Skip to main content
Top
Published in: Journal of Assisted Reproduction and Genetics 1/2020

01-01-2020 | Infertility | Genetics

A novel FOXL2 mutation in two infertile patients with blepharophimosis–ptosis–epicanthus inversus syndrome

Authors: Jingmei Hu, Hanni Ke, Wei Luo, Yajuan Yang, Hongli Liu, Guangyu Li, Yingying Qin, Jinlong Ma, Shidou Zhao

Published in: Journal of Assisted Reproduction and Genetics | Issue 1/2020

Login to get access

Abstract

Background

Blepharophimosis–ptosis–epicanthus inversus syndrome (BPES) is a rare, autosomal dominant disease. There are two clinical types of BPES: type I patients have eyelid abnormalities accompanied by infertility in affected females, while type II patients only display eyelid malformations. Previous studies have reported that the forkhead box L2 (FOXL2) gene mutations cause BPES.

Purpose

To identify plausible FOXL2 mutation in a Chinese family with BPES and infertility

Methods

Mutational screening of FOXL2 was performed in the affected members and 223 controls. Functional characterization of the novel mutation identified was carried out in vitro by luciferase reporter assay and subcellular localization experiment.

Results

A novel heterozygous mutation c.188 T > A (p.I63N) in FOXL2 was identified in two BPES patients in this family. The mutation abolished the transcriptional repression of FOXL2 on the promoters of CYP19A1 and CCND2 genes, as shown by luciferase reporter assays. However, no dominant-negative effect was observed for the mutation, and it did not impact FOXL2 protein nuclear localization and distribution.

