Genetic Aspects of Premature Ovarian Failure: a Literature Review

Total Page:16

File Type:pdf, Size:1020Kb

Genetic Aspects of Premature Ovarian Failure: a Literature Review See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/49711859 Genetic aspects of premature ovarian failure: A literature review Article in Archives of Gynecology · March 2011 DOI: 10.1007/s00404-010-1815-4 · Source: PubMed CITATIONS READS 56 185 5 authors, including: Emerson Barchi Cordts Denise Christofolini Faculdade de Medicina do ABC Faculdade de Medicina do ABC 19 PUBLICATIONS 92 CITATIONS 123 PUBLICATIONS 702 CITATIONS SEE PROFILE SEE PROFILE Bianca Bianco Caio Parente Barbosa Faculdade de Medicina do ABC Faculdade de Medicina do ABC 127 PUBLICATIONS 744 CITATIONS 121 PUBLICATIONS 625 CITATIONS SEE PROFILE SEE PROFILE All in-text references underlined in blue are linked to publications on ResearchGate, Available from: Bianca Bianco letting you access and read them immediately. Retrieved on: 15 September 2016 Genetic aspects of premature ovarian failure: a literature review Archives of Gynecology and Obstetrics ISSN 0932-0067 Volume 283 Number 3 Arch Gynecol Obstet (2010) 283:635-643 DOI 10.1007/ s00404-010-1815-4 1 23 Your article is protected by copyright and all rights are held exclusively by Springer- Verlag. This e-offprint is for personal use only and shall not be self-archived in electronic repositories. If you wish to self-archive your work, please use the accepted author’s version for posting to your own website or your institution’s repository. You may further deposit the accepted author’s version on a funder’s repository at a funder’s request, provided it is not made publicly available until 12 months after publication. 1 23 Author's personal copy Arch Gynecol Obstet (2011) 283:635–643 DOI 10.1007/s00404-010-1815-4 REPRODUCTIVE MEDICINE Genetic aspects of premature ovarian failure: a literature review Emerson Barchi Cordts • Denise Maria Christofolini • Aline Amaro dos Santos • Bianca Bianco • Caio Parente Barbosa Received: 27 September 2010 / Accepted: 8 December 2010 / Published online: 29 December 2010 Ó Springer-Verlag 2010 Abstract Keywords Premature ovarian failure Á Infertility Á Background The diagnosis of premature ovarian failure Genetics Á Chromosomal abnormalities Á Polymorphism (POF) is based on the finding of amenorrhea before the age of 40 years associated with follicle-stimulating hormone levels in the menopausal range. It is a heterogeneous dis- Introduction order affecting approximately 1% of women \40 years, 1:10,000 women by age 20 years and 1:1,000 women by Premature ovarian failure (POF) (MIM—311360), or pre- age 30 years. POF is generally characterized by low levels mature ovarian insufficiency, is an early ovarian dysfunc- of gonadal hormones (estrogens and inhibins) and high tion clinically defined as the cessation of ovarian function levels of gonadotropins (LH and FSH) (hypergonadotropic with elevated gonadotrophin and low estrogen level before amenorrhea). or at the age of 40 years [1]. This condition is character- Methods Review of significant articles regarding genetic ized by the presence of primary or secondary amenorrhea causes that are associated with POF. for at least 4 months, hypoestrogenism and elevated serum Results Heterogeneity of POF is reflected by a variety of gonadotropin concentrations. The diagnosis is confirmed possible causes, including autoimmunity, toxics, drugs, as by two blood tests at least 1 month apart to measure FSH well as genetic defects. Changes at a single autosomal [2–4]. locus and many X-linked loci have been implicated in POF incidence in patients with 46, XX karyotype was women with POF. X chromosome abnormalities (e.g., estimated in around 1:1,000 women under 30 years old, Turner syndrome) represent the major cause of primary 1:250 around 35 years old and 1:100 at 40 years old [5]. amenorrhea associated with ovarian dysgenesis. Many Multiple causes of POF can be defined and result in fol- genes have been involved in POF development, among licle reduction and/or defects in the follicular development them BMP15, FMR1, FMR2, LHR, FSHR, INHA, FOXL2, stimulus mechanism [5]. Ovarian dysfunction can be sec- FOXO3, ERa, SF1, ERb and CYP19A1 genes. ondary to autoimmune diseases, infections (e.g., mumps), Conclusion Despite the description of several candidate chemotherapy and radiation treatment and metabolic dis- genes, the cause of POF remains undetermined in the vast eases (e.g., galactosemia), but for most of the cases, the majority of cases. etiology is idiopathic and probably genetic [6]. The genetic basis to the disease is supported by the occurrence of families with several affected women [3, 7–9]. Regarding the genetic causes of POF, they can be chromosomal or caused by single genes, involving the X E. B. Cordts Á D. M. Christofolini Á A. A. dos Santos Á chromosome or autosomes [10]. The X chromosome B. Bianco Á C. P. Barbosa (&) abnormalities represent 13% of the cases, followed by the Division of Human Reproduction, Department of Gynecology FMR1 premutation that represents 6% of the cases [11, and