S. J., and Cronin, M. T. (2011). A review of the use of in silico methods to predict the chemistry of molecular initiating events related to drug toxicity. Expert Opin. Drug Metab. Toxicol. 7, 1481–1495.
A review of the use of in silico methods to predict the chemistry of molecular initiating events related to drug toxicity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhsV2hsLbP&md5=5c2a9fc03106d752681fbf49daa19917CAS | 22032332PubMed |

Engelbregt, M. J., Houdijk, M. E., Popp-Snijders, C., and Delemarre-van de Waal, H. A. (2000). The effects of intra-uterine growth retardation and postnatal undernutrition on onset of puberty in male and female rats. Pediatr. Res. 48, 803–807.
The effects of intra-uterine growth retardation and postnatal undernutrition on onset of puberty in male and female rats.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD3M7hsVGrtA%3D%3D&md5=120799df390eb7c102e6a5eddc511078CAS | 11102550PubMed |

Eustache, F., Mondon, F., Canivenc-Lavier, M. C., Lesaffre, C., Fulla, Y., Berges, R., Cravedi, J. P., Vaiman, D., and Auger, J. (2009). Chronic dietary exposure to a low-dose mixture of genistein and vinclozolin modifies the reproductive axis, testis transcriptome, and fertility. Environ. Health Perspect. 117, 1272–1279.
Chronic dietary exposure to a low-dose mixture of genistein and vinclozolin modifies the reproductive axis, testis transcriptome, and fertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtVKrtLvE&md5=930dca8c410a6cc9e0035cc24cde58b0CAS | 19672408PubMed |

Fauque, P., Jouannet, P., Lesaffre, C., Ripoche, M. A., Dandolo, L., Vaiman, D., and Jammes, H. (2007). Assisted reproductive technology affects developmental kinetics, H19 imprinting control region methylation and H19 gene expression in individual mouse embryos. BMC Dev. Biol. 7, 116.
Assisted reproductive technology affects developmental kinetics, H19 imprinting control region methylation and H19 gene expression in individual mouse embryos.Crossref | GoogleScholarGoogle Scholar | 17949482PubMed |

Fauque, P., Mondon, F., Letourneur, F., Ripoche, M. A., Journot, L., Barbaux, S., Dandolo, L., Patrat, C., Wolf, J. P., Jouannet, P., Jammes, H., and Vaiman, D. (2010a). In vitro fertilization and embryo culture strongly impact the placental transcriptome in the mouse model. PLoS ONE 5, e9218.
In vitro fertilization and embryo culture strongly impact the placental transcriptome in the mouse model.Crossref | GoogleScholarGoogle Scholar | 20169163PubMed |

Fauque, P., Ripoche, M. A., Tost, J., Journot, L., Gabory, A., Busato, F., Le Digarcher, A., Mondon, F., Gut, I., Jouannet, P., Vaiman, D., Dandolo, L., and Jammes, H. (2010b). Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos. Hum. Mol. Genet. 19, 1779–1790.
Modulation of imprinted gene network in placenta results in normal development of in vitro manipulated mouse embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXksFKns70%3D&md5=016d89aa5f20e920e348302f6fc17499CAS | 20150233PubMed |

Favaro, E., Ramachandran, A., McCormick, R., Gee, H., Blancher, C., Crosby, M., Devlin, C., Blick, C., Buffa, F., Li, J. L., Vojnovic, B., Pires das Neves, R., Glazer, P., Iborra, F., Ivan, M., Ragoussis, J., and Harris, A. L. (2010). MicroRNA-210 regulates mitochondrial free radical response to hypoxia and Krebs cycle in cancer cells by targeting iron sulfur cluster protein ISCU. PLoS ONE 5, e10345.
MicroRNA-210 regulates mitochondrial free radical response to hypoxia and Krebs cycle in cancer cells by targeting iron sulfur cluster protein ISCU.Crossref | GoogleScholarGoogle Scholar | 20436681PubMed |

Flint, S., Markle, T., Thompson, S., and Wallace, E. (2012). Bisphenol A exposure, effects, and policy: a wildlife perspective. J. Environ. Manage. 104, 19–34.
Bisphenol A exposure, effects, and policy: a wildlife perspective.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XmvFeksb0%3D&md5=42cf37285a897cbf92360e4650f3d27aCAS | 22481365PubMed |

Fukuda, T., Fukuda, N., Agostinho, A., Hernandez-Hernandez, A., Kouznetsova, A., and Hoog, C. (2014). STAG3-mediated stabilization of REC8 cohesin complexes promotes chromosome synapsis during meiosis. EMBO J. 33, 1243–1255.
| 1:CAS:528:DC%2BC2cXhtFWgsr%2FE&md5=e193b4b52ab6ed3fd54ce9d30d4213a1CAS | 24797475PubMed |

Girardi, G., Yarilin, D., Thurman, J. M., Holers, V. M., and Salmon, J. E. (2006). Complement activation induces dysregulation of angiogenic factors and causes fetal rejection and growth restriction. J. Exp. Med. 203, 2165–2175.
Complement activation induces dysregulation of angiogenic factors and causes fetal rejection and growth restriction.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XptlSgsL4%3D&md5=de32ac09d8e579590cd0e4b208db7d28CAS | 16923853PubMed |

Goto, K., Kinoshita, A., Takuma, Y., and Ogawa, K. (1990). Fertilisation of bovine oocytes by the injection of immobilised, killed spermatozoa. Vet. Rec. 127, 517–520.
| 1:STN:280:DyaK3M7is12nsg%3D%3D&md5=54ec5e5d91a0986677a593ab9a35e8faCAS | 2281585PubMed |

Grafodatskaya, D., Cytrynbaum, C., and Weksberg, R. (2013). The health risks of ART. EMBO Rep. 14, 129–135.
The health risks of ART.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtFOis7k%3D&md5=d4b66edfc56f0b7a63869443ad8a4ae3CAS | 23337626PubMed |

Grechukhina, O., Petracco, R., Popkhadze, S., Massasa, E., Paranjape, T., Chan, E., Flores, I., Weidhaas, J. B., and Taylor, H. S. (2012). A polymorphism in a let-7 microRNA binding site of KRAS in women with endometriosis. EMBO Mol. Med. 4, 206–217.
A polymorphism in a let-7 microRNA binding site of KRAS in women with endometriosis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XjsVSqs7o%3D&md5=b5d1ae2ddb50eb14dcbdac3572900540CAS | 22307873PubMed |

