Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Brain ; 144(5): 1451-1466, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-33855352

RESUMEN

Abnormal gut motility is a feature of several mitochondrial encephalomyopathies, and mutations in genes such as TYMP and POLG, have been linked to these rare diseases. The human genome encodes three DNA ligases, of which only one, ligase III (LIG3), has a mitochondrial splice variant and is crucial for mitochondrial health. We investigated the effect of reduced LIG3 activity and resulting mitochondrial dysfunction in seven patients from three independent families, who showed the common occurrence of gut dysmotility and neurological manifestations reminiscent of mitochondrial neurogastrointestinal encephalomyopathy. DNA from these patients was subjected to whole exome sequencing. In all patients, compound heterozygous variants in a new disease gene, LIG3, were identified. All variants were predicted to have a damaging effect on the protein. The LIG3 gene encodes the only mitochondrial DNA (mtDNA) ligase and therefore plays a pivotal role in mtDNA repair and replication. In vitro assays in patient-derived cells showed a decrease in LIG3 protein levels and ligase activity. We demonstrated that the LIG3 gene defects affect mtDNA maintenance, leading to mtDNA depletion without the accumulation of multiple deletions as observed in other mitochondrial disorders. This mitochondrial dysfunction is likely to cause the phenotypes observed in these patients. The most prominent and consistent clinical signs were severe gut dysmotility and neurological abnormalities, including leukoencephalopathy, epilepsy, migraine, stroke-like episodes, and neurogenic bladder. A decrease in the number of myenteric neurons, and increased fibrosis and elastin levels were the most prominent changes in the gut. Cytochrome c oxidase (COX) deficient fibres in skeletal muscle were also observed. Disruption of lig3 in zebrafish reproduced the brain alterations and impaired gut transit in vivo. In conclusion, we identified variants in the LIG3 gene that result in a mitochondrial disease characterized by predominant gut dysmotility, encephalopathy, and neuromuscular abnormalities.


Asunto(s)
ADN Ligasa (ATP)/genética , Enfermedades Gastrointestinales/genética , Motilidad Gastrointestinal/genética , Encefalomiopatías Mitocondriales/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Animales , Femenino , Enfermedades Gastrointestinales/patología , Humanos , Masculino , Encefalomiopatías Mitocondriales/patología , Mutación , Linaje , Pez Cebra
2.
Cytogenet Genome Res ; 160(3): 118-123, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32248198

RESUMEN

We present 2 cases of double mosaic aneuploidy harboring 2 or more different aneuploid cell lines, but no line with a normal chromosome constitution. One of these cases presented mosaicism of sex chromosome aneuploid cell lines (47,XXX/45,X) along with another line containing an autosomal trisomy (47,XX,+8), while the other case showed mosaicism of 2 different autosomal trisomy cell lines (47,XY,+5 and 47,XY,+8). To elucidate the mechanisms underlying these mosaicisms, we conducted molecular cytogenetic analyses. Genotyping data from the SNP microarray indicated that 2 sequential meiotic or early postzygotic segregation errors likely had occurred followed by natural selection. These cases suggest that frequent segregation errors and selection events in the meiotic and early postzygotic stages lead to this condition.


Asunto(s)
Linaje de la Célula/genética , Mosaicismo , Cromosomas Sexuales/genética , Trisomía/genética , Aneuploidia , Análisis Citogenético , Femenino , Humanos , Lactante , Persona de Mediana Edad , Trisomía/patología
3.
J Hum Genet ; 65(8): 705-709, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32277176

RESUMEN

Sex-chromosome discordant chimerism (XX/XY chimerism) is a rare chromosomal disorder in humans. We report a boy with ambiguous genitalia and hypospadias, showing 46,XY[26]/46,XX[4] in peripheral blood cells. To clarify the mechanism of how this chimerism took place, we carried out whole-genome genotyping using a SNP array and microsatellite analysis. The B-allele frequency of the SNP array showed a mixture of three and five allele combinations, which excluded mosaicism but not chimerism, and suggested the fusion of two embryos or a shared parental haplotype between the two parental cells. All microsatellite markers showed a single maternal allele. From these results, we concluded that this XX/XY chimera is composed of two different paternal alleles and a single duplicated maternal genome. This XX/XY chimera likely arose from a diploid maternal cell that was formed via endoduplication of the maternal genome just before fertilization, being fertilized with both X and Y sperm.