Obstetrics, Faculdade de Medicina do ABC, Avenida Prı´ncipe de Gales, 821, Santo Andre´, SP CEP 09060-650, Brazil 12]. Besides, there are many reports of mutations and e-mail: [email protected] polymorphisms in genes related to the sporadic form of 123 Author's personal copy 636 Arch Gynecol Obstet (2011) 283:635–643 the disease that will be discussed in details in this as genitourinary abnormalities, ranging from unilateral manuscript. kidney and renal dysplasia to ovarian malformations [19]. Pubertal onset and sexual development are usually normal in trisomy X; however, there have been cases of ovarian or The X chromosome defects uterine dysgenesis described in children and young adults with trisomy X [17]. Defects involving large aberrations on the X chromosome There are multiple case reports of women with trisomy have been associated with POF including complete dele- X found to have POF, with endocrine findings of hyper- tion of one X (Turner syndrome), trisomy X, partial dele- gonadotropic hypogonadism. The ages of these cases have tions or X/autosome translocations [10]. ranged from 19 to 40 years [20]. Studies on the prevalence of POF in adolescents or adults with trisomy X have not yet been performed. One study that performed genetic Turner syndrome screening on women presenting with POF identified tri- somy X in 3% of cases [21]. In trisomy X, a large per- The Turner syndrome has an incidence of 1 in 2,500 centage of the reported cases of POF have also been females and is characterized cytogenetically by X chro- associated with autoimmune diseases, including autoim- mosome monosomy (45,X). In about 60% of cases, how- mune thyroid disorder [21]. ever, in addition to the 45,X cell line, another cell line is observed that has the complete chromosome number but presents one structurally abnormal X or Y chromosome The X chromosomes rearrangements [13, 14]. Clinically, the syndrome is characterized by gonadal dysgenesis with primary amenorrhea, sexual There is a ‘‘critical region’’ for ovarian development and infantilism, webbing of the neck, cubitus valgus and short function on the long arm of the X chromosome that ranges stature in phenotypic women [14]. Ovarian failure in TS is from Xq13.3 to q27. Alternative mechanisms proposed for due to accelerated follicular atresia, usually manifesting in the explanation of the ovarian defect account for the size of childhood but sometimes later in life [15]. Infertility in the critical Xq region. They include the direct disruption of 45,X patients is caused by oocyte loss in the early stages of relevant loci or a ‘position effect’ caused by the rear- the meiotic prophase, before the pachytene meiotic stage, rangements on contiguous genes. The ‘position effect’ is a resulting in ovarian dysgenesis and streak ovaries. Ogata mechanism involving the deletion or translocation of reg- and Matsuo [16] argued that ovarian failure in X monos- ulatory domains to different position on the genome that omies could be caused by non-specific pairing errors at might cause changes in gene transcription [22]. However, it meiosis that increase the probability of germ cell atresia— has been stated that deletions of the short arm of the X the extent of ovarian failure correlates with the extent of chromosome usually result in primary amenorrhea, pairing failure. whereas deletions of the long arm of the X chromosome result in either primary or secondary ovarian failure [23]. Consequently, both the short and long arm of the X chro- Trisomy X mosome seem to contain important genes for ovarian function. Trisomy X (47,XXX) is a sex chromosome aneuploidy condition and occurs in approximately 1 in 1,000 female births; however, it is estimated that only approximately Genes involved in premature ovarian failure 10% of cases are diagnosed. Although non-mosaic 47,XXX karyotypes are the most frequent, mosaicism occurs in Many genes have been involved in POF development. approximately 10% of cases and in many combinations The genes responsible for the most number of cases such as 46,XX/47,XXX or 47,XXX/48,XXXX, or in are described on Table 1 according to chromosomal combinations including Turner syndrome cell lines such as disposition. 45,X/47,XXX or 45,X/46,XX/47,XXX [17]. Clinical characteristics include epicanthal folds, hypertelorism, BMP15 (bone morphogenetic protein 15) upslanting palpebral fissures, clinodactyly, overlapping digits, pes planus and pectus excavatum. Hypotonia and BMP15, located at Xp11.2, is a member of the large joint hyperextensibility may also be present [18]. Although superfamily of the transforming growth factor b (TGFb) major medical problems are not present in most cases, proteins involved in diverse cellular processes during some medical problems may be associated with trisomy X embryonic development and tissue formation [24]. Studies 123 Author's personal copy Arch Gynecol Obstet (2011) 283:635–643 637 Table 1 The genes involved in POF development The human mutation was shown to act as dominant nega- tive and to decrease in vitro growth of granulosa cells after Gene Chromosomelocation stimulation with wild-type BMP15.