Hanley, N. A., Hagan, D. M., Clement-Jones, M., Ball, S. G., Strachan, T., Salas-Cortes, L., McElreavey, K., Lindsay, S., Robson, S., Bullen, P., Ostrer, H., and Wilson, D. I. (2000). SRY, SOX9, and DAX1 expression patterns during human sex determination and gonadal development. Mech. Dev. 91, 403–407.
SRY, SOX9, and DAX1 expression patterns during human sex determination and gonadal development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXhsVagtL0%3D&md5=9f522a594d1db1e535f9596dafeb9c65CAS | 10704874PubMed |

Harbuz, R., Zouari, R., Pierre, V., Ben Khelifa, M., Kharouf, M., Coutton, C., Merdassi, G., Abada, F., Escoffier, J., Nikas, Y., Vialard, F., Koscinski, I., Triki, C., Sermondade, N., Schweitzer, T., Zhioua, A., Zhioua, F., Latrous, H., Halouani, L., Ouafi, M., Makni, M., Jouk, P. S., Sele, B., Hennebicq, S., Satre, V., Viville, S., Arnoult, C., Lunardi, J., and Ray, P. F. (2011). A recurrent deletion of DPY19L2 causes infertility in man by blocking sperm head elongation and acrosome formation. Am. J. Hum. Genet. 88, 351–361.
A recurrent deletion of DPY19L2 causes infertility in man by blocking sperm head elongation and acrosome formation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXjtFOht78%3D&md5=a28167191437fa427ab4a4aa3ce99547CAS | 21397064PubMed |

Hart, R., and Norman, R. J. (2013a). The longer-term health outcomes for children born as a result of IVF treatment. Part II: mental health and development outcomes. Hum. Reprod. Update 19, 244–250.
The longer-term health outcomes for children born as a result of IVF treatment. Part II: mental health and development outcomes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXmt1CgsrY%3D&md5=34b2c8df5051e429bab7bc041725031fCAS | 23449643PubMed |

Hart, R., and Norman, R. J. (2013b). The longer-term health outcomes for children born as a result of IVF treatment: Part I: general health outcomes. Hum. Reprod. Update 19, 232–243.
The longer-term health outcomes for children born as a result of IVF treatment: Part I: general health outcomes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXmt1Cgsrc%3D&md5=b7a2c8a426e93aac3c6230c9cefd1e9aCAS | 23449642PubMed |

Hartmann, S., Bergmann, M., Bohle, R. M., Weidner, W., and Steger, K. (2006). Genetic imprinting during impaired spermatogenesis. Mol. Hum. Reprod. 12, 407–411.
Genetic imprinting during impaired spermatogenesis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XmsVeisro%3D&md5=515ca97eac5e7ca1c3ea7f4ea147e426CAS | 16608903PubMed |

Heaton, N. D., and Pryor, J. P. (1990). Vasa aplasia and cystic fibrosis. Br. J. Urol. 66, 538–540.
Vasa aplasia and cystic fibrosis.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK3M%2FmsVGrtQ%3D%3D&md5=a1694931423a20de35918750a88513c4CAS | 2249127PubMed |

Henrion, D., Terzi, F., Matrougui, K., Duriez, M., Boulanger, C. M., Colucci-Guyon, E., Babinet, C., Briand, P., Friedlander, G., Poitevin, P., and Levy, B. I. (1997). Impaired flow-induced dilation in mesenteric resistance arteries from mice lacking vimentin. J. Clin. Invest. 100, 2909–2914.
Impaired flow-induced dilation in mesenteric resistance arteries from mice lacking vimentin.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXotVejtbg%3D&md5=d8b6ac9cf1736fc85745c4fe8a627b58CAS | 9389758PubMed |

Hildebrand, M. S., Avenarius, M. R., Fellous, M., Zhang, Y., Meyer, N. C., Auer, J., Serres, C., Kahrizi, K., Najmabadi, H., Beckmann, J. S., and Smith, R. J. (2010). Genetic male infertility and mutation of CATSPER ion channels. Eur. J. Hum. Genet. 18, 1178–1184.
Genetic male infertility and mutation of CATSPER ion channels.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhtlWmur3O&md5=7ee9abc614f34f98f78c06fe11fe5cb3CAS | 20648059PubMed |

Holt, J. E., Pye, V., Boon, E., Stewart, J. L., Garcia-Higuera, I., Moreno, S., Rodriguez, R., Jones, K. T., and McLaughlin, E. A. (2014). The APC/C activator FZR1 is essential for meiotic prophase I in mice. Development 141, 1354–1365.
The APC/C activator FZR1 is essential for meiotic prophase I in mice.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXntVehs70%3D&md5=c72e8e719380fdad057a548bad6789f8CAS | 24553289PubMed |

Hopkins, J., Hwang, G., Jacob, J., Sapp, N., Bedigian, R., Oka, K., Overbeek, P., Murray, S., and Jordan, P. W. (2014). Meiosis-specific cohesin component, Stag3 is essential for maintaining centromere chromatid cohesion, and required for DNA repair and synapsis between homologous chromosomes. PLoS Genet. 10, e1004413.
Meiosis-specific cohesin component, Stag3 is essential for maintaining centromere chromatid cohesion, and required for DNA repair and synapsis between homologous chromosomes.Crossref | GoogleScholarGoogle Scholar | 24992337PubMed |

Inoue, S., Tomasini, R., Rufini, A., Elia, A. J., Agostini, M., Amelio, I., Cescon, D., Dinsdale, D., Zhou, L., Harris, I. S., Lac, S., Silvester, J., Li, W. Y., Sasaki, M., Haight, J., Brustle, A., Wakeham, A., McKerlie, C., Jurisicova, A., Melino, G., and Mak, T. W. (2014). TAp73 is required for spermatogenesis and the maintenance of male fertility. Proc. Natl Acad. Sci. USA 111, 1843–1848.
TAp73 is required for spermatogenesis and the maintenance of male fertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXitFOmurk%3D&md5=20dfa64accc05a6610f2779cb3ecb91bCAS | 24449892PubMed |

Kang, E. R., Iqbal, K., Tran, D. A., Rivas, G. E., Singh, P., Pfeifer, G. P., and Szabo, P. E. (2011). Effects of endocrine disruptors on imprinted gene expression in the mouse embryo. Epigenetics 6, 937–950.
Effects of endocrine disruptors on imprinted gene expression in the mouse embryo.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xht1OrsLw%3D&md5=e4623b91812d40e6c78f5d427312e64aCAS | 21636974PubMed |