Asunto(s)
Quimera/genética , Quimerismo , Trastornos del Desarrollo Sexual/genética , Partenogénesis/genética , Trastornos de los Cromosomas Sexuales/genética , Alelos , Trastornos del Desarrollo Sexual/diagnóstico por imagen , Haplotipos , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Masculino , Repeticiones de Microsatélite/genética , Mosaicismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple , Aberraciones Cromosómicas Sexuales , Trastornos de los Cromosomas Sexuales/sangre , Trastornos de los Cromosomas Sexuales/diagnóstico por imagen
4.
BMC Cancer ; 20(1): 1162, 2020 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-33246418

RESUMEN

BACKGROUND: Aggressive systemic mastocytosis (ASM) is a rare malignant disease characterized by disordered mast cell accumulation in various organs. We here describe a female ASM patient with a previous history of ovarian dysgerminoma. METHODS: Molecular cytogenomic analyses were performed to elucidate an etiological link between the ASM and dysgerminoma of the patient. RESULTS: This patient was affected by ovarian dysgerminoma which was treated by chemotherapy and surgical resection. Having subsequently been in complete remission for 2 years, she developed symptoms of ASM. A somatic D816A mutation in the KIT gene was detected in her bone marrow, which facilitated the diagnosis of ASM. Unexpectedly, this KIT D816A variant was also detected in the prior ovarian dysgerminoma sample. Whole-exome sequencing allowed us to identify a somatic nonsense mutation of the TP53 gene in the bone marrow, but not in the dysgerminoma. Microarray analysis of the patient's bone marrow revealed a copy-number-neutral loss of heterozygosity at the TP53 locus, suggestive of the homozygous nonsense mutation in the TP53 gene. In addition, the loss of heterozygosity at the TP53 locus was also detected in the dysgerminoma. CONCLUSIONS: These results indicated that either the mast cells causing the ASM in this case had originated from the preceding ovarian dysgerminoma as a clonal evolution of a residual tumor cell, which acquired the TP53 mutation, or that both tumors developed from a common cancer stem cell carrying the KIT D816A variation.


Asunto(s)
Disgerminoma/complicaciones , Mastocitosis Sistémica/etiología , Neoplasias de Células Germinales y Embrionarias/complicaciones , Neoplasias Ováricas/complicaciones , Disgerminoma/patología , Femenino , Humanos , Mastocitosis Sistémica/patología , Persona de Mediana Edad , Neoplasias de Células Germinales y Embrionarias/patología , Neoplasias Ováricas/patología
5.
BMC Med Genet ; 19(1): 210, 2018 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-30541480

RESUMEN

BACKGROUND: Ornithine transcarbamylase deficiency (OTCD) is an X-linked recessive disorder involving a defect in the urea cycle caused by OTC gene mutations. Although a total of 417 disease-causing mutations in OTC have been reported, structural abnormalities in this gene are rare. We here describe a female OTCD case caused by an exonic duplication of the OTC gene (exons 1-6). CASE PRESENTATION: A 23-year-old woman with late-onset OTCD diagnosed by biochemical testing was subjected to subsequent genetic testing. Sanger sequencing revealed no pathogenic mutation throughout the coding exons of the OTC gene, but multiplex ligation-dependent probe amplification (MLPA) revealed duplication of exons 1-6. Further genetic analyses revealed an inversion of duplicated exon 1 and a tandem duplication of exons 2-6. Each of the junctions of the inversion harbored a microhomology and non-templated microinsertion, respectively, suggesting a replication-based mechanism. The duplication was also of de novo origin but segregation analysis indicated that it took place in the paternal chromosome. CONCLUSION: We report the first OTCD case harboring an exonic duplication in the OTC gene. The functional defects caused by this anomaly were determined via structural analysis of its complex rearrangements.