Recommended publications
  • Regulation of Sex Determination in Mice by a Non-Coding Genomic Region
    HIGHLIGHTED ARTICLE GENETICS OF SEX Regulation of Sex Determination in Mice by a Non-coding Genomic Region Valerie A. Arboleda,* Alice Fleming,* Hayk Barseghyan,* Emmanuèle Délot,*,† Janet S. Sinsheimer,*,‡ and Eric Vilain*,†,§,1 *Department of Human Genetics, †Department of Pediatrics, ‡Department of Biomathematics, and §Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, California 90095-7088 ABSTRACT To identify novel genomic regions that regulate sex determination, we utilized the powerful C57BL/6J-YPOS (B6-YPOS) model of XY sex reversal where mice with autosomes from the B6 strain and a Y chromosome from a wild-derived strain, Mus domesticus poschiavinus (YPOS), show complete sex reversal. In B6-YPOS, the presence of a 55-Mb congenic region on chromosome 11 protects from sex reversal in a dose-dependent manner. Using mouse genetic backcross designs and high-density SNP arrays, we narrowed the congenic region to a 1.62-Mb genomic region on chromosome 11 that confers 80% protection from B6-YPOS sex reversal when one copy is present and complete protection when two copies are present. It was previously believed that the protective congenic region originated from the 129S1/SviMJ (129) strain. However, genomic analysis revealed that this region is not derived from 129 and most likely is derived from the semi-inbred strain POSA. We show that the small 1.62-Mb congenic region that protects against B6-YPOS sex reversal is located within the Sox9 promoter and promotes the expression of Sox9, thereby driving testis de- velopment within the B6-YPOS background. Through 30 years of backcrossing, this congenic region was maintained, as it promoted male sex determination and fertility despite the female-promoting B6-YPOS genetic background.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Suppression of Notch Signaling Stimulates Progesterone Synthesis by Enhancing the Expression of NR5A2 and NR2F2 in Porcine Granulosa Cells
    G C A T T A C G G C A T genes Article Suppression of Notch Signaling Stimulates Progesterone Synthesis by Enhancing the Expression of NR5A2 and NR2F2 in Porcine Granulosa Cells Rihong Guo 1,2, Fang Chen 2 and Zhendan Shi 1,2,* 1 Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; [email protected] 2 Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China; [email protected] * Correspondence: [email protected] Received: 16 December 2019; Accepted: 18 January 2020; Published: 22 January 2020 Abstract: The conserved Notch pathway is reported to be involved in progesterone synthesis and secretion; however, the exact effects remain controversial. To determine the role and potential mechanisms of the Notch signaling pathway in progesterone biosynthesis in porcine granulosa cells (pGCs), we first used a pharmacological γ-secretase inhibitor, N-(N-(3,5-difluorophenacetyl-l-alanyl))-S-phenylglycine t-butyl ester (DAPT), to block the Notch pathway in cultured pGCs and then evaluated the expression of genes in the progesterone biosynthesis pathway and key transcription factors (TFs) regulating steroidogenesis. We found that DAPT dose- and time-dependently increased progesterone secretion. The expression of steroidogenic proteins NPC1 and StAR and two TFs, NR5A2 and NR2F2, was significantly upregulated, while the expression of HSD3B was significantly downregulated. Furthermore, knockdown of both NR5A2 and NR2F2 with specific siRNAs blocked the upregulatory effects of DAPT on progesterone secretion and reversed the effects of DAPT on the expression of NPC1, StAR, and HSD3B.
