1
|
Federici S, Rossetti R, Moleri S, Munari EV, Frixou M, Bonomi M, Persani L. Primary ovarian insufficiency: update on clinical and genetic findings. Front Endocrinol (Lausanne) 2024; 15:1464803. [PMID: 39391877 PMCID: PMC11466302 DOI: 10.3389/fendo.2024.1464803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/02/2024] [Indexed: 10/12/2024] Open
Abstract
Primary ovarian insufficiency (POI) is a disorder of insufficient ovarian follicle function before the age of 40 years with an estimated prevalence of 3.7% worldwide. Its relevance is emerging due to the increasing number of women desiring conception late or beyond the third decade of their lives. POI clinical presentation is extremely heterogeneous with a possible exordium as primary amenorrhea due to ovarian dysgenesis or with a secondary amenorrhea due to different congenital or acquired abnormalities. POI significantly impacts non only on the fertility prospect of the affected women but also on their general, psychological, sexual quality of life, and, furthermore, on their long-term bone, cardiovascular, and cognitive health. In several cases the underlying cause of POI remains unknown and, thus, these forms are still classified as idiopathic. However, we now know the age of menopause is an inheritable trait and POI has a strong genetic background. This is confirmed by the existence of several candidate genes, experimental and natural models. The most common genetic contributors to POI are the X chromosome-linked defects. Moreover, the variable expressivity of POI defect suggests it can be considered as a multifactorial or oligogenic defect. Here, we present an updated review on clinical findings and on the principal X-linked and autosomal genes involved in syndromic and non-syndromic forms of POI. We also provide current information on the management of the premature hypoestrogenic state as well as on fertility preservation in subjects at risk of POI.
Collapse
Affiliation(s)
- Silvia Federici
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Raffaella Rossetti
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Silvia Moleri
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Elisabetta V. Munari
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Frixou
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Marco Bonomi
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Luca Persani
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
2
|
Issa NM, Elhady GM. Cytogenetic abnormalities in a sample of females with premature ovarian failure. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2022. [DOI: 10.1186/s43043-022-00098-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Premature ovarian failure (POF) is a complex heterogeneous disorder characterized by the triad of amenorrhea, hypergonadotropinism, and hypoestrogenism in women before the expected age of menopause. In most POF patients, the etiology is idiopathic. X chromosome abnormalities are known to be responsible for many POF cases but the effect of sex chromosome low level mosaicism on ovarian function still remains unclear. The aim of this study was to investigate the prevalence and type of cytogenetic abnormalities as well as low-level sex chromosome mosaicism in Egyptian females with POF.
Results
The present study recruited thirty women with POF and thirty women with normal reproductive history as a control group. Conventional cytogenetic analysis was carried out on POF patients in order to detect cytogenetic abnormalities. FISH on interphase and metaphase nuclei from patients with normal karyotype as well as from thirty control women with normal reproductive history was performed using X, Y, and 18 centromeric probes to evaluate low-level sex chromosome mosaicism. Conventional cytogenetic analysis of peripheral blood lymphocytes demonstrated chromosomal aberrations in 7 cases. FISH revealed that the rate of X chromosome mosaicism was significantly higher in POF patients than in the control group.
Conclusion
We concluded that X chromosome abnormalities including low level mosaicism may be underlying the pathology of POF as increased mosaicism may lead to accelerated oocyte aging and premature follicular atresia.
Collapse
|
3
|
Turkyilmaz A, Alavanda C, Ates EA, Geckinli BB, Polat H, Gokcu M, Karakaya T, Cebi AH, Soylemez MA, Guney Aİ, Ata P, Arman A. Whole-exome sequencing reveals new potential genes and variants in patients with premature ovarian insufficiency. J Assist Reprod Genet 2022; 39:695-710. [PMID: 35066699 PMCID: PMC8995228 DOI: 10.1007/s10815-022-02408-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/19/2022] [Indexed: 10/19/2022] Open
Abstract
PURPOSE Premature ovarian insufficiency (POI) is a heterogeneous disorder characterized by the cessation of menstrual cycles before the age of 40 years due to the depletion or dysfunction of the ovarian follicles. POI is a highly heterogeneous disease in terms of etiology. The aim of this study is to reveal the genetic etiology in POI patients. METHODS A total of 35 patients (mean age: 27.2 years) from 28 different families diagnosed with POI were included in the study. Karyotype, FMR1 premutation analysis, single nucleotide polymorphism (SNP) array, and whole-exome sequencing (WES) were conducted to determine the genetic etiology of patients. RESULTS A total of 35 patients with POI were first evaluated by karyotype analysis, and chromosomal anomaly was detected in three (8.5%) and FMR1 premutation was detected in six patients (17%) from two different families. A total of 29 patients without FMR1 premutation were included in the SNP array analysis, and one patient had a 337-kb deletion in the chromosome 6q26 region including PARK2 gene, which was thought to be associated with POI. Twenty-nine cases included in SNP array analysis were evaluated simultaneously with WES analysis, and genetic variant was detected in 55.1% (16/29). CONCLUSION In the present study, rare novel variants were identified in genes known to be associated with POI, which contribute to the mutation spectrum. The effects of detected novel genes and variations on different pathways such as gonadal development, meiosis and DNA repair, or metabolism need to be investigated by experimental studies. Molecular etiology allows accurate genetic counseling to the patient and family as well as fertility planning.
Collapse
Affiliation(s)
- Ayberk Turkyilmaz
- Department of Medical Genetics, School of Medicine, Karadeniz Technical University, Trabzon, Turkey.
| | - Ceren Alavanda
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Esra Arslan Ates
- grid.414850.c0000 0004 0642 8921Department of Medical Genetics, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Bilgen Bilge Geckinli
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Hamza Polat
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Mehmet Gokcu
- grid.31564.350000 0001 2186 0630Department of Medical Genetics, School of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Taner Karakaya
- Department of Medical Genetics, Isparta City Hospital, Isparta, Turkey
| | - Alper Han Cebi
- grid.31564.350000 0001 2186 0630Department of Medical Genetics, School of Medicine, Karadeniz Technical University, Trabzon, Turkey
| | - Mehmet Ali Soylemez
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ahmet İlter Guney
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Pinar Ata
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| | - Ahmet Arman
- grid.16477.330000 0001 0668 8422Department of Medical Genetics, School of Medicine, Marmara University, Istanbul, Turkey
| |
Collapse
|
4
|
Piedade KC, Spencer H, Persani L, Nelson LM. Optimizing Fertility in Primary Ovarian Insufficiency: Case Report and Literature Review. Front Genet 2021; 12:676262. [PMID: 34249096 PMCID: PMC8261244 DOI: 10.3389/fgene.2021.676262] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/12/2021] [Indexed: 12/31/2022] Open
Abstract
Primary ovarian insufficiency (POI) is a clinical spectrum of ovarian dysfunction. Overt POI presents with oligo/amenorrhea and hypergonadotropic hypogonadism before age 40 years. Overt POI involves chronic health problems to include increased morbidity and mortality related to estradiol deficiency and the associated osteoporosis and cardiovascular disease as well as psychological and psychiatric disorders related to the loss of reproductive hormones and infertility. Presently, with standard clinical testing, a mechanism for Overt POI can only be identified in about 10% of cases. Now discovery of new mechanisms permits an etiology to be identified in a research setting in 25-30% of overt cases. The most common genetic cause of Overt POI is premutation in FMR1. The associated infertility is life altering. Oocyte donation is effective, although many women prefer to conceive with their own ova. Surprisingly, the majority who have Overt POI still have detectable ovarian follicles (70%). The major mechanism of follicle dysfunction in Overt POI has been histologically defined by a prospective NIH study: inappropriate follicle luteinization due to the tonically elevated serum LH levels. A trial of physiologic hormone replacement therapy, clinically proven to suppress the elevated LH levels in these women, may improve follicle function and increase the chance of ovulation. Here, we report the case of a woman with Overt POI diagnosed at age 35 years. To attempt pregnancy, she elected a trial of intrauterine insemination (IUI) in conjunction with follicle monitoring and physiologic hormone replacement therapy. She conceived on the eighth cycle of treatment and delivered a healthy baby. Our report calls for a concerted effort to define the best methods by which to optimize fertility for women who have POI.