Asunto(s)
Cromosomas Humanos X/química , Exones , Duplicación de Gen , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Ornitina Carbamoiltransferasa/genética , Edad de Inicio , Secuencia de Bases , Femenino , Expresión Génica , Genes Recesivos , Humanos , Reacción en Cadena de la Polimerasa Multiplex , Ornitina Carbamoiltransferasa/metabolismo , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/metabolismo , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/fisiopatología , Herencia Paterna , Translocación Genética , Adulto Joven
6.
BMC Med Genet ; 19(1): 166, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30217189

RESUMEN

BACKGROUND: Nectins are cell adhesion molecules that play a pivotal role in adherens junctions and tight junctions. Our previous study using whole-genome oligonucleotide microarrays revealed that nectin-4 was upregulated in pre-eclamptic placentas. We investigated the role of nectin-4 in the etiology of pre-eclampsia. METHODS: We investigated the expression of nectin-4 using real-time RT-PCR, western blot and immunostaining. Additionally, we performed matrigel invasion assay and cytotoxicity assay using cells overexpressing the nectin-4. RESULTS: NECTIN4 transcripts were elevated in pre-eclamptic placentas relative to uncomplicated pregnancies. Nectin-4 protein levels in pre-eclamptic placentas were higher on a semi-quantitative western blot. Nectin-4 was localized at the apical cell membrane in syncytiotrophoblast cells and not at the adherens junctions. Nectin-4 was also detected in cytotrophoblasts and a subset of cells in the decidua. Nectin-4 overexpressing trophoblast cells migrated normally in the matrix. However, Natural killer (NK) cells showed a strong cytotoxic effect against nectin-4 overexpressing trophoblast cells. No causative genetic variation was evident in the NECTIN4 gene from a pre-eclamptic placenta. CONCLUSIONS: There are as yet unknown factors that induce nectin-4 overexpression in trophoblast cells that may contribute to abnormal placentation via an aberrant immune response and the onset of a pre-eclamptic pregnancy.


Asunto(s)
Moléculas de Adhesión Celular/genética , Decidua/inmunología , Preeclampsia/genética , ARN Mensajero/genética , Trofoblastos/inmunología , Adulto , Estudios de Casos y Controles , Moléculas de Adhesión Celular/inmunología , Cesárea , Citotoxicidad Inmunológica , Decidua/patología , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Preeclampsia/inmunología , Preeclampsia/patología , Preeclampsia/cirugía , Embarazo , ARN Mensajero/inmunología , Trofoblastos/patología
7.
Pediatr Int ; 56(4): 462-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24980921

RESUMEN

Constitutional t(11;22)(q23;q11) is the most frequent recurrent non-Robertsonian translocation in humans. Balanced carriers of t(11;22) usually manifest no clinical symptoms, and are often identified after the birth of offspring with an unbalanced form of this translocation, known as Emanuel syndrome. To determine the prevalence of the disorder, we sent surveillance questionnaires to 735 core hospitals in Japan. The observed number of Emanuel syndrome cases was 36 and that of t(11;22) balanced translocation carriers, 40. On the basis of the de novo t(11;22) translocation frequency in sperm from healthy men, we calculated the frequency of the translocations in the general population. Accordingly, the prevalence of Emanuel syndrome was estimated at 1 in 110,000. Based on this calculation, the estimated number of Emanuel syndrome cases in Japan is 1063 and of t(11;22) balanced translocation carriers, 16,604, which are much higher than the numbers calculated from the questionnaire responses. It is possible that this discordance is partly attributable to a lack of disease identification. Further efforts should be made to increase the awareness of Emanuel syndrome to ensure a better quality of life for affected patients and their families.