    [Show full text]
  • Journal.Pbio.2005886 August 10, 2018 1 / 47 Muscle Clock Directs Diurnal Metabolism
    RESEARCH ARTICLE Transcriptional programming of lipid and amino acid metabolism by the skeletal muscle circadian clock Kenneth Allen Dyar1,2*, MichaeÈl Jean Hubert1, Ashfaq Ali Mir1, Stefano Ciciliot2, Dominik Lutter1, Franziska Greulich1, Fabiana Quagliarini1, Maximilian Kleinert1, Katrin Fischer1, Thomas Oliver Eichmann3, Lauren Emily Wright4, Marcia Ivonne Peña Paz2, Alberto Casarin5, Vanessa Pertegato5, Vanina Romanello2, Mattia Albiero2, Sara Mazzucco6, Rosario Rizzuto4, Leonardo Salviati5, Gianni Biolo6, Bert Blaauw2,4, a1111111111 Stefano Schiaffino2, N. Henriette Uhlenhaut1,7* a1111111111 1 Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), Institute for a1111111111 Diabetes and Obesity (IDO), Munich, Germany, 2 Venetian Institute of Molecular Medicine (VIMM), Padova, a1111111111 Italy, 3 Institute of Molecular Biosciences, University of Graz, Graz, Austria, 4 Department of Biomedical a1111111111 Sciences, University of Padova, Padova, Italy, 5 Clinical Genetics Unit, Department of Woman and Child Health, University of Padova, and IRP Città della Speranza, Padova, Italy, 6 Clinica Medica, Department of Medical Sciences, University of Trieste, Trieste, Italy, 7 Gene Center, Ludwig-Maximilians-Universitaet (LMU), Munich, Germany * [email protected] (KAD); [email protected] (NHU) OPEN ACCESS Citation: Dyar KA, Hubert MJ, Mir AA, Ciciliot S, Lutter D, Greulich F, et al. (2018) Transcriptional programming of lipid and amino acid metabolism Abstract by the skeletal
    [Show full text]
  • Epigenetic Services Citations
    Active Motif Epigenetic Services Publications The papers below contain data generated by Active Motif’s Epigenetic Services team. To learn more about our services, please give us a call or visit us at www.activemotif.com/services. Technique Target Journal Year Reference Justin C. Boucher et al. CD28 Costimulatory Domain- ATAC-Seq, Cancer Immunol. Targeted Mutations Enhance Chimeric Antigen Receptor — 2021 RNA-Seq Res. T-cell Function. Cancer Immunol. Res. doi: 10.1158/2326- 6066.CIR-20-0253. Satvik Mareedu et al. Sarcolipin haploinsufficiency Am. J. Physiol. prevents dystrophic cardiomyopathy in mdx mice. RNA-Seq — Heart Circ. 2021 Am J Physiol Heart Circ Physiol. doi: 10.1152/ Physiol. ajpheart.00601.2020. Gabi Schutzius et al. BET bromodomain inhibitors regulate Nature Chemical ChIP-Seq BRD4 2021 keratinocyte plasticity. Nat. Chem. Biol. doi: 10.1038/ Biology s41589-020-00716-z. Siyun Wang et al. cMET promotes metastasis and ChIP-qPCR FOXO3 J. Cell Physiol. 2021 epithelial-mesenchymal transition in colorectal carcinoma by repressing RKIP. J. Cell Physiol. doi: 10.1002/jcp.30142. Sonia Iyer et al. Genetically Defined Syngeneic Mouse Models of Ovarian Cancer as Tools for the Discovery of ATAC-Seq — Cancer Discovery 2021 Combination Immunotherapy. Cancer Discov. doi: doi: 10.1158/2159-8290 Vinod Krishna et al. Integration of the Transcriptome and Genome-Wide Landscape of BRD2 and BRD4 Binding BRD2, BRD4, RNA Motifs Identifies Key Superenhancer Genes and Reveals ChIP-Seq J. Immunol. 2021 Pol II the Mechanism of Bet Inhibitor Action in Rheumatoid Arthritis Synovial Fibroblasts. J. Immunol. doi: doi: 10.4049/ jimmunol.2000286. Daniel Haag et al.