Kiani, J., Grandjean, V., Liebers, R., Tuorto, F., Ghanbarian, H., Lyko, F., Cuzin, F., and Rassoulzadegan, M. (2013). RNA-mediated epigenetic heredity requires the cytosine methyltransferase Dnmt2. PLoS Genet. 9, e1003498.
RNA-mediated epigenetic heredity requires the cytosine methyltransferase Dnmt2.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXpsFeksr4%3D&md5=a0687fb15849018f60c3660b06b3663dCAS | 23717211PubMed |

Kiger, N., Chaouat, G., Kolb, J. P., Wegmann, T. G., and Guenet, J. L. (1985). Immunogenetic studies of spontaneous abortion in mice. Preimmunization of females with allogeneic cells. J. Immunol. 134, 2966–2970.
| 1:STN:280:DyaL2M7lvFSjsg%3D%3D&md5=2cca5acc9e51af585879f7e209853ee6CAS | 3980987PubMed |

Kjeldsen, L. S., Ghisari, M., and Bonefeld-Jorgensen, E. C. (2013). Currently used pesticides and their mixtures affect the function of sex hormone receptors and aromatase enzyme activity. Toxicol. Appl. Pharmacol. 272, 453–464.
Currently used pesticides and their mixtures affect the function of sex hormone receptors and aromatase enzyme activity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhsVylu7fE&md5=746c1f3839411e09c96ceb73e1e966f6CAS | 23871939PubMed |

Kobayashi, H., and Iwamatsu, T. (2005). Sex reversal in the medaka Oryzias latipes by brief exposure of early embryos to estradiol-17beta. Zoolog. Sci. 22, 1163–1167.
Sex reversal in the medaka Oryzias latipes by brief exposure of early embryos to estradiol-17beta.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XnvFSjsg%3D%3D&md5=8ef5c14b580c82a1ddc75babd0873ddeCAS | 16286729PubMed |

Kooijman, S. A., and Lika, K. (2014). Resource allocation to reproduction in animals. Biol. Rev. Camb. Philos. Soc , .
Resource allocation to reproduction in animals.Crossref | GoogleScholarGoogle Scholar | 24517882PubMed |

Koopman, P., Gubbay, J., Vivian, N., Goodfellow, P., and Lovell-Badge, R. (1991). Male development of chromosomally female mice transgenic for Sry. Nature 351, 117–121.
Male development of chromosomally female mice transgenic for Sry.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXktVagsrk%3D&md5=2f99b4c04e2ca4a06b7659bedd4e990bCAS | 2030730PubMed |

Krawetz, S. A., Kruger, A., Lalancette, C., Tagett, R., Anton, E., Draghici, S., and Diamond, M. P. (2011). A survey of small RNAs in human sperm. Hum. Reprod. 26, 3401–3412.
A survey of small RNAs in human sperm.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhsVymtb7P&md5=896aa2f36638eb5f0d0b3bc1256187f9CAS | 21989093PubMed |

Kusakabe, H., Yanagimachi, R., and Kamiguchi, Y. (2008). Mouse and human spermatozoa can be freeze-dried without damaging their chromosomes. Hum. Reprod. 23, 233–239.
Mouse and human spermatozoa can be freeze-dried without damaging their chromosomes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXptlClsw%3D%3D&md5=faebe6fc332bbd5cfa942db2eacad256CAS | 18056060PubMed |

Leroy, J. L., Opsomer, G., Van Soom, A., Goovaerts, I. G., and Bols, P. E. (2008). Reduced fertility in high-yielding dairy cows: are the oocyte and embryo in danger? Part I. The importance of negative energy balance and altered corpus luteum function to the reduction of oocyte and embryo quality in high-yielding dairy cows. Reprod. Domest. Anim. 43, 612–622.
Reduced fertility in high-yielding dairy cows: are the oocyte and embryo in danger? Part I. The importance of negative energy balance and altered corpus luteum function to the reduction of oocyte and embryo quality in high-yielding dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD1cnksVeqsg%3D%3D&md5=3feb32c9e0ebf23c778c65c7591b8131CAS | 18384499PubMed |

Lhuillier, P., Rode, B., Escalier, D., Lores, P., Dirami, T., Bienvenu, T., Gacon, G., Dulioust, E., and Toure, A. (2009). Absence of annulus in human asthenozoospermia: case report. Hum. Reprod. 24, 1296–1303.
Absence of annulus in human asthenozoospermia: case report.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD1MzltFOruw%3D%3D&md5=b785039a28da6e6fbff51b2a4722750aCAS | 19221096PubMed |

Lin, M. H., Kuo-Kuang Lee, R., Li, S. H., Lu, C. H., Sun, F. J., and Hwu, Y. M. (2008). Sperm chromatin structure assay parameters are not related to fertilization rates, embryo quality, and pregnancy rates in in vitro fertilization and intracytoplasmic sperm injection, but might be related to spontaneous abortion rates. Fertil. Steril. 90, 352–359.
Sperm chromatin structure assay parameters are not related to fertilization rates, embryo quality, and pregnancy rates in in vitro fertilization and intracytoplasmic sperm injection, but might be related to spontaneous abortion rates.Crossref | GoogleScholarGoogle Scholar | 17904130PubMed |

Llano, E., Gomez, H. L., Garcia-Tunon, I., Sanchez-Martin, M., Caburet, S., Barbero, J. L., Schimenti, J. C., Veitia, R. A., and Pendas, A. M. (2014). STAG3 is a strong candidate gene for male infertility. Hum. Mol. Genet. 23, 3421–3431.
STAG3 is a strong candidate gene for male infertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXpvV2rsbw%3D&md5=ef5759fe7739492519de9a522678b082CAS | 24608227PubMed |

Luconi, M., Forti, G., and Baldi, E. (2006). Pathophysiology of sperm motility. Front. Biosci. 11, 1433–1447.
Pathophysiology of sperm motility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XislSnsr8%3D&md5=607e7a33838cb9dea3270e2908df612fCAS | 16368527PubMed |

Matzuk, M. M., and Lamb, D. J. (2002). Genetic dissection of mammalian fertility pathways. Nat. Cell Biol. 4, S33–S40.
Genetic dissection of mammalian fertility pathways.Crossref | GoogleScholarGoogle Scholar |

Matzuk, M. M., and Lamb, D. J. (2008). The biology of infertility: research advances and clinical challenges. Nat. Med. 14, 1197–1213.
The biology of infertility: research advances and clinical challenges.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtlCjtL%2FM&md5=717de840e336842f45d71121dac96aabCAS | 18989307PubMed |