Asunto(s)
Trastornos de los Cromosomas/epidemiología , Fisura del Paladar/epidemiología , Cardiopatías Congénitas/epidemiología , Discapacidad Intelectual/epidemiología , Hipotonía Muscular/epidemiología , Biología Computacional , Monitoreo Epidemiológico , Femenino , Humanos , Japón/epidemiología , Masculino , Prevalencia , Encuestas y Cuestionarios
8.
Genes Cells ; 17(11): 897-912, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23039116

RESUMEN

Meiotic chromosome segregation requires homologous pairing, synapsis and crossover recombination during meiotic prophase. The checkpoint kinase ATR has been proposed to be involved in the quality surveillance of these processes, although the underlying mechanisms remain largely unknown. In our present study, we generated mice lacking HORMAD2, a protein that localizes to unsynapsed meiotic chromosomes. We show that this Hormad2 deficiency hampers the proper recruitment of ATR activity to unsynapsed chromosomes. Male Hormad2-deficient mice are infertile due to spermatocyte loss as a result of characteristic impairment of sex body formation; an ATR- and γH2AX-enriched repressive chromatin domain is formed, but is partially dissociated from the elongated sex chromosome axes. In contrast to males, Hormad2-deficient females are fertile. However, our analysis of Hormad2/Spo11 double-mutant females shows that the oocyte number is negatively correlated with the frequency of pseudo-sex body formation in a Hormad2 gene dosage-dependent manner. This result suggests that the elimination of Spo11-deficient asynaptic oocytes is associated with the HORMAD2-dependent pseudo-sex body formation that is likely initiated by local concentration of ATR activity in the absence of double-strand breaks. Our results thus show a HORMAD2-dependent quality control mechanism that recognizes unsynapsis and recruits ATR activity during mammalian meiosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Emparejamiento Cromosómico , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular/genética , Roturas del ADN de Doble Cadena , Endodesoxirribonucleasas/deficiencia , Endodesoxirribonucleasas/genética , Femenino , Fertilidad/genética , Silenciador del Gen , Masculino , Profase Meiótica I , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Oocitos/metabolismo , Oocitos/fisiología , Ovario/citología , Ovario/enzimología , Transporte de Proteínas , Espermatocitos/metabolismo , Espermatocitos/fisiología , Testículo/citología
9.
Genes Cells ; 17(6): 439-54, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22530760

RESUMEN

Meiotic pachytene checkpoints monitor the failure of homologous recombination and synapsis to ensure faithful chromosome segregation during gamete formation. To date, the molecular basis of the mammalian pachytene checkpoints has remained largely unknown. We here report that mouse HORMAD1 is required for a meiotic prophase checkpoint that eliminates asynaptic oocytes. Hormad1-deficient mice are infertile and show an extensive failure of homologous pairing and synapsis, consistent with the evolutionarily conserved function of meiotic HORMA domain proteins. Unexpectedly, Hormad1-deficient ovaries contain a normal number of oocytes despite asynapsis and consequently produce aneuploid oocytes, indicating a checkpoint failure. By the analysis of Hormad1/Spo11 double mutants, the Hormad1 deficiency was found to abrogate the massive oocyte loss in the Spo11-deficient ovary. The Hormad1 deficiency also causes the eventual loss of pseudo sex body in the Spo11-deficient ovary and testis. These results suggest the involvement of HORMAD1 in the repressive chromatin domain formation that is proposed to be important in the meiotic prophase checkpoints. We also show the extensive phosphorylation of HORMAD1 in the Spo11-deficient testis and ovary, suggesting an involvement of novel DNA damage-independent phosphorylation signaling in the surveillance mechanism. Our present results provide clues to HORMAD1-dependent checkpoint in response to asynapsis in mammalian meiosis.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Emparejamiento Cromosómico , Meiosis , Animales , Proteínas de Ciclo Celular/genética , Endodesoxirribonucleasas/genética , Endodesoxirribonucleasas/metabolismo , Femenino , Genes cdc , Masculino , Ratones , Ratones Noqueados , Oocitos/citología , Oocitos/metabolismo , Fosforilación , Espermatocitos/citología , Espermatocitos/metabolismo
10.
Hum Mol Genet ; 19(13): 2630-7, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20392709