    [Show full text]
  • Supplementary File 1
    Supplementary Materials Figure S1. RNA sequencing quality control report. The upper panel showed per base sequence quality of (a) miRNA from 786-O and ACHN, and (b) RNA from 786-O and ACHN; the lower panel showed the per sequence quality scores of (c) miRNA from 786-O and ACHN, and (d) RNA from 786-O and ACHN. The quality score from 15 to 99 bp fell into the green background, indicating good quality calls. The majority read > 35 indicated good sequencing quality. Table S1. The list of miRNA with significant changes (ACHN vs 786-O) #miRNA Fold Change #miRNA Fold Change #miRNA Fold Change hsa-let-7a-2-3p -490.00 hsa-miR-192-5p 21.37 hsa-miR-323a-3p 135.76 hsa-let-7c-3p -10.30 hsa-miR-194-3p 31.18 hsa-miR-323b-3p 53.75 hsa-miR-100-3p -26.39 hsa-miR-194-5p 21.87 hsa-miR-326 -16.46 hsa-miR-100-5p -63.93 hsa-miR-195-5p 8.31 hsa-miR-329-3p 101.81 hsa-miR-105-5p -288.00 hsa-miR-196a-3p 5.28 hsa-miR-329-5p 436 hsa-miR-1185-1-3p 32.08 hsa-miR-199b-5p -9.75 hsa-miR-335-3p 73.67 hsa-miR-1185-2-3p 37.22 hsa-miR-203a-3p 222.25 hsa-miR-335-5p 71.58 hsa-miR-1185-5p 91.13 hsa-miR-205-5p -5.12 hsa-miR-337-3p 1696 hsa-miR-1197 7.38 hsa-miR-20b-5p 5.28 hsa-miR-337-5p 12141 hsa-miR-125b-1-3p -301.58 hsa-miR-211-5p 118 hsa-miR-34b-3p 1110.62 hsa-miR-125b-2-3p 5.91 hsa-miR-215-5p 18.88 hsa-miR-34b-5p 796.38 hsa-miR-1262 16.28 hsa-miR-218-5p 14.37 hsa-miR-34c-3p 10751 hsa-miR-1266-5p 6.62 hsa-miR-224-5p 118 hsa-miR-34c-5p 836.81 hsa-miR-127-3p 1723.39 hsa-miR-2277-3p 6.65 hsa-miR-3614-5p 141 hsa-miR-127-5p 4334.00 hsa-miR-2682-3p 6.63 hsa-miR-3617-5p -140 hsa-miR-1271-5p
    [Show full text]
  • Hes1 in the Somatic Cells of the Murine Ovary Is Necessary for Oocyte Survival and Maturation
    Developmental Biology 375 (2013) 140–151 Contents lists available at SciVerse ScienceDirect Developmental Biology journal homepage: www.elsevier.com/locate/developmentalbiology Hes1 in the somatic cells of the murine ovary is necessary for oocyte survival and maturation Iris Manosalva a,b,n, Aitor Gonza´lez a,b,1, Ryoichiro Kageyama a,b,n a Institute for Virus Research, Kyoto University, Kyoto, Japan b Japan Science and Technology Agency, CREST, Kyoto, Japan article info abstract Article history: The Notch pathway plays an important role in ovary development in invertebrates like Drosophila. Received 25 February 2012 However its role for the mammalian ovary is unclear. Mammalian Hes genes encode transcriptional Received in revised form factors that mediate many of the activities of the Notch pathway. Here, we have studied the function of 18 December 2012 Hes1 during embryonic development of the mouse ovary. We find that Hes1 protein is present in Accepted 19 December 2012 somatic cells and oocyte cytoplasm and decreases between E15.5 and P0. Conventional Hes1 knock-out Available online 27 December 2012 (KO), Hes1 conditional KO in the ovarian somatic, and chemical inhibition of Notch signaling decrease Keywords: the total number, size and maturation of oocytes and increase the number of pregranulosa cells at P0. Mouse These defects correlate with abnormal proliferation and enhanced apoptosis. Expression of the Ovary proapoptotic gene Inhbb is increased, while the levels of the antiapoptotic and oocyte maturation Notch pathway marker Kit are decreased in the Hes1 KO ovaries. Conversely, overactivation of the Notch pathway Hes1 Oocytes in ovarian somatic cells increases the number of mature oocytes and decreases the number of Fertility pregranulosa cells.