Conclusions

The mutation c.188 T > A (p.I63N) in FOXL2 might be causative for BPES and infertility in this family and further amplified the spectrum of FOXL2 mutations.
Literature
1.
go back to reference Bertini V, Valetto A, Baldinotti F, Azzarà A, Cambi F, Toschi B, et al. Blepharophimosis, ptosis, epicanthus inversus syndrome: new report with a 197-kb deletion upstream of FOXL2 and review of the literature. Molecular Syndromology. 2019;10:147–53.CrossRef Bertini V, Valetto A, Baldinotti F, Azzarà A, Cambi F, Toschi B, et al. Blepharophimosis, ptosis, epicanthus inversus syndrome: new report with a 197-kb deletion upstream of FOXL2 and review of the literature. Molecular Syndromology. 2019;10:147–53.CrossRef
2.
go back to reference Verdin H, De Baere E. Blepharophimosis, ptosis, and epicanthus inversus. University of Washington: GeneReviews; 2015. p. 1–18. Verdin H, De Baere E. Blepharophimosis, ptosis, and epicanthus inversus. University of Washington: GeneReviews; 2015. p. 1–18.
3.
go back to reference Zlotogora J, Sagi M, Cohen T. The blepharophimosis, ptosis, and epicanthus inversus syndrome: delineation of two types. Am J Hum Genet. 1983;35:1020–7.PubMedPubMedCentral Zlotogora J, Sagi M, Cohen T. The blepharophimosis, ptosis, and epicanthus inversus syndrome: delineation of two types. Am J Hum Genet. 1983;35:1020–7.PubMedPubMedCentral
4.
go back to reference Crisponi L, Deiana M, Loi A, Chiappe F, Uda M, Amati P, et al. The putative forkhead transcription factor FOXL2 is mutated in blepharophimosis/ptosis/epicanthus inversus syndrome. Nat Genet. 2001;27:159–66.CrossRef Crisponi L, Deiana M, Loi A, Chiappe F, Uda M, Amati P, et al. The putative forkhead transcription factor FOXL2 is mutated in blepharophimosis/ptosis/epicanthus inversus syndrome. Nat Genet. 2001;27:159–66.CrossRef
5.
go back to reference Bunyan DJ, Thomas NS. Screening of a large cohort of blepharophimosis, ptosis, and epicanthus inversus syndrome patients reveals a very strong paternal inheritance bias and a wide spectrum of novel FOXL2 mutations. EUR J MED GENET. 2019;62:103668.CrossRef Bunyan DJ, Thomas NS. Screening of a large cohort of blepharophimosis, ptosis, and epicanthus inversus syndrome patients reveals a very strong paternal inheritance bias and a wide spectrum of novel FOXL2 mutations. EUR J MED GENET. 2019;62:103668.CrossRef
6.
go back to reference Verdin H, De Baere E. FOXL2 impairment in human disease. Horm Res Paediatr. 2012;77:2–11.CrossRef Verdin H, De Baere E. FOXL2 impairment in human disease. Horm Res Paediatr. 2012;77:2–11.CrossRef
7.
go back to reference Cocquet J, Pailhoux E, Jaubert F, Servel N, Xia X, Pannetier M, et al. Evolution and expression of FOXL2. J Med Genet. 2002;39:916–21.CrossRef Cocquet J, Pailhoux E, Jaubert F, Servel N, Xia X, Pannetier M, et al. Evolution and expression of FOXL2. J Med Genet. 2002;39:916–21.CrossRef
8.
go back to reference Uda M, Ottolenghi C, Crisponi L, Garcia JE, Deiana M, Kimber W, et al. Foxl2 disruption causes mouse ovarian failure by pervasive blockage of follicle development. Hum Mol Genet. 2004;13:1171–81.CrossRef Uda M, Ottolenghi C, Crisponi L, Garcia JE, Deiana M, Kimber W, et al. Foxl2 disruption causes mouse ovarian failure by pervasive blockage of follicle development. Hum Mol Genet. 2004;13:1171–81.CrossRef
9.
go back to reference Schmidt D, Ovitt CE, Anlag K, Fehsenfeld S, Gredsted L, Treier AC, et al. The murine winged-helix transcription factor Foxl2 is required for granulosa cell differentiation and ovary maintenance. DEVELOPMENT. 2004;131:933–42.CrossRef Schmidt D, Ovitt CE, Anlag K, Fehsenfeld S, Gredsted L, Treier AC, et al. The murine winged-helix transcription factor Foxl2 is required for granulosa cell differentiation and ovary maintenance. DEVELOPMENT. 2004;131:933–42.CrossRef
10.
go back to reference Bentsi-Barnes IK, Kuo FT, Barlow GM, Pisarska MD. Human forkhead L2 represses key genes in granulosa cell differentiation including aromatase, P450scc, and cyclin D2. Fertil Steril. 2010;94:353–6.CrossRef Bentsi-Barnes IK, Kuo FT, Barlow GM, Pisarska MD. Human forkhead L2 represses key genes in granulosa cell differentiation including aromatase, P450scc, and cyclin D2. Fertil Steril. 2010;94:353–6.CrossRef
11.
go back to reference Kuo FT, Bentsi-Barnes IK, Barlow GM, Pisarska MD. Mutant Forkhead L2 (FOXL2) proteins associated with premature ovarian failure (POF) dimerize with wild-type FOXL2, leading to altered regulation of genes associated with granulosa cell differentiation. ENDOCRINOLOGY. 2011;152:3917–29.CrossRef Kuo FT, Bentsi-Barnes IK, Barlow GM, Pisarska MD. Mutant Forkhead L2 (FOXL2) proteins associated with premature ovarian failure (POF) dimerize with wild-type FOXL2, leading to altered regulation of genes associated with granulosa cell differentiation. ENDOCRINOLOGY. 2011;152:3917–29.CrossRef
12.