Collapse
Affiliation(s)
| | - Hillary Spencer
- Vanderbilt University Medical Center, Nashville, TN, United States
| | - Luca Persani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- Department of Endocrine and Metabolic Diseases, Istituto Auxologico Italiano, Milan, Italy
| | | |
Collapse
|
5
|
Shi X, Tang T, Lin Q, Liu H, Qin Y, Liang X, Cong P, Mo D, Liu X, Chen Y, He Z. Efficient generation of bone morphogenetic protein 15-edited Yorkshire pigs using CRISPR/Cas9†. Biol Reprod 2020; 103:1054-1068. [PMID: 32761111 DOI: 10.1093/biolre/ioaa138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/25/2020] [Accepted: 08/06/2020] [Indexed: 12/15/2022] Open
Abstract
Bone morphogenetic protein 15 (BMP15), a member of the transforming growth factor beta superfamily, plays an essential role in ovarian follicular development in mono-ovulatory mammalian species. Studies using a biallelic knockout mouse model revealed that BMP15 potentially has just a minimal impact on female fertility and ovarian follicular development in polyovulatory species. In contrast, our previous study demonstrated that in vivo knockdown of BMP15 significantly affected porcine female fertility, as evidenced by the dysplastic ovaries containing significantly decreased numbers of follicles and an increased number of abnormal follicles. This finding implied that BMP15 plays an important role in the regulation of female fertility and ovarian follicular development in polyovulatory species. To further investigate the regulatory role of BMP15 in porcine ovarian and follicular development, here, we describe the efficient generation of BMP15-edited Yorkshire pigs using CRISPR/Cas9. Using artificial insemination experiments, we found that the biallelically edited gilts were all infertile, regardless of different genotypes. One monoallelically edited gilt #4 (Δ66 bp/WT) was fertile and could deliver offspring with a litter size comparable to that of wild-type gilts. Further analysis established that the infertility of biallelically edited gilts was caused by the arrest of follicular development at preantral stages, with formation of numerous structurally abnormal follicles, resulting in streaky ovaries and the absence of obvious estrous cycles. Our results strongly suggest that the role of BMP15 in nonrodent polyovulatory species may be as important as that in mono-ovulatory species.
Collapse
Affiliation(s)
- Xuan Shi
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Tao Tang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Qiyuan Lin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Hongbo Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yufeng Qin
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xinyu Liang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Peiqing Cong
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Xiaohong Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| | - Zuyong He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, P.R. China
| |
Collapse
|
6
|
Vaz B, El Mansouri F, Liu X, Taketo T. Premature ovarian insufficiency in the XO female mouse on the C57BL/6J genetic background. Mol Hum Reprod 2020; 26:678-688. [PMID: 32634219 PMCID: PMC7473787 DOI: 10.1093/molehr/gaaa049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 06/17/2020] [Indexed: 12/15/2022] Open
Abstract
In humans, all but 1% of monosomy 45.X embryos die in utero and those who reach term suffer from congenital abnormalities and infertility termed Turner's syndrome (TS). By contrast, XO female mice on various genetic backgrounds show much milder physical defects and normal fertility, diminishing their value as an animal model for studying the infertility of TS patients. In this article, we report that XO mice on the C57BL/6J (B6) genetic background showed early oocyte loss, infertility or subfertility and high embryonic lethality, suggesting that the effect of monosomy X in the female germline may be shared between mice and humans. First, we generated XO mice on either a mixed N2(C3H.B6) or B6 genetic background and compared the number of oocytes in neonatal ovaries; N2.XO females retained 45% of the number of oocytes in N2.XX females, whereas B6.XO females retained only 15% of that in B6.XX females. Second, while N2.XO females were as fertile as N2.XX females, both the frequency of delivery and the total number of pups delivered by B6.XO females were significantly lower than those by B6.XX females. Third, after mating with B6 males, both N2.XO and B6.XO females rarely produced XO pups carrying paternal X chromosomes, although a larger percentage of embryos was found to be XO before implantation. Furthermore, B6.XO females delivered 20% XO pups among female progeny after mating with C3H males. We conclude that the impact of monosomy X on female mouse fertility depends on the genetic background.
Collapse
Affiliation(s)
- B Vaz
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada
| | - F El Mansouri
- Department of Surgery, McGill University, Montreal, QC H4A3J1, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC H4A3J1, Canada
| | - X Liu
- Department of Surgery, McGill University, Montreal, QC H4A3J1, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC H4A3J1, Canada
| | - T Taketo
- Department of Biology, McGill University, Montreal, QC H3A 1B1, Canada
- Department of Surgery, McGill University, Montreal, QC H4A3J1, Canada
- Research Institute of the McGill University Health Centre, Montreal, QC H4A3J1, Canada
- Department of Obstetrics & Gynecology, McGill University, Montreal, QC H4A3J1, Canada
| |
Collapse
|
7
|
Jo HC, Park JK, Baek JC, Park JE, Kang MY, Cho IA. Clinicopathological features of premature ovarian insufficiency associated with chromosome abnormalities. ACTA ACUST UNITED AC 2019. [DOI: 10.5734/jgm.2019.16.1.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Hyen Chul Jo
- Department of Obstetrics and Gynecology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Ji Kwon Park
- Department of Obstetrics and Gynecology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Jong Chul Baek
- Department of Obstetrics and Gynecology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Ji Eun Park
- Department of Obstetrics and Gynecology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - Min Young Kang
- Department of Obstetrics and Gynecology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon, Korea
| | - In Ae Cho
- Department of Obstetrics and Gynecology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Korea
| |
Collapse
|
8
|
Delcour C, Amazit L, Patino LC, Magnin F, Fagart J, Delemer B, Young J, Laissue P, Binart N, Beau I. ATG7 and ATG9A loss-of-function variants trigger autophagy impairment and ovarian failure. Genet Med 2019; 21:930-938. [PMID: 30224786 DOI: 10.1038/s41436-018-0287-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/17/2018] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Primary ovarian insufficiency (POI) is a frequent disorder that affects ~1% of women under 40 years of age. POI, which is characterized by the premature depletion of ovarian follicles and elevated plasma levels of follicle-stimulating hormone (FSH), leads to infertility. Although various etiological factors have been described, including chromosomal abnormalities and gene variants, most cases remain idiopathic. The aim of the present study was to identify and validate functionally new sequence variants in ATG (autophagy-related genes) leading to POI. METHODS We have reanalyzed, in silico, the exome sequencing data from a previously reported work performed in 69 unrelated POI women. Functional experiments using a classical hallmark of autophagy, the microtubule-associated protein 1 light chain 3β (LC3), were then used to link these genes to this lysosomal degradation pathway. RESULTS We venture a functional link between ATG7 and ATG9A variants and POI. We demonstrated that variant ATG7 and ATG9A led to a decrease in autophagosome biosynthesis and consequently to an impairment of autophagy, a key biological process implicated in the preservation of the primordial follicles forming the ovarian reserve. CONCLUSION Our results unveil that impaired autophagy is a novel pathophysiological mechanism involved in human POI.
Collapse
Affiliation(s)
- Clémence Delcour
- Inserm U1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Larbi Amazit
- Inserm U1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Institut Biomédical de Bicêtre, UMS-32, Le Kremlin-Bicêtre, France
| | - Liliana C Patino
- Center For Research in Genetics and Genomics (CIGGUR), GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, DC, Colombia
| | - Françoise Magnin
- Inserm U1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Jérôme Fagart
- Inserm U1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Brigitte Delemer
- Service d'Endocrinologie-Diabète-Nutrition, CHU de Reims-Hôpital Robert-Debré, Reims, France
| | - Jacques Young
- Inserm U1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Service d'Endocrinologie et des Maladies de la Reproduction, APHP, Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - Paul Laissue
- Center For Research in Genetics and Genomics (CIGGUR), GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, DC, Colombia
| | - Nadine Binart
- Inserm U1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France
- Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Isabelle Beau
- Inserm U1185, Faculté de Médecine Paris Sud, Le Kremlin-Bicêtre, France.
- Univ Paris Sud, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| |
Collapse
|
9
|
Rossetti R, Ferrari I, Bonomi M, Persani L. Genetics of primary ovarian insufficiency. Clin Genet 2016; 91:183-198. [PMID: 27861765 DOI: 10.1111/cge.12921] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/10/2016] [Accepted: 11/11/2016] [Indexed: 12/15/2022]
Abstract
Primary ovarian insufficiency (POI) is characterized by a loss of ovarian function before the age of 40 and account for one major cause of female infertility. POI relevance is continuously growing because of the increasing number of women desiring conception beyond 30 years of age, when POI prevalence is >1%. POI is highly heterogeneous and can present with ovarian dysgenesis and primary amenorrhea, or with secondary amenorrhea, and it can be associated with other congenital or acquired abnormalities. In most cases POI remains classified as idiopathic. However, the age of menopause is an inheritable trait and POI has a strong genetic component. This is confirmed by the existence of several candidate genes, experimental and natural models. The variable expressivity of POI defect may indicate that, this disease may frequently be considered as a multifactorial or oligogenic defect. The most common genetic contributors to POI are the X chromosome-linked defects. Here, we review the principal X-linked and autosomal genes involved in syndromic and non-syndromic forms of POI with the expectation that this list will soon be upgraded, thus allowing the possibility to predict the risk of an early age at menopause in families with POI.
Collapse
Affiliation(s)
- R Rossetti
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - I Ferrari
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - M Bonomi
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - L Persani
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| |
Collapse
|
10
|
Next generation sequencing identifies abnormal Y chromosome and candidate causal variants in premature ovarian failure patients. Genomics 2016; 108:209-215. [PMID: 27989800 DOI: 10.1016/j.ygeno.2016.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 10/24/2016] [Accepted: 10/28/2016] [Indexed: 12/31/2022]
Abstract
Premature ovarian failure (POF) is characterized by heterogeneous genetic causes such as chromosomal abnormalities and variants in causal genes. Recently, development of techniques made next generation sequencing (NGS) possible to detect genome wide variants including chromosomal abnormalities. Among 37 Korean POF patients, XY karyotype with distal part deletions of Y chromosome, Yp11.32-31 and Yp12 end part, was observed in two patients through NGS. Six deleterious variants in POF genes were also detected which might explain the pathogenesis of POF with abnormalities in the sex chromosomes. Additionally, the two POF patients had no mutation in SRY but three non-synonymous variants were detected in genes regarding sex reversal. These findings suggest candidate causes of POF and sex reversal and show the propriety of NGS to approach the heterogeneous pathogenesis of POF.