    [Show full text]
  • Supplement. Transcriptional Factors (TF), Protein Name and Their Description Or Function
    Supplement. Transcriptional factors (TF), protein name and their description or function. TF Protein name TF description/function ARID3A AT rich interactive domain 3A (BRIGHT-like) This gene encodes a member of the ARID (AT-rich interaction domain) family of DNA binding proteins. ATF4 Activating Transcription Factor 4 Transcriptional activator. Binds the cAMP response element (CRE) (consensus: 5-GTGACGT[AC][AG]-3), a sequence present in many viral and cellular promoters. CTCF CCCTC-Binding Factor Chromatin binding factor that binds to DNA sequence specific sites. Involved in transcriptional regulation by binding to chromatin insulators and preventing interaction between promoter and nearby enhancers and silencers. The protein can bind a histone acetyltransferase (HAT)-containing complex and function as a transcriptional activator or bind a histone deacetylase (HDAC)-containing complex and function as a transcriptional repressor. E2F1-6 E2F transcription factors 1-6 The protein encoded by this gene is a member of the E2F family of transcription factors. The E2F family plays a crucial role in the control of cell cycle and action of tumor suppressor proteins and is also a target of the transforming proteins of small DNA tumor viruses. The E2F proteins contain several evolutionally conserved domains found in most members of the family. These domains include a DNA binding domain, a dimerization domain which determines interaction with the differentiation regulated transcription factor proteins (DP), a transactivation domain enriched in acidic amino acids, and a tumor suppressor protein association domain which is embedded within the transactivation domain. EBF1 Transcription factor COE1 EBF1 has been shown to interact with ZNF423 and CREB binding proteins.
    [Show full text]
  • Integrated Single-Nucleotide and Structural Variation Signatures Of
    bioRxiv preprint doi: https://doi.org/10.1101/267500; this version posted February 18, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Integrated single-nucleotide and structural variation sig- natures of DNA-repair deficient human cancers Tyler Funnell1, Allen Zhang1, Yu-Jia Shiah2, Diljot Grewal1, Robert Lesurf2, Steven McKinney1, Ali Bashashati1, Yi Kan Wang1, Paul C. Boutros2, Sohrab P. Shah1;3 1Department of Molecular Oncology, BC Cancer Agency, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada 2Informatics & Biocomputing, Ontario Institute for Cancer Research, Toronto, Canada 3Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada Correspondence and requests for materials should be addressed to S.P.S. (email: [email protected]) Keywords: mutation signatures, structural variants, single nucleotide variants, cancer genome, ovarian cancer, breast cancer, statistical model, correlated topic models 1 bioRxiv preprint doi: https://doi.org/10.1101/267500; this version posted February 18, 2018. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Mutation signatures in cancer genomes reflect endogenous and exogenous mutational pro- cesses, offering insights into tumour etiology, features for prognostic and biologic stratifica- tion and vulnerabilities to be exploited therapeutically. We present a novel machine learning formalism for improved signature inference, based on multi-modal correlated topic models (MMCTM) which can at once infer signatures from both single nucleotide and structural variation counts derived from cancer genome sequencing data.
    [Show full text]
  • Alternative Splicing and Thermosensitive Expression of Dmrt1 During Urogenital Development in the Painted Turtle, Chrysemys Picta
    Alternative splicing and thermosensitive expression of Dmrt1 during urogenital development in the painted turtle, Chrysemys picta Beatriz Mizoguchi and Nicole Valenzuela Department of Ecology, Evolution and Organismal Biology, Iowa State University, Ames, IA, United States of America ABSTRACT Background. The doublesex and mab-3 related transcription factor 1 (Dmrt1) is a highly conserved gene across numerous vertebrates and invertebrates in sequence and function. Small aminoacid changes in Dmrt1 are associated with turnovers in sex determination in reptiles. Dmrt1 is upregulated in males during gonadal development in many species, including the painted turtle, Chrysemys picta, a reptile with temperature- dependent sex determination (TSD). Dmrt1 is reported to play different roles during sex determination and differentiation, yet whether these functions are controlled by distinct Dmrt1 spliceoforms remains unclear. While Dmrt1 isoforms have been characterized in various vertebrates, no study has investigated their existence in any turtle. Methods. We examine the painted turtle to identify novel Dmrt1 isoforms that may be present during urogenital development using PCR, profile their expression by RNA-seq across five embryonic stages at male- and female-producing temperatures, and validate their expression pattern via qPCR with transcript-specific fluorescent probes. Results. A novel Dmrt1 spliceoform was discovered for the first time in chelonians, lacking exons 2 and 3 (Dmrt1 1Ex2Ex3). Dmrt1 canonical and 1Ex2Ex3 transcripts were differentialy expressed by temperature at stages 19 and 22 in developing gonads of painted turtles, after the onset of sex determination, and displayed a significant male- Submitted 9 April 2019 biased expression pattern. This transcriptional pattern differs from studies in other Accepted 27 January 2020 turtles and vertebrates that reported Dmrt1 differential expression before or at the onset Published 19 March 2020 of sex determination.