McMillen, I. C., and Robinson, J. S. (2005). Developmental origins of the metabolic syndrome: prediction, plasticity, and programming. Physiol. Rev. 85, 571–633.
Developmental origins of the metabolic syndrome: prediction, plasticity, and programming.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXjt12lsLw%3D&md5=e60f1e5087cede1852b656d288b1cc2dCAS | 15788706PubMed |

Merzenich, H., Zeeb, H., and Blettner, M. (2010). Decreasing sperm quality: a global problem? BMC Public Health 10, 24.
Decreasing sperm quality: a global problem?Crossref | GoogleScholarGoogle Scholar | 20085639PubMed |

Modi, S., Hughes, M., Garrow, A., and White, A. (2012). The value of in silico chemistry in the safety assessment of chemicals in the consumer goods and pharmaceutical industries. Drug Discov. Today 17, 135–142.
The value of in silico chemistry in the safety assessment of chemicals in the consumer goods and pharmaceutical industries.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XitVSlt7Y%3D&md5=82f0f561fd9f03495123966fa71ac50fCAS | 22063083PubMed |

Moll, A. C., Imhof, S. M., Cruysberg, J. R., Schouten-van Meeteren, A. Y., Boers, M., and van Leeuwen, F. E. (2003). Incidence of retinoblastoma in children born after in-vitro fertilisation. Lancet 361, 309–310.
Incidence of retinoblastoma in children born after in-vitro fertilisation.Crossref | GoogleScholarGoogle Scholar | 12559867PubMed |

Moniot, B., Declosmenil, F., Barrionuevo, F., Scherer, G., Aritake, K., Malki, S., Marzi, L., Cohen-Solal, A., Georg, I., Klattig, J., Englert, C., Kim, Y., Capel, B., Eguchi, N., Urade, Y., Boizet-Bonhoure, B., and Poulat, F. (2009). The PGD2 pathway, independently of FGF9, amplifies SOX9 activity in Sertoli cells during male sexual differentiation. Development 136, 1813–1821.
The PGD2 pathway, independently of FGF9, amplifies SOX9 activity in Sertoli cells during male sexual differentiation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXot1aktLY%3D&md5=e0812235c4a4ea383cc9c50c2cdb28d0CAS | 19429785PubMed |

Moore, D. J., Onoufriadis, A., Shoemark, A., Simpson, M. A., zur Lage, P. I., de Castro, S. C., Bartoloni, L., Gallone, G., Petridi, S., Woollard, W. J., Antony, D., Schmidts, M., Didonna, T., Makrythanasis, P., Bevillard, J., Mongan, N. P., Djakow, J., Pals, G., Lucas, J. S., Marthin, J. K., Nielsen, K. G., Santoni, F., Guipponi, M., Hogg, C., Antonarakis, S. E., Emes, R. D., Chung, E. M., Greene, N. D., Blouin, J. L., Jarman, A. P., and Mitchison, H. M. (2013). Mutations in ZMYND10, a gene essential for proper axonemal assembly of inner and outer dynein arms in humans and flies, cause primary ciliary dyskinesia. Am. J. Hum. Genet. 93, 346–356.
Mutations in ZMYND10, a gene essential for proper axonemal assembly of inner and outer dynein arms in humans and flies, cause primary ciliary dyskinesia.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtFOgu73I&md5=18eaeb94c6f9fb613666e79cf9158f3eCAS | 23891471PubMed |

Mussa, A., Russo, S., De Crescenzo, A., Chiesa, N., Molinatto, C., Selicorni, A., Richiardi, L., Larizza, L., Silengo, M. C., Riccio, A., and Ferrero, G. B. (2013). Prevalence of Beckwith–Wiedemann syndrome in north west of Italy. Am. J. Med. Genet. A. 161, 2481–2486.
| 1:CAS:528:DC%2BC3sXhsFGgt7vK&md5=b52029173d31a9c0b40a8d11a68ac08cCAS |

Nair, R. R., Khanna, A., and Singh, K. (2013). Role of inflammatory proteins S100A8 and S100A9 in pathophysiology of recurrent early pregnancy loss. Placenta 34, 824–827.
Role of inflammatory proteins S100A8 and S100A9 in pathophysiology of recurrent early pregnancy loss.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtFSlsr3P&md5=8af3ba1725169bec2632a0e5eb74bb4eCAS | 23850136PubMed |

O’Brien, S. J., Wildt, D. E., Bush, M., Caro, T. M., FitzGibbon, C., Aggundey, I., and Leakey, R. E. (1987). East African cheetahs: evidence for two population bottlenecks? Proc. Natl Acad. Sci. USA 84, 508–511.
East African cheetahs: evidence for two population bottlenecks?Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL2s%2Fpt1CgtQ%3D%3D&md5=ad74c425435bb2bc059411267d3ae445CAS | 3467370PubMed |

Ogonuki, N., Mochida, K., Miki, H., Inoue, K., Fray, M., Iwaki, T., Moriwaki, K., Obata, Y., Morozumi, K., Yanagimachi, R., and Ogura, A. (2006). Spermatozoa and spermatids retrieved from frozen reproductive organs or frozen whole bodies of male mice can produce normal offspring. Proc. Natl Acad. Sci. USA 103, 13 098–13 103.
Spermatozoa and spermatids retrieved from frozen reproductive organs or frozen whole bodies of male mice can produce normal offspring.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xptlemu7g%3D&md5=2b7e438f9ce2cb69d3e6a711b4b330adCAS |

Okun, N., and Sierra, S. (2014). Pregnancy outcomes after assisted human reproduction. J. Obstet. Gynaecol. Can. 36, 64–83.
| 24444289PubMed |

Okutsu, T., Suzuki, K., Takeuchi, Y., Takeuchi, T., and Yoshizaki, G. (2006). Testicular germ cells can colonize sexually undifferentiated embryonic gonad and produce functional eggs in fish. Proc. Natl Acad. Sci. USA 103, 2725–2729.
Testicular germ cells can colonize sexually undifferentiated embryonic gonad and produce functional eggs in fish.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XksF2rtrc%3D&md5=72c61366f99b2ec5ebf39b7bc3f2fe27CAS | 16473947PubMed |

Onoufriadis, A., Shoemark, A., Munye, M. M., James, C. T., Schmidts, M., Patel, M., Rosser, E. M., Bacchelli, C., Beales, P. L., Scambler, P. J., Hart, S. L., Danke-Roelse, J. E., Sloper, J. J., Hull, S., Hogg, C., Emes, R. D., Pals, G., Moore, A. T., Chung, E. M., and Mitchison, H. M. (2014). Combined exome and whole-genome sequencing identifies mutations in ARMC4 as a cause of primary ciliary dyskinesia with defects in the outer dynein arm. J. Med. Genet. 51, 61–67.
Combined exome and whole-genome sequencing identifies mutations in ARMC4 as a cause of primary ciliary dyskinesia with defects in the outer dynein arm.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXktVyku78%3D&md5=1448ab501e0db6956307948ab031a148CAS | 24203976PubMed |