Collapse
|
11
|
Ferrari I, Bouilly J, Beau I, Guizzardi F, Ferlin A, Pollazzon M, Salerno M, Binart N, Persani L, Rossetti R. Impaired protein stability and nuclear localization ofNOBOXvariants associated with premature ovarian insufficiency. Hum Mol Genet 2016; 25:5223-5233. [DOI: 10.1093/hmg/ddw342] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/29/2016] [Indexed: 11/14/2022] Open
|
12
|
Chauhan P, Jaiswal SK, Lakhotia AR, Rai AK. Molecular cytogenetic characterization of two Turner syndrome patients with mosaic ring X chromosome. J Assist Reprod Genet 2016; 33:1161-8. [PMID: 27387888 DOI: 10.1007/s10815-016-0761-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/20/2016] [Indexed: 01/15/2023] Open
Abstract
PURPOSE In the present study, we reported two cases of TS with mosaic ring X chromosome showing common clinical characteristics of TS like growth retardation and ovarian dysfunction. The purpose of the present study was to cytogenetically characterize both cases. METHODS Whole blood culture and G-banding were performed for karyotyping the cases following standard protocol. Origin of the ring chromosome and degree of mosaicism were further determined by fluorescence in situ hybridization (FISH). Breakpoints and loss of genetic material in formation of different ring X chromosomes r (X) in cases were determined with the help of cytogenetic microarray. RESULTS Cases 1 and 2 with ring chromosome were cytogenetically characterized as 45, X [114]/46Xr (X) (p22.11q21.32) [116] and 45, X [170]/46, Xr (X) (p22.2q21.33) [92], respectively. Sizes of these ring X chromosomes were found to be ~75 and ~95 Mb in cases 1 and 2, respectively, using visual estimation as part of cytogenetic observation. In both cases, we observed breakpoints on Xq chromosome were within relatively narrow region between Xq21.33 and Xq22.1 compared to regions in previously reported cases associated with ovarian dysgenesis. CONCLUSIONS Our observation agrees with the fact that despite of large heterogeneity, severity of the cases with intact X-inactive specific transcript (XIST) is dependent on degree of mosaicism and extent of Xq deletion having crucial genes involved directly or indirectly in various physiological involving ovarian cyclicity.
Collapse
Affiliation(s)
- Pooja Chauhan
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sushil Kumar Jaiswal
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, India
| | | | - Amit Kumar Rai
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
13
|
Daan NMP, Fauser BCJM. Menopause prediction and potential implications. Maturitas 2015; 82:257-65. [PMID: 26278873 DOI: 10.1016/j.maturitas.2015.07.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 07/22/2015] [Indexed: 02/05/2023]
Abstract
Reproductive ageing in women is characterized by a decline in both the quantity and quality of oocytes. Menopause is reached upon exhaustion of the resting primordial follicle pool, occurring on average at 51 years of age (range 40-60 years). The mean global age at natural menopause (ANM) appears robust, suggesting a distinct genetic control. Accordingly, a strong correlation in ANM is observed between mothers and daughters. Few specific genetic determinants of ANM have been identified. Substantial efforts have been made to predict ANM by using anti-Müllerian hormone (AMH) levels. AMH serum concentrations at reproductive age predict ANM, but precision is currently limited. Early ANM is associated with early preceding fertility loss, whereas late menopause is associated with reduced morbidity and mortality later in life. Menopause affects various women's health aspects, including bone density, breast, the cardiovascular system, mood/cognitive function and sexual well-being. If the current trend of increasing human life expectancy persists, women will soon spend half their life postmenopause. Unfortunately, increased longevity does not coincide with an equal increase in years spend in good health. Future research should focus on determinants of long term health effects of ANM, and efforts to improve women's postmenopausal health and quality of life.
Collapse
Affiliation(s)
- Nadine M P Daan
- Department of Reproductive Medicine & Gynecology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Bart C J M Fauser
- Department of Reproductive Medicine & Gynecology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
14
|
Bouilly J, Roucher-Boulez F, Gompel A, Bry-Gauillard H, Azibi K, Beldjord C, Dodé C, Bouligand J, Mantel AG, Hécart AC, Delemer B, Young J, Binart N. New NOBOX mutations identified in a large cohort of women with primary ovarian insufficiency decrease KIT-L expression. J Clin Endocrinol Metab 2015; 100:994-1001. [PMID: 25514101 DOI: 10.1210/jc.2014-2761] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
CONTEXT Primary ovarian insufficiency (POI) is a major cause of anovulation and infertility in women. This disease affects 1% of women before 40 years, and several genetic causes have been reported. OBJECTIVE The aim of the study was to evaluate the prevalence of NOBOX mutations in a new large cohort of women with POI and to characterize these variants and identify a NOBOX novel target gene. PATIENTS AND METHODS A total of 213 unrelated patients with POI were screened for NOBOX mutations, and luciferase reporter assays were performed for the mutations identified. RESULTS We reported 3 novel and 2 recurrent heterozygous missense NOBOX rare variants found in 12 patients but not in 724 alleles from ethnic-matched individual women with occurrence of menopause at a normal age. Their functional impact had been tested on the classic growth differentiation factor-9 (GDF9) promoter and on KIT-L, a new NOBOX target gene. The p.Gly91Thr, p.Gly111Arg, p.Arg117Trp, p.Lys371Thr, and p.Pro619Leu mutations were deleterious for protein function. CONCLUSIONS In our series, 5.6% of the patients with POI displayed heterozygous NOBOX mutations. We demonstrate that KIT-L could be now a direct NOBOX target. These findings replicate the high prevalence of the association between the NOBOX rare variants and POI.
Collapse
Affiliation(s)
- Justine Bouilly
- Inserm U693 (J.B., J.B., A.G.M., J.Y., N.B.), Le Kremlin-Bicêtre, F-94276, France; Université Paris-Sud (J.B., J.Y., N.B.), Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre, F-94276, France; Service d'Hormonologie, d'Endocrinologie Moléculaire et Des Maladies Rares (F.R.-B.), Centre De Biologie et Pathologie Est, Université Lyon 1, 69677 Bron, France; Unité de Gynécologie Endocrinienne (A.G.), Université Paris-Descartes, l'Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, 75014 Paris, France; l'Assistance Publique-Hôpitaux de Paris (H.B-G., J.Y., N.B.), Hôpital de Bicêtre, Service d'Endocrinologie et des Maladies de la Reproduction, Le Kremlin-Bicêtre, F-94276, France; Centre d'Aide Médicale à la Procréation (H.B-G.), CHI 94000 Créteil, France; Service de Biochimie et Génétique Moléculaire (K.A., C.B., C.D.), Hôpital Cochin, l'Assistance Publique-Hôpitaux de Paris Université Paris-Descartes, 75006 Paris, France; l'Assistance Publique-Hôpitaux de Paris (J.B., A.G.M.), Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Le Kremlin-Bicêtre, F-94276, France; and Service d'Endocrinologie-Diabète-Nutrition (A.-C.H., B.D.), Centre Hospitalier Universitaire de Reims-Hôpital Robert-Debré, 51092 Reims, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Rudd D, Axelsen M, Epping EA, Andreasen N, Wassink T. Childhood-onset schizophrenia case with 2.2 Mb deletion at chromosome 3p12.2-p12.1 and two large chromosomal abnormalities at 16q22.3-q24.3 and Xq23-q28. Clin Case Rep 2015; 3:201-7. [PMID: 25914809 PMCID: PMC4405302 DOI: 10.1002/ccr3.192] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 07/07/2014] [Accepted: 08/02/2014] [Indexed: 01/27/2023] Open
Abstract
Childhood-onset schizophrenia is rare, comprising 1% of known schizophrenia cases. Here, we report a patient with childhood-onset schizophrenia who has three large chromosomal abnormalities: an inherited 2.2 Mb deletion of chromosome 3p12.2–p12.1, a de novo 16.7 Mb duplication of 16q22.3–24.3, and a de novo 43 Mb deletion of Xq23–q28.