    [Show full text]
  • Circadian Proteins and Reproduction: an Update
    Asian Journal of Biological and Life Sciences Review Article Circadian proteins and reproduction: An update Kumari Nishi1, Swarndeep Singh Hundal2*, Vinoth Prasanna Gunasekaran3 1, 2 Department of Zoology, Punjab Agricultural University, Ludhiana, Punjab-141004, India 3 Department of Biomedical Science, School of Basic Medical Sciences, Bharathidasan University, Tiruchirappalli, Tamilnadu - 620 024, India. E-mail : [email protected], [email protected], [email protected] Contact No : +91 98722 74244 Submitted : 19.02.2013 Accepted :27.04.2013 Published : 31.08.2013 Abstract Endogenous circadian rhythms play an important role in effective functioning of complex organisms to enhance their survival. The master clock or suprachiasmatic nucleus in the ventral hypothalamus helps synchronize all independent peripheral clocks for temporal regulation of all physiological activities. The important clock proteins are Circadian Locomotor Output Cycle Kaput (CLOCK), Brain-Muscle-Arnt-Like protein 1 (BMAL1), Cryptochrome (CRY) and Period (PER). These proteins have a role in reproduction and functioning of gonads. The expression of these proteins is controlled by a histone deacetylase Sirtuin1 (SIRT1). The presence, role and functioning of these compounds in the reproductive tissues is discussed. INTRODUCTION Cryptochrome(Cry1, Cry2), Period (Per1, Per2, Per3), Clock homologue (mouse) (Clock; Circadian Locomotor Output Cycle urvival of an organism depends on two factors i.e. food Kaput), and Aryl hydrocarbon Receptor Nuclear Translocator- Savailability and its reproductive ability. For effective [7-9]. reproduction, the organism should respond to the environmental like (Arntl) cues appropriately. Animals inhabiting outside the tropics, adapt These proteins include Circadian Locomotor Output Cycle to seasonal changes in environment by altering their physiology Kaput (CLOCK) and Brain-Muscle-Arnt-Like protein 1 and behavior including reproduction, migration, hibernation, (BMAL1).
    [Show full text]
  • Primary Sex Determination in Birds Depends on DMRT1 Dosage, but Gonadal Sex Does Not Determine Adult Secondary Sex Characteristics
    Primary sex determination in birds depends on DMRT1 dosage, but gonadal sex does not determine adult secondary sex characteristics Jason Ioannidisa,1,2, Gunes Taylorb,1, Debiao Zhaoa, Long Liuc, Alewo Idoko-Akoha, Daoqing Gongc, Robin Lovell-Badgeb, Silvana Guiolib,3, Mike J. McGrewa,2,3, and Michael Clintona,3 aDivision of Functional Genomics and Development, The Roslin Institute, Royal (Dick) School of Veterinary Studies, EH25 9RG Midlothian, United Kingdom; bLaboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, NW1 1AT London, United Kingdom; and cCollege of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, People’s Republic of China Edited by Scott V. Edwards, Harvard University, Cambridge, MA, and approved January 11, 2021 (received for review October 12, 2020) In birds, males are the homogametic sex (ZZ) and females the override genetic sex determination (GSD) in marsupial neonates heterogametic sex (ZW). Primary sex determination is thought to (7). In chickens, blocking E2 synthesis in female embryos leads to depend on a sex chromosome gene dosage mechanism, and the masculinization of the gonads while the addition of E2 to male most likely sex determinant is the Z chromosome gene Doublesex embryos leads to feminization of the gonads (8–10). and Mab-3–Related Transcription factor 1 (DMRT1). To clarify this In birds, the male is the homogametic sex (ZZ), and the fe- issue, we used a CRISPR-Cas9–based monoallelic targeting ap- male is the heterogametic sex (ZW), but, as yet, there is no ev- proach and sterile surrogate hosts to generate birds with targeted idence for an ovary-determining gene located on the female- mutations in the DMRT1 gene.
    [Show full text]