Pailhoux, E., Vigier, B., Chaffaux, S., Servel, N., Taourit, S., Furet, J. P., Fellous, M., Grosclaude, F., Cribiu, E. P., Cotinot, C., and Vaiman, D. (2001). A 11.7-kb deletion triggers intersexuality and polledness in goats. Nat. Genet. 29, 453–458.
A 11.7-kb deletion triggers intersexuality and polledness in goats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXovFCrsro%3D&md5=3d9e840a519087f8606fb5ae6937db27CAS | 11726932PubMed |

Parma, P., Radi, O., Vidal, V., Chaboissier, M. C., Dellambra, E., Valentini, S., Guerra, L., Schedl, A., and Camerino, G. (2006). R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nat. Genet. 38, 1304–1309.
R-spondin1 is essential in sex determination, skin differentiation and malignancy.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XhtFeisbjF&md5=75425dff8527236f116c3e0f2123d1a4CAS | 17041600PubMed |

Perrin, J., Metzler-Guillemain, C., Karsenty, G., Grillo, J. M., Mitchell, M. J., and Guichaoua, M. R. (2006). Meiotic arrest at the midpachytene stage in a patient with complete azoospermia factor b deletion of the Y chromosome. Fertil. Steril. 85, 494e5–494.e8.
Meiotic arrest at the midpachytene stage in a patient with complete azoospermia factor b deletion of the Y chromosome.Crossref | GoogleScholarGoogle Scholar |

Perrin, A., Coat, C., Nguyen, M. H., Talagas, M., Morel, F., Amice, J., and De Braekeleer, M. (2013). Molecular cytogenetic and genetic aspects of globozoospermia: a review. Andrologia 45, 1–9.
Molecular cytogenetic and genetic aspects of globozoospermia: a review.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtlGhsL0%3D&md5=23537b06b0368843269363a186dc4e65CAS | 22571172PubMed |

Pierre, V., Martinez, G., Coutton, C., Delaroche, J., Yassine, S., Novella, C., Pernet-Gallay, K., Hennebicq, S., Ray, P. F., and Arnoult, C. (2012). Absence of Dpy19l2, a new inner nuclear membrane protein, causes globozoospermia in mice by preventing the anchoring of the acrosome to the nucleus. Development 139, 2955–2965.
Absence of Dpy19l2, a new inner nuclear membrane protein, causes globozoospermia in mice by preventing the anchoring of the acrosome to the nucleus.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhsVWlu7rN&md5=90b710264939c25cca29eba92579cb8aCAS | 22764053PubMed |

Piersma, A. H. (2013). Innovations in testing strategies in reproductive toxicology. Methods Mol. Biol. 947, 327–341.
Innovations in testing strategies in reproductive toxicology.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXktlaqt74%3D&md5=0a5b76c09604789337a78aff4f1b13e5CAS | 23138915PubMed |

Pinborg, A., Wennerholm, U. B., Romundstad, L. B., Loft, A., Aittomaki, K., Soderstrom-Anttila, V., Nygren, K. G., Hazekamp, J., and Bergh, C. (2013). Why do singletons conceived after assisted reproduction technology have adverse perinatal outcome? Systematic review and meta-analysis. Hum. Reprod. Update 19, 87–104.
Why do singletons conceived after assisted reproduction technology have adverse perinatal outcome? Systematic review and meta-analysis.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BC3s7js1SktA%3D%3D&md5=1651ea91b4d54a2a4b75a0066bb8786cCAS | 23154145PubMed |

Ponter, A. A., Guyader-Joly, C., Nuttinck, F., Grimard, B., and Humblot, P. (2012). Oocyte and embryo production and quality after OPU-IVF in dairy heifers given diets varying in their n-6/n-3 fatty acid ratio. Theriogenology 78, 632–645.
Oocyte and embryo production and quality after OPU-IVF in dairy heifers given diets varying in their n-6/n-3 fatty acid ratio.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XmtFertrk%3D&md5=c7356c2056a0a3b2fdc23e849ee6ebcdCAS | 22537996PubMed |

Prefumo, F., Fratelli, N., Soares, S. C., and Thilaganathan, B. (2007). Uterine artery Doppler velocimetry at 11–14 weeks in singleton pregnancies conceived by assisted reproductive technology. Ultrasound Obstet. Gynecol. 29, 141–145.
Uterine artery Doppler velocimetry at 11–14 weeks in singleton pregnancies conceived by assisted reproductive technology.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD2s%2FlsFOgtA%3D%3D&md5=6a2bd2d8926e721bf82c773be7f83ab7CAS | 17072900PubMed |

Prüfer, K., Racimo, F., Patterson, N., Jay, F., Sankararaman, S., Sawyer, S., Heinze, A., Renaud, G., Sudmant, P. H., de Filippo, C., Li, H., Mallick, S., Dannemann, M., Fu, Q., Kircher, M., Kuhlwilm, M., Lachmann, M., Meyer, M., Ongyerth, M., Siebauer, M., Theunert, C., Tandon, A., Moorjani, P., Pickrell, J., Mullikin, J. C., Vohr, S. H., Green, R. E., Hellmann, I., Johnson, P. L., Blanche, H., Cann, H., Kitzman, J. O., Shendure, J., Eichler, E. E., Lein, E. S., Bakken, T. E., Golovanova, L. V., Doronichev, V. B., Shunkov, M. V., Derevianko, A. P., Viola, B., Slatkin, M., Reich, D., Kelso, J., and Paabo, S. (2014). The complete genome sequence of a Neanderthal from the Altai Mountains. Nature 505, 43–49.
The complete genome sequence of a Neanderthal from the Altai Mountains.Crossref | GoogleScholarGoogle Scholar | 24352235PubMed |

Rahmioglu, N., Nyholt, D. R., Morris, A. P., Missmer, S. A., Montgomery, G. W., and Zondervan, K. T. (2014). Genetic variants underlying risk of endometriosis: insights from meta-analysis of eight genome-wide association and replication datasets. Hum. Reprod. Update 20, 702–716.
Genetic variants underlying risk of endometriosis: insights from meta-analysis of eight genome-wide association and replication datasets.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtlyku7nI&md5=b1c665272493336c1383b56dbfb9bfc4CAS | 24676469PubMed |