Collapse
Affiliation(s)
- Danielle Rudd
- Interdisciplinary Graduate Program in Genetics, University of Iowa Iowa City, Iowa ; Department of Psychiatry, University of Iowa Iowa City, Iowa
| | - Michael Axelsen
- Department of Psychiatry, University of Iowa Iowa City, Iowa
| | - Eric A Epping
- Department of Psychiatry, University of Iowa Iowa City, Iowa
| | - Nancy Andreasen
- Department of Psychiatry, University of Iowa Iowa City, Iowa
| | - Thomas Wassink
- Department of Psychiatry, University of Iowa Iowa City, Iowa
| |
Collapse
|
16
|
|
17
|
Persani L, Rossetti R, Di Pasquale E, Cacciatore C, Fabre S. The fundamental role of bone morphogenetic protein 15 in ovarian function and its involvement in female fertility disorders. Hum Reprod Update 2014; 20:869-83. [PMID: 24980253 DOI: 10.1093/humupd/dmu036] [Citation(s) in RCA: 145] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND A large number of studies have contributed to understanding the general mechanisms driving ovarian folliculogenesis in humans and show a complex endocrine dialog between the central nervous system, the pituitary and the ovary, integrated by various intraovarian paracrine messages. The role of intraovarian paracrine regulation has acquired more relevance in the recent years owing to the discovery of previously unknown factors, such as the oocyte-derived bone morphogenetic protein (BMP)15. METHODS A thorough literature search was carried out in order to summarize what has been reported so far on the role of BMP15, and the BMP15 paralog, growth and differentiation factor 9 (GDF9), in ovarian function and female fertility. Research articles published in English until March 2014 were included. RESULTS The biological actions of BMP15 include: (i) the promotion of follicle growth and maturation starting from the primary gonadotrophin-independent phases of folliculogenesis; (ii) the regulation of follicular granulosa cell (GC) sensitivity to FSH action and the determination of ovulation quota; (iii) the prevention of GC apoptosis and (iv) the promotion of oocyte developmental competence. The existence of biologically active heterodimers with GDF9, and/or the synergistic co-operation of BMP15 and GDF9 homodimers are indeed relevant in this context. Experimental disruption of the bmp15 gene in mice resulted in a mild fertility defect limited to females, whereas natural missense mutations in ewes cause variable phenotypes (ranging from hyperprolificacy to complete sterility) depending on a fine gene dosage mechanism also involving GDF9. Strong evidence supports the concept that such a mechanism plays an important role in the regulation of ovulation rate across mammalian and non-mammalian species. Following the discovery of sheep fecundity genes, several research groups have focused on alterations in human BMP15 associated with primary ovarian insufficiency (POI) or polycystic ovary syndrome. Several variants of BMP15 are significantly associated with POI supporting their pathogenic role, but the underlying biological mechanism is still under investigation and of great interest in medicine. BMP15 maps to the Xp locus involved in the determination of the ovarian defect in Turner syndrome and significantly contributes to the determination of ovarian reserve. Pioneering studies in women undergoing controlled ovarian stimulation indicate that BMP15 may represent a marker of ovarian response or oocyte quality. CONCLUSIONS BMP15, an oocyte-derived growth and differentiation factor, is a critical regulator of folliculogenesis and GC activities. Variations in BMP15 gene dosage have a relevant influence on ovarian function and can account for several defects of female fertility. The modulation of BMP15 action may have interesting pharmacological perspectives and the analysis of BMP15 may become a useful marker in IVF procedures. Recent outcomes indicate that the close interactions of BMP15/GDF9 have a critical biological impact that should be taken into account in future studies.
Collapse
Affiliation(s)
- Luca Persani
- Department of Clinical Sciences & Community Health, University of Milan, 20100 Milan, Italy Laboratory of Endocrine & Metabolic Research and Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy
| | - Raffaella Rossetti
- Department of Clinical Sciences & Community Health, University of Milan, 20100 Milan, Italy
| | - Elisa Di Pasquale
- Institute of Genetic and Biomedical Research - UOS of Milan, National Research Council (CNR) and Istituto Clinico Humanitas, Via Manzoni 59, Rozzano, 20089 Milan, Italy
| | - Chiara Cacciatore
- Department of Clinical Sciences & Community Health, University of Milan, 20100 Milan, Italy Laboratory of Endocrine & Metabolic Research and Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia 20, 20149 Milan, Italy
| | - Stéphane Fabre
- INRA, Université de Toulouse INPT, UMR1388, Génétique, Physiologie et Systèmes D'Elevage, F-31326 Castanet-Tolosan, France
| |
Collapse
|
18
|
Homozygous inactivating mutation in NANOS3 in two sisters with primary ovarian insufficiency. BIOMED RESEARCH INTERNATIONAL 2014; 2014:787465. [PMID: 25054146 PMCID: PMC4098983 DOI: 10.1155/2014/787465] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/30/2014] [Accepted: 06/03/2014] [Indexed: 12/27/2022]
Abstract
Despite the increasing understanding of female reproduction, the molecular diagnosis of primary ovarian insufficiency (POI) is seldom obtained. The RNA-binding protein NANOS3 poses as an interesting candidate gene for POI since members of the Nanos family have an evolutionarily conserved function in germ cell development and maintenance by repressing apoptosis. We performed mutational analysis of NANOS3 in a cohort of 85 Brazilian women with familial or isolated POI, presenting with primary or secondary amenorrhea, and in ethnically-matched control women. A homozygous p.Glu120Lys mutation in NANOS3 was identified in two sisters with primary amenorrhea. The substituted amino acid is located within the second C2HC motif in the conserved zinc finger domain of NANOS3 and in silico molecular modelling suggests destabilization of protein-RNA interaction. In vitro analyses of apoptosis through flow cytometry and confocal microscopy show that NANOS3 capacity to prevent apoptosis was impaired by this mutation. The identification of an inactivating missense mutation in NANOS3 suggests a mechanism for POI involving increased primordial germ cells (PGCs) apoptosis during embryonic cell migration and highlights the importance of NANOS proteins in human ovarian biology.
Collapse
|
19
|
Úbeda F, Ohtsuki H, Gardner A, Baalen M. Ecology drives intragenomic conflict over menopause. Ecol Lett 2014; 17:165-74. [PMID: 24320989 PMCID: PMC3912906 DOI: 10.1111/ele.12208] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/19/2013] [Accepted: 09/30/2013] [Indexed: 12/28/2022]
Abstract
Menopause is the transition from reproductive to non-reproductive life well before natural death. Rather than involving a smooth, rapid change, it is normally preceded by a long period of erratic hormonal fluctuation that is accompanied by a plethora of unpleasant symptoms. Here, we (1) suggest that this turbulent period owes to conflict, between a woman's maternally inherited (MI) and paternally inherited (PI) genes, over the trade-off between reproduction and communal care; (2) perform a theoretical analysis to show that this conflict is resolved either through silencing or fluctuating expression of one of the genes; (3) highlight which of the symptoms preceding menopause may result from antagonistic co-evolution of MI and PI genes; (4) argue that ecological differences between ancestral human populations may explain the variability in menopause among different ethnic groups; (5) discuss how these insights may be used to inform family planning and cancer risk assessment based on a woman's ancestral background.
Collapse
Affiliation(s)
- Francisco Úbeda
- School of Biological Sciences, Royal Holloway University of LondonEgham, Surrey, TW20 0EX, UK
| | - Hisashi Ohtsuki
- Department of Evolutionary Studies of Biosystems, The Graduate University for Advanced StudiesShonan Village, Kanagawa, Hayama, 240-0193, Japan
| | - Andy Gardner
- School of Biology, University of St AndrewsSt Andrews, KY16 9TH, UK
| | - Minus Baalen
- School of Biological Sciences, Royal Holloway University of LondonEgham, Surrey, TW20 0EX, UK
- Department of Evolutionary Studies of Biosystems, The Graduate University for Advanced StudiesShonan Village, Kanagawa, Hayama, 240-0193, Japan
- School of Biology, University of St AndrewsSt Andrews, KY16 9TH, UK
| |
Collapse
|
20
|
Castronovo C, Rossetti R, Rusconi D, Recalcati MP, Cacciatore C, Beccaria E, Calcaterra V, Invernizzi P, Larizza D, Finelli P, Persani L. Gene dosage as a relevant mechanism contributing to the determination of ovarian function in Turner syndrome. Hum Reprod 2013; 29:368-79. [PMID: 24324027 PMCID: PMC3896225 DOI: 10.1093/humrep/det436] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
STUDY QUESTION What is the burden of X chromosome mosaicism in the occurrence of spontaneous menarche (SM) in Turner syndrome (TS)? SUMMARY ANSWER SM was significantly associated with X chromosome mosaicism in the TS patients; a mosaicism with around 10% euploid cell line may predict spontaneous pubertal development when determined by molecular-cytogenetic techniques on uncultivated tissues. WHAT IS KNOWN ALREADY Spontaneous puberty can be observed in a minority of patients with TS, more frequently, but not exclusively, in those with a high level of 46,XX/45,X mosaicism at standard karyotype. The genetic mechanisms contributing to ovarian function in TS patients are still not determined. However, submicroscopic X-linked and autosomal copy number variations (CNVs) have recently emerged as an important genetic risk category for premature ovarian insufficiency and may be involved in modulating the TS ovarian phenotype. STUDY DESIGN, SIZE, DURATION A group of 40 patients with a diagnosis of TS at conventional karyotyping participated in the study; 6 patients had SM and 34 patients had primary amenorrhoea (PA). All clinical data and the patients’ DNA samples were collected over the years at a single paediatric clinic. PARTICIPANTS/MATERIALS, SETTING, METHODS The patients' samples were used to perform both genetic (Copy Number Assay) and molecular-cytogenetic (array-CGH and iFISH, interphase-FISH) analyses in order to evaluate the X chromosome mosaicism rate and to detect possible rare CNVs of genes with a known or predicted role in female fertility. MAIN RESULTS AND THE ROLE OF CHANCE All TS patients showed variable percentages of the 46,XX lineage, but these percentages were higher in the SM group (P < 0.01). A mosaicism around 10% for the euploid cell line may predict spontaneous pubertal development when determined by molecular-cytogenetic techniques performed in uncultivated tissues. A few CNVs involving autosomal and X-linked ovary-related loci were identified by array-CGH analysis and confirmed by real-time quantitative PCR, including a BMP15 gene duplication at Xp11.22, a deletion interrupting the PAPPA gene at 9q33.1, and an intragenic duplication involving the PDE8A gene at 15q25.3. LIMITATIONS, REASONS FOR CAUTION This is a pilot study on a relatively small sample size and confirmation in larger TS cohorts may be required. The ovarian tissue could not be studied in any patients and in a subgroup of patients, the mosaicism was estimated in tissues of different embryonic origin. WIDER IMPLICATIONS OF THE FINDINGS The combined determination of X chromosome mosaicism by molecular and molecular-cytogenetic techniques may become useful for the prediction of SM in TS. The detection of CNVs in both X-linked and autosomal ovary-related genes further suggests gene dosage as a relevant mechanism contributing to the ovarian phenotype of TS patients. These CNVs may pinpoint novel candidates relevant to female fertility and generate further insights into the mechanisms contributing to ovarian function. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by Telethon Foundation (grant no: GGP09126 to L.P.), the Italian Ministry of the University and Research (grant number: 2006065999 to P.F.) and a Ministry of Health grant ‘Ricerca Corrente’ to IRCCS Istituto Auxologico Italiano (grant number: 08C704-2006). The authors have no conflict of interest to declare.