RESUMEN

The constitutional t(11;22) is the most frequent recurrent non-Robertsonian translocation in humans, the breakpoints of which are located within palindromic AT-rich repeats on 11q23 and 22q11 (PATRR11 and PATRR22). Genetic variation of the PATRR11 was found to affect de novo t(11;22) translocation frequency in sperm derived from normal healthy males, suggesting the hypothesis that polymorphisms of the PATRR22 might also influence the translocation frequency. Although the complicated structure of the PATRR22 locus prevented determining the genotype of the PATRR22 in each individual, genotyping of flanking markers as well as identification of rare variants allowed us to demonstrate an association between the PATRR22 allele type and the translocation frequency. We found that size and symmetry of the PATRR22 affect the de novo translocation frequency, which is lower for the shorter or more asymmetric versions. These data lend support to our hypothesis that the PATRRs form secondary structures in the nucleus that induce genomic instability leading to the recurrent translocation.


Asunto(s)
Cromosomas Humanos Par 11/genética , Cromosomas Humanos Par 22/genética , Espermatozoides , Translocación Genética/genética , Secuencia Rica en At/genética , Alelos , Rotura Cromosómica , ADN/química , ADN/genética , Variación Genética , Humanos , Masculino , Datos de Secuencia Molecular , Polimorfismo Genético , Análisis de Secuencia de ADN
11.
Am J Hum Genet ; 84(1): 14-20, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19110213

RESUMEN

Aneuploidy, a chromosomal numerical abnormality in the conceptus or fetus, occurs in at least 5% of all pregnancies and is the leading cause of early pregnancy loss in humans. Accumulating evidence now suggests that the correct segregation of chromosomes is affected by events occurring in prophase during meiosis I. These events include homologous chromosome pairing, sister-chromatid cohesion, and meiotic recombination. In our current study, we show that mutations in SYCP3, a gene encoding an essential component of the synaptonemal complex that is central to the interaction of homologous chromosomes, are associated with recurrent pregnancy loss. Two out of 26 women with recurrent pregnancy loss of unknown cause were found to carry independent heterozygous nucleotide alterations in this gene, neither of which was present among a group of 150 fertile women. Analysis of transcripts from minigenes harboring each of these two mutations revealed that both affected normal splicing, possibly resulting in the production of C-terminally mutated proteins. The mutant proteins were found to interact with their wild-type counterpart in vitro and inhibit the normal fiber formation of the SYCP3 protein when coexpressed in a heterologous system. These data suggest that these mutations are likely to generate an aberrant synaptonemal complex in a dominant-negative manner and contribute to abnormal chromosomal behavior that might lead to recurrent miscarriage. Combined with the fact that similar mutations have been previously identified in two males with azoospermia, our current data suggest that sexual dimorphism in response to meiotic disruption occurs even in humans.


Asunto(s)
Aborto Habitual/genética , Predisposición Genética a la Enfermedad , Proteínas Nucleares/genética , Adulto , Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Femenino , Humanos , Mutación , Embarazo , Complejo Sinaptonémico/genética
12.
J Cell Sci ; 123(Pt 3): 484-96, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20067997

RESUMEN

During the development of the vertebrate nervous system, mitosis of neural progenitor cells takes place near the lumen, the apical side of the neural tube, through a characteristic movement of nuclei known as interkinetic nuclear migration (INM). Furthermore, during the proliferative period, neural progenitor cells exhibit planar cell divisions to produce equivalent daughter cells. Here, we examine the potential role of extracellular signals in INM and planar divisions using the medaka mutant tacobo (tab). This tab mutant shows pleiotropic phenotypes, including neurogenesis, and positional cloning identified tab as laminin gamma1 (lamc1), providing a unique framework to study the role of extracellular signals in neurogenesis. In tab mutant neural tubes, a number of nuclei exhibit abnormal patterns of migration leading to basally mislocalized mitosis. Furthermore, the orientation of cell division near the apical surface is randomized. Probably because of these defects, neurogenesis is accelerated in the tab neural tube. Detailed analyses demonstrate that extracellular signals mediated by the FAK pathway regulate INM and planar divisions in the neuroepithelium, possibly through interaction with the intracellular dynein-motor system.