go back to reference Caburet S, Georges A, L'Hote D, Todeschini AL, Benayoun BA, Veitia RA. The transcription factor FOXL2: at the crossroads of ovarian physiology and pathology. Mol Cell Endocrinol. 2012;356:55–64.CrossRef Caburet S, Georges A, L'Hote D, Todeschini AL, Benayoun BA, Veitia RA. The transcription factor FOXL2: at the crossroads of ovarian physiology and pathology. Mol Cell Endocrinol. 2012;356:55–64.CrossRef
13.
go back to reference Beysen D, De Paepe A, De Baere E. FOXL2 mutations and genomic rearrangements in BPES. Hum Mutat. 2009;30:158–69.CrossRef Beysen D, De Paepe A, De Baere E. FOXL2 mutations and genomic rearrangements in BPES. Hum Mutat. 2009;30:158–69.CrossRef
14.
go back to reference Fokstuen S, Antonarakis SE, Blouin JL. FOXL2-mutations in blepharophimosis-ptosis-epicanthus inversus syndrome (BPES); challenges for genetic counseling in female patients. Am J Med Genet A. 2003;117A:143–6.CrossRef Fokstuen S, Antonarakis SE, Blouin JL. FOXL2-mutations in blepharophimosis-ptosis-epicanthus inversus syndrome (BPES); challenges for genetic counseling in female patients. Am J Med Genet A. 2003;117A:143–6.CrossRef
15.
go back to reference De Baere E, Beysen D, Oley C, Lorenz B, Cocquet J, De Sutter P, et al. FOXL2 and BPES: mutational hotspots, phenotypic variability, and revision of the genotype-phenotype correlation. Am J Hum Genet. 2003;72:478–87.CrossRef De Baere E, Beysen D, Oley C, Lorenz B, Cocquet J, De Sutter P, et al. FOXL2 and BPES: mutational hotspots, phenotypic variability, and revision of the genotype-phenotype correlation. Am J Hum Genet. 2003;72:478–87.CrossRef
16.
go back to reference De Baere E, Lemercier B, Christin-Maitre S, Durval D, Messiaen L, Fellous M, et al. FOXL2 mutation screening in a large panel of POF patients and XX males. J Med Genet. 2002;39:e43.CrossRef De Baere E, Lemercier B, Christin-Maitre S, Durval D, Messiaen L, Fellous M, et al. FOXL2 mutation screening in a large panel of POF patients and XX males. J Med Genet. 2002;39:e43.CrossRef
17.
go back to reference Dipietromaria A, Benayoun BA, Todeschini AL, Rivals I, Bazin C, Veitia RA. Towards a functional classification of pathogenic FOXL2 mutations using transactivation reporter systems. Hum Mol Genet. 2009;18:3324–33.CrossRef Dipietromaria A, Benayoun BA, Todeschini AL, Rivals I, Bazin C, Veitia RA. Towards a functional classification of pathogenic FOXL2 mutations using transactivation reporter systems. Hum Mol Genet. 2009;18:3324–33.CrossRef
18.
go back to reference Meduri G, Bachelot A, Duflos C, Bstandig B, Poirot C, Genestie C, et al. FOXL2 mutations lead to different ovarian phenotypes in BPES patients: case report. Hum Reprod. 2010;25:235–43.CrossRef Meduri G, Bachelot A, Duflos C, Bstandig B, Poirot C, Genestie C, et al. FOXL2 mutations lead to different ovarian phenotypes in BPES patients: case report. Hum Reprod. 2010;25:235–43.CrossRef
19.
go back to reference Elzaiat M, Todeschini AL, Caburet S, Veitia RA. The genetic make-up of ovarian development and function: the focus on the transcription factor FOXL2. Clin Genet. 2017;91:173–82.CrossRef Elzaiat M, Todeschini AL, Caburet S, Veitia RA. The genetic make-up of ovarian development and function: the focus on the transcription factor FOXL2. Clin Genet. 2017;91:173–82.CrossRef
20.
go back to reference Pisarska MD, Barlow G, Kuo FT. Minireview: roles of the forkhead transcription factor FOXL2 in granulosa cell biology and pathology. ENDOCRINOLOGY. 2011;152:1199–208.CrossRef Pisarska MD, Barlow G, Kuo FT. Minireview: roles of the forkhead transcription factor FOXL2 in granulosa cell biology and pathology. ENDOCRINOLOGY. 2011;152:1199–208.CrossRef
21.
go back to reference Pisarska MD, Bae J, Klein C, Hsueh AJ. Forkhead l2 is expressed in the ovary and represses the promoter activity of the steroidogenic acute regulatory gene. ENDOCRINOLOGY. 2004;145:3424–33.CrossRef Pisarska MD, Bae J, Klein C, Hsueh AJ. Forkhead l2 is expressed in the ovary and represses the promoter activity of the steroidogenic acute regulatory gene. ENDOCRINOLOGY. 2004;145:3424–33.CrossRef
22.
go back to reference Park M, Suh DS, Lee K, Bae J. Positive cross talk between FOXL2 and antimullerian hormone regulates ovarian reserve. Fertil Steril. 2014;102:847–55.CrossRef Park M, Suh DS, Lee K, Bae J. Positive cross talk between FOXL2 and antimullerian hormone regulates ovarian reserve. Fertil Steril. 2014;102:847–55.CrossRef
23.
go back to reference Harris SE, Chand AL, Winship IM, Gersak K, Aittomaki K, Shelling AN. Identification of novel mutations in FOXL2 associated with premature ovarian failure. Mol Hum Reprod. 2002;8:729–33.CrossRef Harris SE, Chand AL, Winship IM, Gersak K, Aittomaki K, Shelling AN. Identification of novel mutations in FOXL2 associated with premature ovarian failure. Mol Hum Reprod. 2002;8:729–33.CrossRef
24.