Collapse
Affiliation(s)
- Chiara Castronovo
- Medical Cytogenetics and Molecular Genetics Lab, IRCSS Istituto Auxologico Italiano, 20145 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Gao F, Zhang J, Wang X, Yang J, Chen D, Huff V, Liu YX. Wt1 functions in ovarian follicle development by regulating granulosa cell differentiation. Hum Mol Genet 2013; 23:333-41. [DOI: 10.1093/hmg/ddt423] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
22
|
Kalantari H, Madani T, Zari Moradi S, Mansouri Z, Almadani N, Gourabi H, Mohseni Meybodi A. Cytogenetic analysis of 179 Iranian women with premature ovarian failure. Gynecol Endocrinol 2013; 29:588-91. [PMID: 23656387 DOI: 10.3109/09513590.2013.788625] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The importance of chromosomal abnormalities in etiology of premature ovarian failure (POF) is well known but in many cases, POF still remains idiopathic. We investigated the frequency and type of chromosomal aberrations in Iranian women diagnosed with idiopathic POF. Standard cytogenetic analysis was carried out in a total of 179 patients. Karyotype analysis of these patients revealed that 161 (89.95%) patients had normal female karyotype and 18 (10.05%) patients had abnormal karyotypes. The abnormal karyotypes included sex reverse sex determining region Y (SRY) negative (five Cases), X chromosome mosaicism (five cases), abnormal X chromosomes (three cases), abnormal autosomes (three cases) and X-autosome translocation (two cases). The overall prevalence of chromosomal abnormalities was 10.05% in this first large-scale report of chromosomal aberrations in Iranian women with POF. The results confirm previous observations and emphasis on the critical role of X chromosome abnormalities as one of the possible etiologies for POF.
Collapse
Affiliation(s)
- Hamid Kalantari
- Department of Genetics at Reproductive Biomedicine Research Center, Royan Institute for Reproductive biomedicine, ACECR, Tehran, Iran
| | | | | | | | | | | | | |
Collapse
|
23
|
Zhao Z, Wei D, Mu Y, Qin Y, Li G, Cui L, Chen ZJ. Mutational analysis of SKP2 and P27 in Chinese Han women with premature ovarian failure. Reprod Biomed Online 2013; 27:104-6. [PMID: 23665265 DOI: 10.1016/j.rbmo.2013.03.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 03/28/2013] [Accepted: 03/28/2013] [Indexed: 10/27/2022]
Abstract
P27 and SKP2, a major regulator of P27, play a crucial role in ovarian function in mice. Both P27-deficient and SKP2-deficient female mice develop premature ovarian failure (POF). The coding regions of SKP2 and P27 were examined in 200 Chinese women with POF and 200 control volunteers. This study is the first to investigate SKP2 in POF. No plausible pathogenic mutations were detected. The results suggest that mutations in SKP2 and P27 are not common in Chinese Han women with POF.
Collapse
Affiliation(s)
- Zhiyi Zhao
- Center for Reproductive Medicine, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan 250021, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Saranya B, Kavitha Devi D, Chandra RS, Jayashankar M, Santhiya ST. Translocation t(X;11)(q22;q25) in a woman with premature ovarian failure. Sex Dev 2013; 7:216-21. [PMID: 23429171 DOI: 10.1159/000346958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2012] [Indexed: 11/19/2022] Open
Abstract
Genetic, autoimmune, environmental, iatrogenic, and idiopathic factors are known to cause premature ovarian failure (POF). This report describes an X;11 translocation, t(X;11)(q22;q25), in a woman diagnosed with POF. The FSH level was found to be elevated. Menstrual cycle was regular initially, and she had a spontaneous abortion at the 5th month of gestation at 16 years of age. Her mother was karyotypically normal while her father was not investigated. Male carriers of X;autosome translocations are mostly infertile, and hence the translocation is presumed to be of de novo origin. Fluorescence in situ hybridization using whole chromosome paint probes confirmed the rearrangement.
Collapse
Affiliation(s)
- B Saranya
- Department of Genetics, Dr. ALMPG Institute of Basic Medical Sciences, University of Madras, Chennai, India
| | | | | | | | | |
Collapse
|
25
|
Detailed clinical and molecular study of 20 females with Xq deletions with special reference to menstruation and fertility. Eur J Med Genet 2013; 56:1-6. [DOI: 10.1016/j.ejmg.2012.08.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 08/31/2012] [Indexed: 11/21/2022]
|
26
|
Matsumoto T, Sakari M, Okada M, Yokoyama A, Takahashi S, Kouzmenko A, Kato S. The androgen receptor in health and disease. Annu Rev Physiol 2012; 75:201-24. [PMID: 23157556 DOI: 10.1146/annurev-physiol-030212-183656] [Citation(s) in RCA: 166] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Androgens play pivotal roles in the regulation of male development and physiological processes, particularly in the male reproductive system. Most biological effects of androgens are mediated by the action of nuclear androgen receptor (AR). AR acts as a master regulator of downstream androgen-dependent signaling pathway networks. This ligand-dependent transcriptional factor modulates gene expression through the recruitment of various coregulator complexes, the induction of chromatin reorganization, and epigenetic histone modifications at target genomic loci. Dysregulation of androgen/AR signaling perturbs normal reproductive development and accounts for a wide range of pathological conditions such as androgen-insensitive syndrome, prostate cancer, and spinal bulbar muscular atrophy. In this review we summarize recent advances in understanding of the epigenetic mechanisms of AR action as well as newly recognized aspects of AR-mediated androgen signaling in both men and women. In addition, we offer a perspective on the use of animal genetic model systems aimed at eventually developing novel therapeutic AR ligands.
Collapse
Affiliation(s)
- Takahiro Matsumoto
- Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima 770-8503, Japan.
| | | | | | | | | | | | | |
Collapse
|
27
|
Bertini V, Viola D, Vitti P, Simi P, Valetto A. An idic(15) associated with POF (premature ovarian failure): molecular cytogenetic definition of a case and review of the literature. Gene 2012; 503:123-5. [PMID: 22575723 DOI: 10.1016/j.gene.2012.04.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 11/29/2011] [Accepted: 04/22/2012] [Indexed: 10/28/2022]
Abstract
We report on a 36-year-old infertile woman, presenting a premature ovarian failure with an otherwise normal female phenotype. Cytogenetic analyses showed the presence of a supernumerary marker chromosome, that was characterized by FISH (fluorescent in situ hybridization) and array CGH (comparative genomic hybridization). This marker chromosome was derived from chromosome 15, and contained only heterochromatic material. The Prader Willi/Angelman region was not present. No duplications of the 15q regions were detected by array CGH. Supernumerary markers of chromosome 15 have been reported in cases of infertility and amenorrhea, that is also described in cases with marker derived by other acrocentric chromosomes. The case here presented constitutes a further example that etiology of POF is not always associated with a defective gene, but in some cases oocytes atresia can be the consequence of the abnormal meiotic pairing of chromosomes.