Asunto(s)
Núcleo Celular/metabolismo , Proteínas de Peces/metabolismo , Células Neuroepiteliales/metabolismo , Transducción de Señal/fisiología , Animales , Immunoblotting , Inmunohistoquímica , Microscopía Confocal , Células Neuroepiteliales/citología , Oryzias
13.
Genes Cells ; 16(11): 1097-109, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22023386

RESUMEN

Although recent findings showed that some Drosophila doublesex and Caenorhabditis elegans mab-3 related genes are expressed in neural tissues during development, their functions have not been fully elucidated. Here, we isolated a zebrafish mutant, ha2, that shows defects in telencephalic neurogenesis and found that ha2 encodes Doublesex and MAB-3 related transcription factor like family A2 (Dmrta2). dmrta2 expression is restricted to the telencephalon, diencephalon and olfactory placode during somitogenesis. We found that the expression of the proneural gene, neurogenin1, in the posterior and dorsal region of telencephalon (posterior-dorsal telencephalon) is markedly reduced in this mutant at the 14-somite stage without any defects in cell proliferation or cell death. In contrast, the telencephalic expression of her6, a Hes-related gene that is known to encode a negative regulator of neurogenin1, expands dramatically in the ha2 mutant. Based on over-expression experiments and epistatic analyses, we propose that zebrafish Dmrta2 controls neurogenin1 expression by repressing her6 in the posterior-dorsal telencephalon. Furthermore, the expression domains of the telencephalic marker genes, foxg1 and emx3, and the neuronal differentiation gene, neurod, are downregulated in the ha2 posterior-dorsal telencephalon during somitogenesis. These results suggest that Dmrta2 plays important roles in the specification of the posterior-dorsal telencephalic cell fate during somitogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Telencéfalo/embriología , Factores de Transcripción/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Animales , Proteínas de Unión al ADN/genética , Epistasis Genética , Etilnitrosourea/química , Mutagénesis , Mutación , Telencéfalo/metabolismo , Factores de Transcripción/genética , Pez Cebra/genética
14.
J Hum Genet ; 57(8): 515-22, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22648182

RESUMEN

The events that take place during the prophase of meiosis I are essential for the correct segregation of homologous chromosomes. Defects in these processes likely contribute to infertility or recurrent pregnancy loss in humans. To screen for candidate genes for reproductive failure due to meiotic defects, we have analyzed the gene expression patterns in fetal, neonatal and adult gonads of both male and female mice by microarray and thereby identified 241 genes that are expressed specifically during prophase of meiosis I. Combined with our previous data obtained from developing spermatocytes, a total of 99 genes were identified that are upregulated in early prophase I. We confirmed the meiotic prophase I-specific expression of these genes using qRT-PCR. To further screen this panel for candidate genes that fulfill important roles in homologous pairing, synapsis and recombination, we established a gene transfer system for prophase I oocytes in combination with in vitro organ culture of ovaries, and successfully determined the localization of the selected genes. This gene set can thus serve as a resource for targeted sequence analysis via next-generation sequencing to identify the genes associated with human reproduction failure due to meiotic defects.


Asunto(s)
Segregación Cromosómica/genética , Gónadas , Profase Meiótica I/genética , Oocitos , Animales , Emparejamiento Cromosómico/genética , Desarrollo Embrionario , Femenino , Perfilación de la Expresión Génica , Gónadas/citología , Gónadas/metabolismo , Masculino , Ratones , Oocitos/citología , Oocitos/metabolismo , Embarazo , Recombinación Genética , Espermatocitos/citología , Espermatocitos/metabolismo
15.
JIMD Rep ; 63(6): 575-580, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36341163