go back to reference Laissue P, Lakhal B, Benayoun BA, Dipietromaria A, Braham R, Elghezal H, et al. Functional evidence implicating FOXL2 in non-syndromic premature ovarian failure and in the regulation of the transcription factor OSR2. J Med Genet. 2009;46:455–7.CrossRef Laissue P, Lakhal B, Benayoun BA, Dipietromaria A, Braham R, Elghezal H, et al. Functional evidence implicating FOXL2 in non-syndromic premature ovarian failure and in the regulation of the transcription factor OSR2. J Med Genet. 2009;46:455–7.CrossRef
25.
go back to reference Benayoun BA, Caburet S, Dipietromaria A, Bailly-Bechet M, Batista F, Fellous M, et al. The identification and characterization of a FOXL2 response element provides insights into the pathogenesis of mutant alleles. Hum Mol Genet. 2008;17:3118–27.CrossRef Benayoun BA, Caburet S, Dipietromaria A, Bailly-Bechet M, Batista F, Fellous M, et al. The identification and characterization of a FOXL2 response element provides insights into the pathogenesis of mutant alleles. Hum Mol Genet. 2008;17:3118–27.CrossRef
26.
go back to reference Beysen D, Moumné L, Veitia R, Peters H, Leroy BP, De Paepe A, et al. Missense mutations in the forkhead domain of FOXL2 lead to subcellular mislocalization, protein aggregation and impaired transactivation. Hum Mol Genet. 2008;17:2030–8.CrossRef Beysen D, Moumné L, Veitia R, Peters H, Leroy BP, De Paepe A, et al. Missense mutations in the forkhead domain of FOXL2 lead to subcellular mislocalization, protein aggregation and impaired transactivation. Hum Mol Genet. 2008;17:2030–8.CrossRef
27.
go back to reference Todeschini AL, Dipietromaria A, L'Hote D, Boucham FZ, Georges AB, Pandaranayaka PJ, et al. Mutational probing of the forkhead domain of the transcription factor FOXL2 provides insights into the pathogenicity of naturally occurring mutations. Hum Mol Genet. 2011;20:3376–85.CrossRef Todeschini AL, Dipietromaria A, L'Hote D, Boucham FZ, Georges AB, Pandaranayaka PJ, et al. Mutational probing of the forkhead domain of the transcription factor FOXL2 provides insights into the pathogenicity of naturally occurring mutations. Hum Mol Genet. 2011;20:3376–85.CrossRef
28.
go back to reference Saleem RA. Structural and functional analyses of disease-causing missense mutations in the forkhead domain of FOXC1. Hum Mol Genet. 2003;12:2993–3005.CrossRef Saleem RA. Structural and functional analyses of disease-causing missense mutations in the forkhead domain of FOXC1. Hum Mol Genet. 2003;12:2993–3005.CrossRef
29.
go back to reference Uhlenhaut NH, Jakob S, Anlag K, Eisenberger T, Sekido R. Somatic sex reprogramming of adult ovaries to testes by FOXL2 ablation. Uhlenhaut NH, Jakob S, Anlag K, Eisenberger T, Sekido R. Somatic sex reprogramming of adult ovaries to testes by FOXL2 ablation.
30.
go back to reference Georges A, L'Hote D, Todeschini AL, Auguste A, Legois B, Zider A, et al. The transcription factor FOXL2 mobilizes estrogen signaling to maintain the identity of ovarian granulosa cells. ELIFE. 2014;3. Georges A, L'Hote D, Todeschini AL, Auguste A, Legois B, Zider A, et al. The transcription factor FOXL2 mobilizes estrogen signaling to maintain the identity of ovarian granulosa cells. ELIFE. 2014;3.
31.
go back to reference Benayoun BA, Batista F, Auer J, Dipietromaria A, L'Hote D, De Baere E, et al. Positive and negative feedback regulates the transcription factor FOXL2 in response to cell stress: evidence for a regulatory imbalance induced by disease-causing mutations. Hum Mol Genet. 2009;18:632–44.CrossRef Benayoun BA, Batista F, Auer J, Dipietromaria A, L'Hote D, De Baere E, et al. Positive and negative feedback regulates the transcription factor FOXL2 in response to cell stress: evidence for a regulatory imbalance induced by disease-causing mutations. Hum Mol Genet. 2009;18:632–44.CrossRef
32.
go back to reference Benayoun BA, Georges AB, L'Hote D, Andersson N, Dipietromaria A, Todeschini AL, et al. Transcription factor FOXL2 protects granulosa cells from stress and delays cell cycle: role of its regulation by the SIRT1 deacetylase. Hum Mol Genet. 2011;20:1673–86.CrossRef Benayoun BA, Georges AB, L'Hote D, Andersson N, Dipietromaria A, Todeschini AL, et al. Transcription factor FOXL2 protects granulosa cells from stress and delays cell cycle: role of its regulation by the SIRT1 deacetylase. Hum Mol Genet. 2011;20:1673–86.CrossRef
Metadata
Title
A novel FOXL2 mutation in two infertile patients with blepharophimosis–ptosis–epicanthus inversus syndrome
Authors
Jingmei Hu
Hanni Ke
Wei Luo
Yajuan Yang
Hongli Liu
Guangyu Li
Yingying Qin
Jinlong Ma
Shidou Zhao
Publication date
01-01-2020
Publisher
Springer US
Published in
Journal of Assisted Reproduction and Genetics / Issue 1/2020
Print ISSN: 1058-0468
Electronic ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-019-01651-2

Other articles of this Issue 1/2020

Journal of Assisted Reproduction and Genetics 1/2020 Go to the issue