Collapse
Affiliation(s)
- Veronica Bertini
- Cytogenetic and Molecular Genetic Unit, Children Department, A.O.U. Pisana, S. Chiara Hospital, Pisa, Italy
| | | | | | | | | |
Collapse
|
28
|
Carp HJ, Selmi C, Shoenfeld Y. The autoimmune bases of infertility and pregnancy loss. J Autoimmun 2012; 38:J266-74. [PMID: 22284905 DOI: 10.1016/j.jaut.2011.11.016] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 11/28/2011] [Accepted: 11/28/2011] [Indexed: 11/26/2022]
|
29
|
Gleicher N, Weghofer A, Barad DH. Cutting edge assessment of the impact of autoimmunity on female reproductive success. J Autoimmun 2011; 38:J74-80. [PMID: 21664106 DOI: 10.1016/j.jaut.2011.05.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/13/2011] [Accepted: 05/18/2011] [Indexed: 11/28/2022]
Abstract
There, likely, is no more controversial issue in reproductive medicine than the effects of autoimmunity on female reproductive success. Published studies are, therefore, often biased. We performed PubMed, Google Scholar and Medline searches for the years 2000-2010 under various key words and phrases, referring to effects of autoimmunity/autoimmune diseases on pregnancy/pregnancy outcomes/pregnancy rates/reproduction/reproductive outcomes/fertility/infertility/fertility treatments/infertility treatments, and a number of similar terms. Reference lists of selected manuscripts were evaluated for additional, potential references. All selected manuscripts were reviewed by at least one author (N.G.). Opinions were reached based on preferential review of only selected studies, which offered data, primarily developed in pursuit of unrelated scientific questions. Data from various medical fields point, surprisingly effectively, toward significant impacts of autoimmunity on female reproductive success. Autoimmunity not only increases miscarriage risks but also reduces female fecundity and infertility treatment success. A, likely, reason why differences of opinion have persisted is that effects are primarily observed in genetically predisposed women, with specific fragile X mental retardation 1 (FMR1) genotypes. This discovery coincides with recently increasing appreciation of the importance of the long arm of the X chromosome (Xq) in control of functional ovarian reserve (reflective of female fertility) and autoimmunity, with FMR1at Xq27.3, located at cross roads of both. Autoimmune effects on female reproductive success deserve recognition. Further investigations must not ignore patient stratification, based on ovarian FMR1 genotypes. Genetic definition of high-risk patients should lead to development of successful therapeutic interventions.
Collapse
|
30
|
Ochalski ME, Engle N, Wakim A, Ravnan BJ, Hoffner L, Rajkovic A, Surti U. Complex X chromosome rearrangement delineated by array comparative genome hybridization in a woman with premature ovarian insufficiency. Fertil Steril 2011; 95:2433.e9-15. [DOI: 10.1016/j.fertnstert.2011.03.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Revised: 03/21/2011] [Accepted: 03/23/2011] [Indexed: 10/18/2022]
|
31
|
Kuan LC, Su MT, Wu CM, Chen M, Kuo PL, Kuo TC. A family with Xq22.3q25 interstitial deletion and normal ovarian function. Fertil Steril 2011; 96:e29-34. [PMID: 21621767 DOI: 10.1016/j.fertnstert.2011.04.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 04/13/2011] [Accepted: 04/18/2011] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To investigate genomic changes in a family with deletion of X chromosome q22.3-q25 associated with normal constitutional and reproductive phenotypes. DESIGN Case report. SETTING Academic district hospital genetic laboratory. PATIENT(S) A family incidentally found to have deletion of X chromosome q22.3-q25. INTERVENTION(S) Cytogenetic analysis and array-based comparative genomic hybridization for amniotic fluid and peripheral blood lymphocyte of family members. MAIN OUTCOME MEASURE(S) Ovarian function and menstrual cycles. RESULT(S) The proband and two daughters showed deletion of Xq22.3q25. This region spans 17.4 Mb and contains 121 genes. CONCLUSION(S) Female subjects with deletion of Xq22.3q25 may present with normal constitutional and reproductive phenotypes.
Collapse
Affiliation(s)
- Long-Ching Kuan
- Department of Obstetrics and Gynecology, Kuo General Hospital, Tainan, Taiwan
| | | | | | | | | | | |
Collapse
|
32
|
Persani L, Rossetti R, Cacciatore C, Fabre S. Genetic defects of ovarian TGF-β-like factors and premature ovarian failure. J Endocrinol Invest 2011; 34:244-51. [PMID: 21297384 DOI: 10.1007/bf03347073] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Premature ovarian failure (POF) is an ovarian defect characterized by the premature depletion of ovarian follicles; POF affects approximately 1-2% of women under the age of 40 yr, thus representing one major cause of female infertility. POF relevance is continuously growing because women tend to conceive always more frequently beyond 30 yr. Frequently, POF is the end-stage of an occult process [primary ovarian insufficiency (POI)]. POI is a heterogeneous disease caused by a variety of mechanisms. Though the underlying cause remains unexplained in the majority of cases, several data indicate that POI has a strong genetic component. These data include the existence of several causal genetic defects in human, experimental, and natural models, as well as the frequent familiarity. The candidate genes are numerous, but POF remains unexplained in most of the cases. Several recent evidences have driven the attention of researchers on the possible involvement of various elements belonging to the transforming growth factor β family, which includes bone morphogenetic proteins, growth/differentiation factors, and inhibins. These peptides are produced by either the oocyte or granulosa cells to constitute a complex paracrine network within the ovarian follicle. Here, we review the studies reporting the genetic alterations of these factors in human and animal defects of ovarian folliculogenesis which support the fundamental roles played by these signals in ovarian morphogenesis and function.
Collapse
Affiliation(s)
- L Persani
- Department of Medical Sciences, University of Milan, Cusano, Milan, Italy.
| | | | | | | |
Collapse
|
33
|
Schuettler J, Peng Z, Zimmer J, Sinn P, von Hagens C, Strowitzki T, Vogt PH. Variable expression of the Fragile X Mental Retardation 1 (FMR1) gene in patients with premature ovarian failure syndrome is not dependent on number of (CGG)n triplets in exon 1. Hum Reprod 2011; 26:1241-51. [PMID: 21335413 DOI: 10.1093/humrep/der018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Increased expression of the Fragile X Mental Retardation 1 (FMR1) gene in blood cells has been claimed to be associated with variable (CGG)(n) triplet numbers in the 5' untranslated region of this gene. Increased CGG triplet numbers, including that of the so-called premutation range (n= 55-200), were shown to have a risk of <26% to impair ovarian reserve leading to primary ovarian insufficiency and premature ovarian failure (POF). METHODS DNA and RNA samples were isolated from 74 patients with idiopathic POF to evaluate quantitatively the expression of FMR1 in leukocytes and CGG triplet number on FMR1 gene alleles. mRNA levels were normalized and compared with those of control women. Expression of the encoded protein (FMRP) was analysed by immunohistochemistry on ovarian biopsy tissue sections. RESULTS A large variance of the FMR1 transcript level was found in the leukocyte RNA samples, but only in patients with POF, and this variability did not correlate to variance of CGG triplet numbers found on both FMR1 alleles (19 < n > 90). During normal folliculogenesis, FMRP is predominantly expressed in granulosa cells. CONCLUSIONS Our data suggest that FMR1 expression during human folliculogenesis is probably a quantitative trait. Proper function of FMRP in granulosa cells seems to depend on an optimal transcript level. All women with CGG triplet numbers outside the range associated with normal folliculogenesis (26 < n > 34) are therefore expected to have a relaxed FMR1 transcription control. FMR1 transcript levels in leukocytes might therefore be diagnostic for altered FMRP levels in granulosa cells, which will affect the process of folliculogenesis.
Collapse
Affiliation(s)
- J Schuettler
- Department of Gynecology Endocrinology, University Women Hospital, Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
34
|
Rajkiewicz M, Szlendak-Sauer K, Sulek A, Gawlik-Zawislak S, Krysa W, Radowicki S, Zaremba J. A molecular and cytogenetic investigation of FMR1 gene premutations in Polish patients with primary ovarian insufficiency. Eur J Obstet Gynecol Reprod Biol 2011; 155:176-9. [PMID: 21276648 DOI: 10.1016/j.ejogrb.2010.12.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 11/29/2010] [Accepted: 12/26/2010] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The aim of this study was to determine the prevalence of premutations in the FMR1 gene that cause primary ovarian insufficiency (POI) in a group of affected women. STUDY DESIGN Forty DNA samples were purified from peripheral blood collected from women with ovarian failure who were under 40 years of age. A routine cytogenetic test was performed to eliminate chromosomal aberrations as the cause of POI. The DNA was analysed by polymerase chain reaction (PCR) with primers specific to the FMR1 gene region. The PCR products were then separated in denaturing polyacrylamide gels using an ABI Prism 377 sequencer. RESULTS Cytogenetic analysis of the samples revealed two X/autosome translocations. DNA analysis identified FMR1 gene premutations in three patients. The frequency of X/autosome translocations in the studied group was 2/40 (5.0%), and the frequency of FMR1 gene premutations was 3/38 cases (7.9%). Thus, genetic tests allowed for the identification of POI in five (12.5%) out of 40 women. CONCLUSION FMR1 gene premutation is a common genetic cause of POI.
Collapse
Affiliation(s)
- Marta Rajkiewicz
- Department of Genetics, Institute of Psychiatry and Neurology, 9 Sobieskiego Street, 02-957 Warsaw, Poland.
| | | | | | | | | | | | | |
Collapse
|
35
|
Bhogal B, Jongens TA. Fragile X syndrome and model organisms: identifying potential routes of therapeutic intervention. Dis Model Mech 2010; 3:693-700. [PMID: 20682752 DOI: 10.1242/dmm.002006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fragile X syndrome (FXS) is a cognitive disorder caused by silencing of the fragile X mental retardation 1 gene (FMR1). Since the discovery of the gene almost two decades ago, most scientific contributions have focused on identifying the molecular function of the fragile X mental retardation protein (FMRP) and understanding how absence of FMR1 gene expression gives rise to the disease phenotypes. The use of model organisms has allowed rapid progression in the FXS field and has given insight into the molecular basis of the disease. The mouse and fly FXS models have enabled studies to identify potential targets and pathways for pharmacological treatment. Here, we briefly review the two primary FXS model systems and describe how studies in these organisms have led us closer to therapeutic treatments for patients afflicted with FXS.