RESUMEN

Maple syrup urine disease (MSUD) is a rare autosomal recessive inherited disorder of branched-chain amino acid metabolism caused by mutations in BCKDHA, BCKDHB, and DBT that encode the E1α, E1ß, and E2 subunits of the branched-chain α-ketoacid dehydrogenase (BCKD) complex. Various MSUD-causing variants have been described; however, no structural rearrangements in BCKDHA have been reported to cause the classic MSUD phenotype. Here, we describe the classic patient with MSUD with compound heterozygous pathogenic variants in BCKDHA: a missense variant (NM_000709.3:c.757G > A, NP_000700.1:p.Ala253Thr) and a paracentric inversion disrupting Intron 1 of BCKDHA, which was identified by whole-genome sequencing and validated by fluorescence in situ hybridization. Using the sequence information of the breakpoint junction, we gained mechanistic insight into the development of this structural rearrangement. Furthermore, the establishment of junction-specific polymerase chain reaction could facilitate identification of the variant in case carrier or future prenatal/preimplantation tests are necessary.

16.
Hum Mol Genet ; 18(18): 3397-406, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19520744

RESUMEN

Palindromic regions are unstable and susceptible to deletion in prokaryotes and eukaryotes possibly due to stalled or slow replication. In the human genome, they also appear to become partially or completely deleted, while two palindromic AT-rich repeats (PATRR) contribute to known recurrent constitutional translocations. To explore the mechanism that causes the development of palindrome instabilities in humans, we compared the incidence of de novo translocations and deletions at PATRRs in human cells. Using a highly sensitive PCR assay that can detect single molecules, de novo deletions were detected neither in human somatic cells nor in sperm. However, deletions were detected at low frequency in cultured cell lines. Inhibition of DNA replication by administration of siRNA against the DNA polymerase alpha 1 (POLA1) gene or introduction of POLA inhibitors increased the frequency. This is in contrast to PATRR-mediated translocations that were never detected in similar conditions but were observed frequently in human sperm samples. Further deletions were found to take place during both leading- and lagging-strand synthesis. Our data suggest that stalled or slow replication induces deletions within PATRRs, but that other mechanisms might contribute to PATRR-mediated recurrent translocations in humans.


Asunto(s)
Replicación del ADN , Eliminación de Gen , Secuencias Invertidas Repetidas , Línea Celular , ADN/genética , ADN/metabolismo , ADN Polimerasa I/genética , ADN Polimerasa I/metabolismo , Regulación de la Expresión Génica , Humanos , ARN Interferente Pequeño/genética , Translocación Genética
17.
Biol Reprod ; 85(1): 165-71, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21451147

RESUMEN

Xlr6 is a novel but uncharacterized X-linked gene that is upregulated in meiotic prophase I during mouse spermatogenesis. Xlr6 belongs to the Xlr gene family, which includes a component of the axial/lateral element of the synaptonemal complex, Sycp3, and its transcripts are abundant in the fetal ovary and adult testis. Immunostaining and Western blot analysis demonstrate a diffuse localization pattern for this protein in the nucleus and an association with chromatin during the leptotene and zygotene stages. In males, XLR6 accumulates at the XY body of early pachytene to midpachytene spermatocytes, although the Xlr6 gene is subjected to meiotic sex chromosome inactivation. During the late pachytene and diplotene stages, the XLR6 protein relocalizes from the XY body to the nucleolus and, eventually, disappears by diakinesis. In females, XLR6 disappears at the pachytene stage, whereas it accumulates at the unpaired chromosomes occasionally observed in wild-type female mice. Although the amino acid sequence of XLR6 has a high similarity with SYCP3, its distinct localization pattern and dynamism suggest a unique chromatin modification function that leads to the transcriptional repression of ribosomal DNA in addition to sex chromosome genes.


Asunto(s)
Nucléolo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Profase Meiótica I , Espermatocitos/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN , Femenino , Genes Ligados a X , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Nucleares , ARN Mensajero/metabolismo , Homología de Secuencia de Aminoácido , Espermatogénesis , Regulación hacia Arriba
18.
J Hum Genet ; 55(5): 293-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20339383