Rassoulzadegan, M., Grandjean, V., Gounon, P., Vincent, S., Gillot, I., and Cuzin, F. (2006). RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse. Nature 441, 469–474.
RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XkvVykurk%3D&md5=0805d8cc9f84252c26d271133cd8d531CAS | 16724059PubMed |

Renfree, M. B., Hore, T. A., Shaw, G., Graves, J. A., and Pask, A. J. (2009). Evolution of genomic imprinting: insights from marsupials and monotremes. Annu. Rev. Genomics Hum. Genet. 10, 241–262.
Evolution of genomic imprinting: insights from marsupials and monotremes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXht12qsrvL&md5=a0a72e3c67095ce3c1ec233e527490e1CAS | 19630559PubMed |

Renfree, M. B., Suzuki, S., and Kaneko-Ishino, T. (2013). The origin and evolution of genomic imprinting and viviparity in mammals. Philos. Trans. R. Soc. Lond. B Biol. Sci. 368, 20120151.
The origin and evolution of genomic imprinting and viviparity in mammals.Crossref | GoogleScholarGoogle Scholar | 23166401PubMed |

Rigot, J. M., Lafitte, J. J., Dumur, V., Gervais, R., Manouvrier, S., Biserte, J., Mazeman, E., and Roussel, P. (1991). Cystic fibrosis and congenital absence of the vas deferens. N. Engl. J. Med. 325, 64–65.
Cystic fibrosis and congenital absence of the vas deferens.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK3M3mtVOqug%3D%3D&md5=58667a31fbdd2f4756d2430a3090e3f9CAS | 2046716PubMed |

Robinson, L., Gallos, I. D., Conner, S. J., Rajkhowa, M., Miller, D., Lewis, S., Kirkman-Brown, J., and Coomarasamy, A. (2012). The effect of sperm DNA fragmentation on miscarriage rates: a systematic review and meta-analysis. Hum. Reprod. 27, 2908–2917.
The effect of sperm DNA fragmentation on miscarriage rates: a systematic review and meta-analysis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhtlymsbbI&md5=1a960fe192ece1a35eb99b7769f544c5CAS | 22791753PubMed |

Rode, B., Dirami, T., Bakouh, N., Rizk-Rabin, M., Norez, C., Lhuillier, P., Lores, P., Jollivet, M., Melin, P., Zvetkova, I., Bienvenu, T., Becq, F., Planelles, G., Edelman, A., Gacon, G., and Toure, A. (2012). The testis anion transporter TAT1 (SLC26A8) physically and functionally interacts with the cystic fibrosis transmembrane conductance regulator channel: a potential role during sperm capacitation. Hum. Mol. Genet. 21, 1287–1298.
The testis anion transporter TAT1 (SLC26A8) physically and functionally interacts with the cystic fibrosis transmembrane conductance regulator channel: a potential role during sperm capacitation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XivVeju7g%3D&md5=0c1f7003267262c2a10452c247033239CAS | 22121115PubMed |

Salum, L. B., Polikarpov, I., and Andricopulo, A. D. (2007). Quantitative structure–activity relationships for a series of selective estrogen receptor-beta modulators. SAR QSAR Environ. Res. 18, 711–727.
Quantitative structure–activity relationships for a series of selective estrogen receptor-beta modulators.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtlGmtb%2FL&md5=86c952f94001b8e9dd3a9569aeca6d8bCAS | 18038369PubMed |

Santulli, P., Marcellin, L., Noel, J. C., Borghese, B., Fayt, I., Vaiman, D., Chapron, C., and Mehats, C. (2012). Sphingosine pathway deregulation in endometriotic tissues. Fertil. Steril. 97, 904–911.
Sphingosine pathway deregulation in endometriotic tissues.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XkvFyqtrg%3D&md5=7520d3be9da3b2eb83702616ba9a80deCAS | 22277765PubMed |

Santulli, P., Borghese, B., Noël, J.-C., Fayt, I., Anaf, V., de Ziegler, D., Batteux, F., Vaiman, D., and Chapron, C. (2014). Hormonal therapy deregulates prostaglandin-endoperoxidase synthase 2 (PTGS2) expression in endometriotic tissues. J. Clin. Endocrinol. Metab. 99, 881–890.
| 1:CAS:528:DC%2BC2cXksFWkt7s%3D&md5=2103b2ed843155011513a71c8af17103CAS | 24423291PubMed |

Sartori, R., Bastos, M. R., and Wiltbank, M. C. (2010). Factors affecting fertilisation and early embryo quality in single- and superovulated dairy cattle. Reprod. Fertil. Dev. 22, 151–158.
Factors affecting fertilisation and early embryo quality in single- and superovulated dairy cattle.Crossref | GoogleScholarGoogle Scholar | 20003858PubMed |

Shioda, T., Rosenthal, N. F., Coser, K. R., Suto, M., Phatak, M., Medvedovic, M., Carey, V. J., and Isselbacher, K. J. (2013). Expressomal approach for comprehensive analysis and visualization of ligand sensitivities of xenoestrogen responsive genes. Proc. Natl Acad. Sci. USA 110, 16 508–16 513.
Expressomal approach for comprehensive analysis and visualization of ligand sensitivities of xenoestrogen responsive genes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhs1Kju7%2FM&md5=f9d54c860418bc358ed4a621b0f5190dCAS |

Smith, J. F., Syritsyna, O., Fellous, M., Serres, C., Mannowetz, N., Kirichok, Y., and Lishko, P. V. (2013). Disruption of the principal, progesterone-activated sperm Ca2+ channel in a CatSper2-deficient infertile patient. Proc. Natl Acad. Sci. USA 110, 6823–6828.
Disruption of the principal, progesterone-activated sperm Ca2+ channel in a CatSper2-deficient infertile patient.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXot1Ghsbk%3D&md5=7a6ca4421e706337d0054f89c72c6750CAS | 23530196PubMed |

Smits, G., Mungall, A. J., Griffiths-Jones, S., Smith, P., Beury, D., Matthews, L., Rogers, J., Pask, A. J., Shaw, G., VandeBerg, J. L., McCarrey, J. R., Renfree, M. B., Reik, W., and Dunham, I. (2008). Conservation of the H19 noncoding RNA and H19–IGF2 imprinting mechanism in therians. Nat. Genet. 40, 971–976.
Conservation of the H19 noncoding RNA and H19–IGF2 imprinting mechanism in therians.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXptVKku7s%3D&md5=0fc9d302b89512fec60d77e239350ad5CAS | 18587395PubMed |

Stouder, C., and Paoloni-Giacobino, A. (2011). Specific transgenerational imprinting effects of the endocrine disruptor methoxychlor on male gametes. Reproduction 141, 207–216.
Specific transgenerational imprinting effects of the endocrine disruptor methoxychlor on male gametes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXisVehsLg%3D&md5=1e509f341778124a702bdeb4bb0a2d3fCAS | 21062904PubMed |

Streichemberger, E., Perrin, J., Saias-Magnan, J., Karsenty, G., Malzac, P., Grillo, J. M., Mitchell, M. J., and Metzler-Guillemain, C. (2012). Case report of apoptosis in testis of four AZFc-deleted patients: increased DNA fragmentation during meiosis, but decreased apoptotic markers in post-meiotic germ cells. Hum. Reprod. 27, 1939–1945.
Case report of apoptosis in testis of four AZFc-deleted patients: increased DNA fragmentation during meiosis, but decreased apoptotic markers in post-meiotic germ cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XptVGis7c%3D&md5=928dfb91be4b2acf1a2efd13986f1b7dCAS | 22508792PubMed |

Sutcliffe, A. G., Peters, C. J., Bowdin, S., Temple, K., Reardon, W., Wilson, L., Clayton-Smith, J., Brueton, L. A., Bannister, W., and Maher, E. R. (2006). Assisted reproductive therapies and imprinting disorders: a preliminary British survey. Hum. Reprod. 21, 1009–1011.
Assisted reproductive therapies and imprinting disorders: a preliminary British survey.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD283hvVKntA%3D%3D&md5=3c6c421c6a8241253cab9b7cc90336b8CAS | 16361294PubMed |

Takasaki, N., Tachibana, K., Ogasawara, S., Matsuzaki, H., Hagiuda, J., Ishikawa, H., Mochida, K., Inoue, K., Ogonuki, N., Ogura, A., Noce, T., Ito, C., Toshimori, K., and Narimatsu, H. (2014). A heterozygous mutation of GALNTL5 affects male infertility with impairment of sperm motility. Proc. Natl Acad. Sci. USA 111, 1120–1125.
A heterozygous mutation of GALNTL5 affects male infertility with impairment of sperm motility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtlertLk%3D&md5=6ce51f3724e4fbf129ac20fa0e5bfe5eCAS | 24398516PubMed |

Tararbit, K., Lelong, N., Thieulin, A. C., Houyel, L., Bonnet, D., Goffinet, F., and Khoshnood, B. (2013). The risk for four specific congenital heart defects associated with assisted reproductive techniques: a population-based evaluation. Hum. Reprod. 28, 367–374.
The risk for four specific congenital heart defects associated with assisted reproductive techniques: a population-based evaluation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhsVOnurw%3D&md5=e3a22f89e63620b123164ece753c7896CAS | 23178272PubMed |

Taylor, S. L., Yoon, S. Y., Morshedi, M. S., Lacey, D. R., Jellerette, T., Fissore, R. A., and Oehninger, S. (2010). Complete globozoospermia associated with PLCzeta deficiency treated with calcium ionophore and ICSI results in pregnancy. Reprod. Biomed. Online 20, 559–564.
Complete globozoospermia associated with PLCzeta deficiency treated with calcium ionophore and ICSI results in pregnancy.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BC3cfgsVeiug%3D%3D&md5=c17d4677dedcdc500dd4891f5b3abd84CAS | 20133201PubMed |

Tong, W., Xie, Q., Hong, H., Shi, L., Fang, H., and Perkins, R. (2004). Assessment of prediction confidence and domain extrapolation of two structure–activity relationship models for predicting estrogen receptor binding activity. Environ. Health Perspect. 112, 1249–1254.
Assessment of prediction confidence and domain extrapolation of two structure–activity relationship models for predicting estrogen receptor binding activity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXotVKjt7c%3D&md5=16f41125a12ab47745002b68cf1068f3CAS | 15345371PubMed |

Toth, B., Wurfel, W., Germeyer, A., Hirv, K., Makrigiannakis, A., and Strowitzki, T. (2011). Disorders of implantation: are there diagnostic and therapeutic options? J. Reprod. Immunol. 90, 117–123.
Disorders of implantation: are there diagnostic and therapeutic options?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhsVGiurzN&md5=f7493dbf309a92fad2859bb01e5910acCAS | 21641658PubMed |

Tsoumpou, I., Mohamed, A. M., Tower, C., Roberts, S. A., and Nardo, L. G. (2011). Failed IVF cycles and the risk of subsequent preeclampsia or fetal growth restriction: a case-control exploratory study. Fertil. Steril. 95, 973–978.
Failed IVF cycles and the risk of subsequent preeclampsia or fetal growth restriction: a case-control exploratory study.Crossref | GoogleScholarGoogle Scholar | 20797707PubMed |

Vaiman, D. (2014). Genetic regulation of recurrent spontaneous abortion in humans. Biom. J. 37, .

Vaiman, D., and Pailhoux, E. (2000). Mammalian sex reversal and intersexuality: deciphering the sex-determination cascade. Trends Genet. 16, 488–494.
Mammalian sex reversal and intersexuality: deciphering the sex-determination cascade.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXosV2js7w%3D&md5=e95685eaa028a6da1a9bc7a2b10ebe10CAS | 11074290PubMed |

Vaiman, D., Gascoin-Lachambre, G., Boubred, F., Mondon, F., Feuerstein, J. M., Ligi, I., Grandvuillemin, I., Barbaux, S., Ghigo, E., Achard, V., Simeoni, U., and Buffat, C. (2011). The intensity of IUGR-induced transcriptome deregulations is inversely correlated with the onset of organ function in a rat model. PLoS ONE 6, e21222.
The intensity of IUGR-induced transcriptome deregulations is inversely correlated with the onset of organ function in a rat model.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXotF2gu78%3D&md5=7c2b68faa1b3d198254bd5e64a482aa9CAS | 21731679PubMed |

Vandenberg, L. N., Colborn, T., Hayes, T. B., Heindel, J. J., Jacobs, D. R., Lee, D. H., Shioda, T., Soto, A. M., vom Saal, F. S., Welshons, W. V., Zoeller, R. T., and Myers, J. P. (2012). Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocr. Rev. 33, 378–455.
Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhtFSrtr7L&md5=deab2425c3239cecc0089bc19b6be058CAS | 22419778PubMed |

Varrault, A., Gueydan, C., Delalbre, A., Bellmann, A., Houssami, S., Aknin, C., Severac, D., Chotard, L., Kahli, M., Le Digarcher, A., Pavlidis, P., and Journot, L. (2006). Zac1 regulates an imprinted gene network critically involved in the control of embryonic growth. Dev. Cell 11, 711–722.
Zac1 regulates an imprinted gene network critically involved in the control of embryonic growth.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xht1Smsr%2FF&md5=e2f0b71bb4ebf0e3e7f2b0c314940efcCAS | 17084362PubMed |

Vatin, M., Bouvier, S., Bellazi, L., Montagutelli, X., Laissue, P., Ziyyat, A., Serres, C., De Mazancourt, P., Dieudonne, M. N., Mornet, E., Vaiman, D., and Gris, J. C. (2014). Polymorphisms of human placental alkaline phosphatase are associated with in vitro fertilization success and recurrent pregnancy loss. Am. J. Pathol. 184, 362–368.
Polymorphisms of human placental alkaline phosphatase are associated with in vitro fertilization success and recurrent pregnancy loss.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtlKhtbk%3D&md5=1d8370d43e3a37e404bd81e5aa080815CAS | 24296104PubMed |

Wang, J. X., Knottnerus, A. M., Schuit, G., Norman, R. J., Chan, A., and Dekker, G. A. (2002). Surgically obtained sperm, and risk of gestational hypertension and pre-eclampsia. Lancet 359, 673–674.
Surgically obtained sperm, and risk of gestational hypertension and pre-eclampsia.Crossref | GoogleScholarGoogle Scholar | 11879865PubMed |

Wildt, D. E., Phillips, L. G., Simmons, L. G., Chakraborty, P. K., Brown, J. L., Howard, J. G., Teare, A., and Bush, M. (1988). A comparative analysis of ejaculate and hormonal characteristics of the captive male cheetah, tiger, leopard, and puma. Biol. Reprod. 38, 245–255.
A comparative analysis of ejaculate and hormonal characteristics of the captive male cheetah, tiger, leopard, and puma.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL1cXhs1Wlu78%3D&md5=cb10481e9596c24891cb0d9bda185771CAS | 2833943PubMed |

Winters, T., McNicoll, F., and Jessberger, R. (2014). Meiotic cohesin STAG3 is required for chromosome axis formation and sister chromatid cohesion. EMBO J. 33, 1256–1270.
| 1:CAS:528:DC%2BC2cXhtFWgsr%2FF&md5=95a0356ced3dd245e0668966eda2a189CAS | 24797474PubMed |

Yan, W. (2009). Male infertility caused by spermiogenic defects: lessons from gene knockouts. Mol. Cell. Endocrinol. 306, 24–32.
Male infertility caused by spermiogenic defects: lessons from gene knockouts.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXms1ait7s%3D&md5=3483d3807bdf403fd65f2d2adec356f5CAS | 19481682PubMed |

Yoon, S. Y., Jellerette, T., Salicioni, A. M., Lee, H. C., Yoo, M. S., Coward, K., Parrington, J., Grow, D., Cibelli, J. B., Visconti, P. E., Mager, J., and Fissore, R. A. (2008). Human sperm devoid of PLC, zeta 1 fail to induce Ca(2+) release and are unable to initiate the first step of embryo development. J. Clin. Invest. 118, 3671–3681.
Human sperm devoid of PLC, zeta 1 fail to induce Ca(2+) release and are unable to initiate the first step of embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtlGltLvF&md5=9d98c235211754a9c423d8e68300feb6CAS | 18924610PubMed |

Yu, L. L., Chang, K., Lu, L. S., Zhao, D., Han, J., Zheng, Y. R., Yan, Y. H., Yi, P., Guo, J. X., Zhou, Y. G., Chen, M., and Li, L. (2013). Lentivirus-mediated RNA interference targeting the H19 gene inhibits cell proliferation and apoptosis in human choriocarcinoma cell line JAR. BMC Cell Biol. 14, 26.
Lentivirus-mediated RNA interference targeting the H19 gene inhibits cell proliferation and apoptosis in human choriocarcinoma cell line JAR.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtVSrsrrJ&md5=52b1118af67e3b4a0b5e46680c7840e7CAS | 23711233PubMed |

Zariwala, M. A., Gee, H. Y., Kurkowiak, M., Al-Mutairi, D. A., Leigh, M. W., Hurd, T. W., Hjeij, R., Dell, S. D., Chaki, M., Dougherty, G. W., Adan, M., Spear, P. C., Esteve-Rudd, J., Loges, N. T., Rosenfeld, M., Diaz, K. A., Olbrich, H., Wolf, W. E., Sheridan, E., Batten, T. F., Halbritter, J., Porath, J. D., Kohl, S., Lovric, S., Hwang, D. Y., Pittman, J. E., Burns, K. A., Ferkol, T. W., Sagel, S. D., Olivier, K. N., Morgan, L. C., Werner, C., Raidt, J., Pennekamp, P., Sun, Z., Zhou, W., Airik, R., Natarajan, S., Allen, S. J., Amirav, I., Wieczorek, D., Landwehr, K., Nielsen, K., Schwerk, N., Sertic, J., Kohler, G., Washburn, J., Levy, S., Fan, S., Koerner-Rettberg, C., Amselem, S., Williams, D. S., Mitchell, B. J., Drummond, I. A., Otto, E. A., Omran, H., Knowles, M. R., and Hildebrandt, F. (2013). ZMYND10 is mutated in primary ciliary dyskinesia and interacts with LRRC6. Am. J. Hum. Genet. 93, 336–345.
ZMYND10 is mutated in primary ciliary dyskinesia and interacts with LRRC6.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtFOnsb%2FK&md5=9bce7734df9657af14ec1426c9dbdd10CAS | 23891469PubMed |

Zhang, L., Sedykh, A., Tripathi, A., Zhu, H., Afantitis, A., Mouchlis, V. D., Melagraki, G., Rusyn, I., and Tropsha, A. (2013). Identification of putative estrogen receptor-mediated endocrine disrupting chemicals using QSAR- and structure-based virtual screening approaches. Toxicol. Appl. Pharmacol. 272, 67–76.
Identification of putative estrogen receptor-mediated endocrine disrupting chemicals using QSAR- and structure-based virtual screening approaches.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhsVClsbfE&md5=4a643038d8ae5d13b4dd7ad1741e41f0CAS | 23707773PubMed |

Zhao, F. Q., Dixon, W. T., and Kennelly, J. J. (1996). Localization and gene expression of glucose transporters in bovine mammary gland. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 115, 127–134.
Localization and gene expression of glucose transporters in bovine mammary gland.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2s%2FltFKktQ%3D%3D&md5=81c523fa976f43713d1126ffa774ebdfCAS | 8896338PubMed |