Collapse
Affiliation(s)
- Balpreet Bhogal
- Department of Genetics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6145, USA
| | | |
Collapse
|
36
|
Abstract
Premature ovarian failure (POF) is a syndrome characterised by amenorrhoea, hypoestrogenism and hypergonadotropinism before the age of 40. It is a disorder affecting approximately 1% of women <40 years, 1/1,000 women by the age of 30 and 1/10,000 women by the age of 20. POF is not merely an early menopause. Up to 50% of the patients with POF will have intermittent and unpredictable ovarian function which may persist for some years. Heterogeneity of POF is also reflected by the variety of possible causes, including autoimmunity, toxics, drugs, radiation, infectious as well as genetic defects. HRT remains the cornerstone of treatment and the only proven method of achieving pregnancy in these patients is by ovum donation.
Collapse
Affiliation(s)
- Arif Kokcu
- IVF Center, Department of Obstetrics and Gynecology, School of Medicine, University of Ondokuz Mayis, Samsun, Turkey.
| |
Collapse
|
37
|
Ferreira SI, Matoso E, Pinto M, Almeida J, Liehr T, Melo JB, Carreira IM. X-chromosome terminal deletion in a female with premature ovarian failure: Haploinsufficiency of X-linked genes as a possible explanation. Mol Cytogenet 2010; 3:14. [PMID: 20646274 PMCID: PMC2916005 DOI: 10.1186/1755-8166-3-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 07/20/2010] [Indexed: 12/02/2022] Open
Abstract
Background Premature ovarian failure (POF) has repeatedly been associated to X-chromosome deletions. FMR1 gene premutation allele's carrier women have an increased risk for POF. We intent to determine the cause of POF in a 29 year old female, evaluating both of these situations. Methods Concomitant analysis of FMR1 gene CGG repeat number and karyotype revealed an X-chromosome terminal deletion. Fluorescence in situ further characterized the breakpoint. A methylation assay for FMR1 gene allowed to determine its methylation status, and hence, the methylation status of the normal X-chromosome. Results We report a POF patient with a 46,X,del(X)(q26) karyotype and with skewed X-chromosome inactivation of the structural abnormal X-chromosome. Conclusions Despite the hemizygosity of FMR1 gene, the patient does not present Fragile X syndrome features, since the normal X-chromosome is not subject to methylation. The described deletion supports the hypothesis that haploinsufficiency of X-linked genes can be on the basis of POF, and special attention should be paid to X-linked genes in region Xq28 since they escape inactivation and might have a role in this disorder. A full clinical and cytogenetic characterization of all POF cases is important to highlight a pattern and help to understand which genes are crucial for normal ovarian development.
Collapse
Affiliation(s)
- Susana I Ferreira
- Laboratório de Citogenética, Instituto de Biologia Médica, Faculdade de Medicina, Universidade de Coimbra, 3000-354 Coimbra, Portugal.
| | | | | | | | | | | | | |
Collapse
|
38
|
Wang B, Ni F, Li L, Wei Z, Zhu X, Wang J, Cao Y, Ma X. Analysis of cyclin-dependent kinase inhibitor 1B mutation in Han Chinese women with premature ovarian failure. Reprod Biomed Online 2010; 21:212-4. [PMID: 20615757 DOI: 10.1016/j.rbmo.2010.04.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 08/17/2009] [Accepted: 03/22/2010] [Indexed: 11/28/2022]
Abstract
The gene for cyclin-dependent kinase inhibitor 1B (CDKN1B; also known as P27(KIP1) and P27) encodes a cyclin-dependent kinase inhibitor and controls ovarian development in mice. In p27-deficient (p27(-/-)) mice, the overactivated follicular pool in ovaries was largely depleted, causing premature ovarian failure. The goal of this research was to investigate whether there are nucleotide changes in the CDKN1B gene of Han Chinese women with premature ovarian failure, as compared with control volunteers using candidate gene sequencing. No novel variations were found in exons encoding for CDKN1B. So the mutations in CDKN1B are not common with premature ovarian failure in Han Chinese women.
Collapse
Affiliation(s)
- Binbin Wang
- Graduate School, Peking Union Medical College, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Molecular cytogenetic definition of a translocation t(X;15) associated with premature ovarian failure. Fertil Steril 2010; 94:1097.e5-8. [PMID: 20338563 DOI: 10.1016/j.fertnstert.2010.02.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2009] [Revised: 02/02/2010] [Accepted: 02/04/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To characterize the breakpoints of a t(X;15) found in a woman with premature ovarian failure (POF). DESIGN Case report. SETTING Molecular and cytogenetics unit in a university-affiliated hospital. PATIENT(S) A 19-year-old infertile woman presenting with a normal female phenotype but primary amenorrhea. INTERVENTION(S) Molecular cytogenetic analyses and genetic counseling. MAIN OUTCOME MEASURE(S) Translocation t(X;15) defined by fluorescence in situ hybridization (FISH) and array comparative genomic hybridization (array CGH). RESULT(S) Chromosome and FISH analysis revealed 46,XX, t(X;15)(Xq22.1;p11); the active X was translocated and had been inherited from her mother. Detailed molecular characterization by FISH showed that the NXF5 (nuclear RNA export factor 5) gene was contained in the clone spanning the breakpoint on the X chromosome. CONCLUSION(S) The NXF5 gene is an appealing candidate for POF because it shows functional homology with the FMR1 (fragile X mental retardation 1) gene. Further analyses of its expression as well as mutation screening in other POF patients will help to elucidate its role.
Collapse
|
40
|
Walters KA, Simanainen U, Handelsman DJ. Molecular insights into androgen actions in male and female reproductive function from androgen receptor knockout models. Hum Reprod Update 2010; 16:543-58. [DOI: 10.1093/humupd/dmq003] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
41
|
Rio M, Malan V, Boissel S, Toutain A, Royer G, Gobin S, Morichon-Delvallez N, Turleau C, Bonnefont JP, Munnich A, Vekemans M, Colleaux L. Familial interstitial Xq27.3q28 duplication encompassing the FMR1 gene but not the MECP2 gene causes a new syndromic mental retardation condition. Eur J Hum Genet 2010; 18:285-90. [PMID: 19844254 PMCID: PMC2987214 DOI: 10.1038/ejhg.2009.159] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 05/22/2009] [Accepted: 07/27/2009] [Indexed: 11/09/2022] Open
Abstract
X-linked mental retardation is a common disorder that accounts for 5-10% of cases of mental retardation in males. Fragile X syndrome is the most common form resulting from a loss of expression of the FMR1 gene. On the other hand, partial duplication of the long arm of the X chromosome is uncommon. It leads to functional disomy of the corresponding genes and has been reported in several cases of mental retardation in males. In this study, we report on the clinical and genetic characterization of a new X-linked mental retardation syndrome characterized by short stature, hypogonadism and facial dysmorphism, and show that this syndrome is caused by a small Xq27.3q28 interstitial duplication encompassing the FMR1 gene. This family broadens the phenotypic spectrum of FMR1 anomalies in an unexpected manner, and we suggest that this condition may represent the fragile X syndrome "contre-type".
Collapse
Affiliation(s)
- Marlène Rio
- Département de Génétique, Université Paris Descartes, INSERM U781, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Voorhuis M, Onland-Moret NC, van der Schouw YT, Fauser BCJM, Broekmans FJ. Human studies on genetics of the age at natural menopause: a systematic review. Hum Reprod Update 2010; 16:364-77. [PMID: 20071357 DOI: 10.1093/humupd/dmp055] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Timing of natural menopause has great implications for fertility and women's health. Age at natural menopause (ANM) is largely influenced by genetic factors. In the past decade, several genetic studies have been conducted to identify genes in ANM, which can help us unravel the biological pathways underlying this trait and the associated infertility and health risks. After providing an overview of the results of the genetic studies performed so far, we give recommendations for future studies in identifying genetic factors involved in determining the variation in timing of natural menopause. METHODS The electronic databases of Pubmed and Embase were systematically searched until September 2009 for genetic studies on ANM, using relevant keywords on the subject. Additional papers identified through hand search were also included. RESULTS Twenty-eight papers emerged from our literature search. A number of genetic regions and variants involved in several possible pathways underlying timing of ANM were identified, including two possible interesting regions (9q21.3 and chromosome 8 at 26 cM) in linkage analyses. Recent genome-wide association studies have identified two genomic regions (19q13.42 and 20p12.3), containing two promising candidate genes (BRKS1 and MCM). In the candidate gene association studies on ANM, very few consistent associations were found. CONCLUSION A number of genetic variants have been discovered in association with ANM, although the overall results have been rather disappointing. We have described possible new strategies for future genetic studies to identify more genetic loci involved in the variation in menopausal age.
Collapse
Affiliation(s)
- Marlies Voorhuis
- Department of Reproductive Medicine and Gynaecology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
43
|
Persani L, Rossetti R, Cacciatore C, Bonomi M. Primary ovarian insufficiency: X chromosome defects and autoimmunity. J Autoimmun 2009; 33:35-41. [DOI: 10.1016/j.jaut.2009.03.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 03/03/2009] [Accepted: 03/11/2009] [Indexed: 01/08/2023]
|
44
|
Guo QS, Qin SY, Zhou SF, He L, Ma D, Zhang YP, Xiong Y, Peng T, Cheng Y, Li XT. Unbalanced translocation in an adult patient with premature ovarian failure and mental retardation detected by spectral karyotyping and array-comparative genomic hybridization. Eur J Clin Invest 2009; 39:729-37. [PMID: 19515099 DOI: 10.1111/j.1365-2362.2009.02141.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND There are only three cases of unbalanced translocation (X;1) reported in childhood in the literature, while no such phenotypic information is available in adults. MATERIALS AND METHODS To delineate the phenotype-genotype relationship of unbalanced translocation (X;1) in adulthood, we reported here a 20-year-old female with an unbalanced translocation (X;1) which was determined by spectral karyotyping, array-comparative genomic hybridization and subtelomeric fluorescence in situ hybridization (FISH). RESULTS The phenotype of partial trisomy 1 and partial monosomy X of the present case was much attenuated, including premature ovarian failure, mental retardation, class I obesity, mild dysmorphism and delayed secondary sexual characteristics. The breakpoints of the unbalanced translocation were accurately located at Xq28 and 1q32.1. The large amplification on Chromosome 1 q arm was found to involve 312 genes and the deletion on Chromosome X q arm also involved 141 genes. Overall, genes associated with physiological process (47 genes), cellular process (33), development (23), response to stimulus (1) and reproduction (1) were observed in the amplification on Chromosome 1 q arm. In addition, genes related to physiological process (23 genes), cellular process (13), development (6) and response to stimulus (2) were observed in the large deletion on chromosome X q arm. Late-replication studies revealed the existence of skewed X inactivation in the derivative X chromosome. CONCLUSIONS The phenotype of partial monosomy X and partial trisomy 1q is much attenuated in case of unbalanced translocation (X;1) in adulthood probably owing to skewed X inactivation in derivative X chromosome.
Collapse
Affiliation(s)
- Q S Guo
- Department of Maternal and Fetal Medicine, The Obstetrics & Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
X;7 Translocation in an Indian Woman with Hypergonadotropic Amenorrhea—A Case Report. Genet Test Mol Biomarkers 2009; 13:533-6. [DOI: 10.1089/gtmb.2009.0051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
46
|
Rossetti R, Di Pasquale E, Marozzi A, Bione S, Toniolo D, Grammatico P, Nelson LM, Beck-Peccoz P, Persani L. BMP15 mutations associated with primary ovarian insufficiency cause a defective production of bioactive protein. Hum Mutat 2009; 30:804-10. [PMID: 19263482 DOI: 10.1002/humu.20961] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Bone morphogenetic protein-15 (BMP15) is selectively synthesized by oocytes as a pre-proprotein and is considered an ovarian follicle organizer whose adequate function is critical for female fertility. Missense mutations were reported in primary ovarian insufficiency (POI) but their biological impact remained unexplored. Here, screening of 300 unrelated idiopathic overt POI women with primary or secondary amenorrhea (SA) led to the identification of six heterozygous BMP15 variations in 29 of them. All alterations are nonconservative and include one insertion of three nucleotides (p.L262_L263insL) and five missense substitutions. Except for the p.S5R located in the signal sequence, the other variants (p.R68W, p.R138H, p.L148P, and p.A180T) localize in the proregion, which is essential for the processing and secretion of bioactive dimers. The mutations p.R68W, p.L148P, and the novel p.R138H lead to marked reductions of mature protein production. Their biological effects, evaluated by a novel luciferase-reporter assay in a human granulosa cell (GC) line, were significantly reduced. Cotransfection experiments of defective mutants with equal amounts of wild-type BMP15 cDNA, thus reproducing the heterozygous state seen in patients, did not generate a complete recovery of wild-type activity. No or minor deleterious effects were detected for the variants p.L262_L263insL, p.A180T, or p.S5R. In conclusion, heterozygous BMP15 mutations associated with the early onset of overt POI lead to defective secretion of bioactive dimers. These findings support the concept that an adequate amount of BMP15 secreted in the follicular fluid is critical for female fertility. We propose to consider the screening of BMP15 mutations among the analyses for the prediction of POI risk.
Collapse
Affiliation(s)
- Raffaella Rossetti
- Department of Medical Sciences, Centro Interuniversitario per Ricerca delle Malattie della Riproduzione, University of Milan, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Perheentupa A, Huhtaniemi I. Aging of the human ovary and testis. Mol Cell Endocrinol 2009; 299:2-13. [PMID: 19059459 DOI: 10.1016/j.mce.2008.11.004] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2008] [Revised: 11/05/2008] [Accepted: 11/05/2008] [Indexed: 11/28/2022]
Abstract
Aging is associated with structural and functional alterations in all organs of the human body. The aging of gonads represents in this respect a special case, because these organs are not functional for the whole lifespan of an individual and their normal function is not indispensable for functions of the rest of the body. Ovarian function lasts for the reproductive life of a woman, i.e., from menarche until menopause. The testicular endocrine function, in contrast, begins already in utero, is interrupted between neonatal life and puberty, and continues thereafter along with spermatogenesis, with only slight decline, until old age. The aging processes of the ovary and testis are therefore very different. We describe in this review the structural and functional alterations in the human ovary and testis upon aging. Special emphasis will be given to clinically significant alterations, which in women concern the causes and consequences of the individual variability of fertility during the latter part of the reproductive age. The clinically important aspect of testicular aging entails the decline of androgen production in aging men.
Collapse
Affiliation(s)
- Antti Perheentupa
- Department of Obstetrics and Gynaecology, University of Turku, Kiinamyllynkatu 10, 20520 Turku, Finland
| | | |
Collapse
|
48
|
Gidoni Y, Holzer H, Tulandi T, Tan SL. Fertility preservation in patients with non-oncological conditions. Reprod Biomed Online 2008; 16:792-800. [PMID: 18549688 DOI: 10.1016/s1472-6483(10)60144-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In addition to cancers, many non-oncological conditions, including chromosomal abnormalities and autoimmune disorders, are currently treated with gonadotoxic agents that can lead to premature ovarian failure. Because of the young age of some of the women affected by these conditions, attempts to preserve fertility and ovarian function are recommended. To date, retrieval of immature oocytes followed by in-vitro maturation and vitrification has been found to be especially useful for women who cannot undergo ovarian stimulation, or when there is a contraindication.
Collapse
Affiliation(s)
- Yariv Gidoni
- Department of Obstetrics and Gynecology, McGill University, 687 Pine Avenue West, Montreal, H3A 1A1, Quebec, Canada
| | | | | | | |
Collapse
|
49
|
Kimura S, Matsumoto T, Matsuyama R, Shiina H, Sato T, Takeyama KI, Kato S. Androgen receptor function in folliculogenesis and its clinical implication in premature ovarian failure. Trends Endocrinol Metab 2007; 18:183-9. [PMID: 17442585 DOI: 10.1016/j.tem.2007.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2006] [Revised: 03/07/2007] [Accepted: 04/05/2007] [Indexed: 01/19/2023]
Abstract
The action of estrogen in the female reproductive organs is well known in terms of the expression pattern and gene regulation of the estrogen receptor (ER). The significance of ERs in female reproduction is undisputed. The role of the androgen receptor (AR) is less clear. Clinical hyperandrogenism, a typical feature of polycystic ovary syndrome (PCOS), highlights pathological androgen production by the ovary. By contrast, the physiological impact of androgen action in female reproductive organs remains elusive. Androgens affect folliculogenesis in a variety of experimental approaches and ARs are expressed in developing follicles. Recent observations have discovered that inactivation of ARs in female mice results in premature ovarian failure (POF), indicating that normal folliculogenesis requires AR-mediated androgen action. Moreover, these results imply that POF might be caused by impairment of AR-mediated androgen action.
Collapse
Affiliation(s)
- Shuhei Kimura
- Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
OBJECTIVES To present an updated review on the etiology, consequences and management of premature ovarian failure. DESIGN A search of the English language literature using the Cochrane Library database and Medline 1966-2006, with a hand search of the references. CONCLUSION Premature ovarian failure is defined as the occurrence of amenorrhea, hypergonadotropinemia and estrogen deficiency in women under the age of 40 years, with the prevalence being 0.9-1.2%. In the majority of cases, the etiology is unknown, but known causes include chemotherapy, radiotherapy, surgery, genetic disorders, particularly involving the X chromosome, associations with autoimmune diseases, infections, smoking and other toxins. The three critical issues of management in these women are the effect of the diagnosis on the psychological health of the patient, the consequent infertility and the long- and short-term effects of estrogen deficiency arising from ovarian decline. Promising methods of screening for premature ovarian failure are being developed.
Collapse
Affiliation(s)
- T A Nippita
- Department of Obstetrics and Gynaecology, Royal North Shore Hospital, St Leonards, and The University of Sydney, Australia
| | | |
Collapse
|