RESUMEN

Prophase I of male meiosis during early spermatogenesis involves dynamic chromosome segregation processes, including synapsis, meiotic recombination and cohesion. Genetic defects in the genes that participate in these processes consistently cause reproduction failure in mice. To identify candidate genes responsible for infertility in humans, we performed gene expression profiling of mouse spermatogenic cells undergoing meiotic prophase I. Cell fractions enriched in spermatogonia, leptotene/zygotene spermatocytes or pachytene spermatocytes from developing mouse testis were separately isolated by density gradient sedimentation and subjected to microarray analysis. A total of 726 genes were identified that were upregulated in leptotene/zygotene spermatocytes. To evaluate the screening efficiency for meiosis-specific genes, we randomly selected 12 genes from this gene set and characterized each gene product using reverse transcription (RT)-PCR of RNA from gonadal tissues, in situ hybridization on testicular tissue sections and subcellular localization analysis of the encoded protein. Four of the 12 genes were confirmed as genes expressed in meiotic stage and 2 of these 4 genes were novel, previously uncharacterized genes. Among the three confirmation methods that were used, RT-PCR appeared to be the most efficient method for further screening. These 726 candidates for human infertility genes might serve as a useful resource for next-generation sequencing combined with exon capture by microarray.


Asunto(s)
Segregación Cromosómica/genética , Predisposición Genética a la Enfermedad/genética , Infertilidad Masculina/genética , Animales , Línea Celular , Endodesoxirribonucleasas , Esterasas/genética , Esterasas/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Hibridación in Situ , Masculino , Profase Meiótica I/genética , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatocitos/citología , Espermatocitos/metabolismo , Testículo/citología , Testículo/metabolismo , Transfección
19.
Mol Genet Genomic Med ; 8(12): e1531, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33085210

RESUMEN

BACKGROUND: Incontinentia pigmenti (IP) is a rare X-linked disorder affecting the skin and other ectodermal tissues that is caused by mutation of the IKBKG/NEMO gene. Previous studies have reported that the overall mutation detection rate in IP is ~75%. We hypothesized that a low-level mosaicism existed in the remaining cases. METHODS: Genomic variations in the IKBKG gene were examined in 30 IP probands and their family members. Standard mutational analyses were performed to detect common deletions, nucleotide alterations, and copy number variations. To assess skewing of the X chromosome inactivation (XCI) pattern, a HUMARA assay was performed. We compared the results of this analysis with phenotype severity. RESULTS: Pathogenic variants were identified in 20 probands (66.7%), the rate of detection was suboptimal. The remaining 10 probands tended to manifest a mild phenotype with no skewed X chromosome inactivation that is generally observed in IP patients. Quantitative nested PCR and digital droplet PCR were performed for the 10 patients and mosaicism of the common IKBKG deletion were identified in five patients. CONCLUSION: Overall, we detected 25 IKBKG mutations (83.3%). Determination of the XCI value in advance of mutational analyses for IP could improve the mutation detection rate. Our improved detection rate for these mutations, particularly those with a low-level mosaicism, may present opportunities for appropriate genetic counseling.


Asunto(s)
Quinasa I-kappa B/genética , Incontinencia Pigmentaria/genética , Mosaicismo , Mutación , Adulto , Preescolar , Femenino , Pruebas Genéticas/métodos , Humanos , Lactante , Masculino , Persona de Mediana Edad , Inactivación del Cromosoma X
20.
J Hum Genet ; 54(5): 253-60, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19373258

RESUMEN

The completion of the human genome project has enabled researchers to characterize the breakpoints for various chromosomal structural abnormalities including deletions, duplications or translocations. This in turn has shed new light on the molecular mechanisms underlying the onset of gross chromosomal rearrangements. On the other hand, advances in genetic manipulation technologies for various model organisms has increased our knowledge of meiotic chromosome segregation, errors which, contribute to chromosomal aneuploidy. This review focuses on the current understanding of germ line chromosomal abnormalities and provides an overview of the mechanisms involved. We refer to our own recent data and those of others to illustrate some of the new paradigms that have arisen in this field. We also discuss some perspectives on the sexual dimorphism of some of the pathways that leads to these chromosomal abnormalities.


Asunto(s)
Aberraciones Cromosómicas , Duplicación de Gen , Reordenamiento Génico/genética , Predisposición Genética a la Enfermedad , Humanos , Meiosis , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA