Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
JIMD Rep ; 63(6): 575-580, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36341163

ABSTRACT

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.

2.
Brain ; 144(5): 1451-1466, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33855352

ABSTRACT

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.


Subject(s)
DNA Ligase ATP/genetics , Gastrointestinal Diseases/genetics , Gastrointestinal Motility/genetics , Mitochondrial Encephalomyopathies/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Animals , Female , Gastrointestinal Diseases/pathology , Humans , Male , Mitochondrial Encephalomyopathies/pathology , Mutation , Pedigree , Zebrafish
3.
BMC Cancer ; 20(1): 1162, 2020 Nov 27.
Article in English | MEDLINE | ID: mdl-33246418

ABSTRACT

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.


Subject(s)
Dysgerminoma/complications , Mastocytosis, Systemic/etiology , Neoplasms, Germ Cell and Embryonal/complications , Ovarian Neoplasms/complications , Dysgerminoma/pathology , Female , Humans , Mastocytosis, Systemic/pathology , Middle Aged , Neoplasms, Germ Cell and Embryonal/pathology , Ovarian Neoplasms/pathology
4.
Mol Genet Genomic Med ; 8(12): e1531, 2020 12.
Article in English | MEDLINE | ID: mdl-33085210

ABSTRACT

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.


Subject(s)
I-kappa B Kinase/genetics , Incontinentia Pigmenti/genetics , Mosaicism , Mutation , Adult , Child, Preschool , Female , Genetic Testing/methods , Humans , Infant , Male , Middle Aged , X Chromosome Inactivation
5.
J Hum Genet ; 65(8): 705-709, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32277176

ABSTRACT

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.


Subject(s)
Chimera/genetics , Chimerism , Disorders of Sex Development/genetics , Parthenogenesis/genetics , Sex Chromosome Disorders/genetics , Alleles , Disorders of Sex Development/diagnostic imaging , Haplotypes , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Microsatellite Repeats/genetics , Mosaicism , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Sex Chromosome Aberrations , Sex Chromosome Disorders/blood , Sex Chromosome Disorders/diagnostic imaging
6.
Cytogenet Genome Res ; 160(3): 118-123, 2020.
Article in English | MEDLINE | ID: mdl-32248198

ABSTRACT

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.


Subject(s)
Cell Lineage/genetics , Mosaicism , Sex Chromosomes/genetics , Trisomy/genetics , Aneuploidy , Cytogenetic Analysis , Female , Humans , Infant , Middle Aged , Trisomy/pathology
7.
BMC Med Genomics ; 12(1): 182, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31806026

ABSTRACT

BACKGROUND: Female carriers of a balanced X; autosome translocation generally undergo selective inactivation of the normal X chromosome. This is because inactivation of critical genes within the autosomal region of the derivative translocation chromosome would compromise cellular function. We here report a female patient with bilateral retinoblastoma and a severe intellectual disability who carries a reciprocal X-autosomal translocation. CASE PRESENTATION: Cytogenetic and molecular analyses, a HUMARA (Human androgen receptor) assay, and methylation specific PCR (MSP) and bisulfite sequencing were performed using peripheral blood samples from the patient. The patient's karyotype was 46,X,t(X;13)(q28;q14.1) by G-banding analysis. Further cytogenetic analysis located the entire RB1 gene and its regulatory region on der(X) with no translocation disruption. The X-inactivation pattern in the peripheral blood was highly skewed but not completely selected. MSP and deep sequencing of bisulfite-treated DNA revealed that an extensive 13q region, including the RB1 promoter, was unusually methylated in a subset of cells. CONCLUSIONS: The der(X) region harboring the RB1 gene was inactivated in a subset of somatic cells, including the retinal cells, in the patient subject which acted as the first hit in the development of her retinoblastoma. In addition, the patient's intellectual disability may be attributable to the inactivation of the der(X), leading to a 13q deletion syndrome-like phenotype, or to an active X-linked gene on der (13) leading to Xq28 functional disomy.


Subject(s)
Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, X/genetics , Intellectual Disability/complications , Intellectual Disability/genetics , Retinoblastoma Binding Proteins/genetics , Retinoblastoma/complications , Translocation, Genetic , Ubiquitin-Protein Ligases/genetics , Female , Humans , Infant , Retinal Neoplasms/complications
9.
Int J Cardiol ; 274: 290-295, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30228022

ABSTRACT

BACKGROUND: Supravalvular aortic stenosis (SVAS) is a congenital heart disease affecting approximately 1:25,000 live births. SVAS may occur sporadically, be inherited in an autosomal dominant manner, or be associated with Williams-Beuren syndrome, a complex developmental disorder caused by a microdeletion of chromosome 7q11.23. ELN on 7q11.23, which encodes elastin, is the only known gene to be recurrently mutated in less than half of SVAS patients. METHODS: Whole-exome sequencing (WES) was performed for seven familial SVAS families to identify other causative gene mutations of SVAS. RESULTS: Three truncating mutations and three intragenic deletions affecting ELN were identified, yielding a diagnostic efficiency of 6/7 (85%). The deletions, which explained 3/7 of the present cohort, spanned 1-29 exons, which might be missed in the course of mutational analysis targeting point mutations. The presence of such deletions was validated by both WES-based copy number estimation and multiplex ligation-dependent probe amplification analyses, and their pathogenicity was reinforced by co-segregation with clinical presentations. CONCLUSIONS: The majority of familial SVAS patients appear to carry ELN mutations, which strongly indicates that elastin is the most important causative gene for SVAS. The frequency of intragenic deletions highlights the need for quantitative tests to analyze ELN for efficient genetic diagnosis of SVAS.


Subject(s)
Aortic Stenosis, Supravalvular/genetics , DNA/genetics , Elastin/genetics , Point Mutation , Aortic Stenosis, Supravalvular/diagnosis , Aortic Stenosis, Supravalvular/metabolism , DNA Mutational Analysis , Elastin/metabolism , Female , Humans , Male , Pedigree , Exome Sequencing/methods
10.
Eur J Med Genet ; 62(3): 224-228, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30031150

ABSTRACT

Jacobsen syndrome refers to a congenital anomaly caused by deletion at 11q23.3-qter. We here describe two siblings with the same 11q23.3-qter deletion. Both parents were healthy with a normal karyotype. Cytogenetic microarray analysis revealed no mosaicism in either parent but the mother showed uniparental disomy encompassing the deleted region found in the two siblings. The pattern of X chromosome inactivation was almost completely skewed in the mother. These data suggested that the mother was a carrier of the 11q23.3-qter deletion but that this had been rescued by disomy formation during early embryogenesis except for her germinal cells.


Subject(s)
Jacobsen Distal 11q Deletion Syndrome/genetics , Phenotype , Uniparental Disomy/genetics , Child, Preschool , Humans , Jacobsen Distal 11q Deletion Syndrome/pathology , Karyotype , Male , Pedigree , Siblings , Uniparental Disomy/pathology , X Chromosome Inactivation
11.
BMC Med Genet ; 19(1): 210, 2018 12 12.
Article in English | MEDLINE | ID: mdl-30541480

ABSTRACT

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.


Subject(s)
Chromosomes, Human, X/chemistry , Exons , Gene Duplication , Ornithine Carbamoyltransferase Deficiency Disease/genetics , Ornithine Carbamoyltransferase/genetics , Age of Onset , Base Sequence , Female , Gene Expression , Genes, Recessive , Humans , Multiplex Polymerase Chain Reaction , Ornithine Carbamoyltransferase/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/metabolism , Ornithine Carbamoyltransferase Deficiency Disease/physiopathology , Paternal Inheritance , Translocation, Genetic , Young Adult
12.
BMC Med Genet ; 19(1): 166, 2018 09 14.
Article in English | MEDLINE | ID: mdl-30217189

ABSTRACT

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.


Subject(s)
Cell Adhesion Molecules/genetics , Decidua/immunology , Pre-Eclampsia/genetics , RNA, Messenger/genetics , Trophoblasts/immunology , Adult , Case-Control Studies , Cell Adhesion Molecules/immunology , Cesarean Section , Cytotoxicity, Immunologic , Decidua/pathology , Female , Gene Expression Regulation , Humans , Immunity, Innate , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Pre-Eclampsia/immunology , Pre-Eclampsia/pathology , Pre-Eclampsia/surgery , Pregnancy , RNA, Messenger/immunology , Trophoblasts/pathology
14.
Eur J Hum Genet ; 24(12): 1702-1706, 2016 12.
Article in English | MEDLINE | ID: mdl-27650967

ABSTRACT

It has been well documented that variants in genes encoding centrosomal proteins cause primary autosomal recessive microcephaly, although the association between centrosomal defects and the etiology of microcephaly syndromes is not fully understood. Polo-like kinase 4 (PLK4) is one of the centrosomal proteins required for centriole duplication. We here describe a patient with microcephaly and chorioretinopathy that harbors compound heterozygous missense variants, c.[442A>G]; [2336G>A], in the PLK4 gene. One of these variants, c.442A>G (p.(M148V)), resides in the kinase domain, and the other, c.2336G>A (p.(C779Y)), in the polo-box domain. Aberrant spindle formation was observed in a LCL derived from this patient. Overexpression experiments of the variant PLK4 proteins demonstrated that the p.(C779Y) but not the p.(M148V) had lost centriole overduplication ability. The altered mobility pattern of both variant proteins on a western blot further suggested alterations in post-translation modification. Our data lend support to the hypothesis that impaired centriole duplication caused by PLK4 variants may be involved in the etiology of microcephaly disorder.


Subject(s)
Choroid Diseases/genetics , Eye Diseases, Hereditary/genetics , Heterozygote , Microcephaly/genetics , Mutation, Missense , Protein Serine-Threonine Kinases/genetics , Retinal Diseases/genetics , Centrioles/metabolism , Centrioles/pathology , Choroid Diseases/diagnosis , Choroid Diseases/metabolism , Eye Diseases, Hereditary/diagnosis , Eye Diseases, Hereditary/metabolism , Female , HeLa Cells , Humans , Infant, Newborn , Microcephaly/diagnosis , Microcephaly/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Retinal Diseases/diagnosis , Retinal Diseases/metabolism
15.
Front Genet ; 7: 125, 2016.
Article in English | MEDLINE | ID: mdl-27462347

ABSTRACT

Palindromic DNA sequences, which can form secondary structures, are widely distributed in the human genome. Although the nature of the secondary structure-single-stranded "hairpin" or double-stranded "cruciform"-has been extensively investigated in vitro, the existence of such unusual non-B DNA in vivo remains controversial. Here, we review palindrome-mediated gross chromosomal rearrangements possibly induced by non-B DNA in humans. Recent advances in next-generation sequencing have not yet overcome the difficulty of palindromic sequence analysis. However, a dozen palindromic AT-rich repeat (PATRR) sequences have been identified at the breakpoints of recurrent or non-recurrent chromosomal translocations in humans. The breakages always occur at the center of the palindrome. Analyses of polymorphisms within the palindromes indicate that the symmetry and length of the palindrome affect the frequency of the de novo occurrence of these palindrome-mediated translocations, suggesting the involvement of non-B DNA. Indeed, experiments using a plasmid-based model system showed that the formation of non-B DNA is likely the key to palindrome-mediated genomic rearrangements. Some evidence implies a new mechanism that cruciform DNAs may come close together first in nucleus and illegitimately joined. Analysis of PATRR-mediated translocations in humans will provide further understanding of gross chromosomal rearrangements in many organisms.

16.
Sci Rep ; 5: 15165, 2015 Oct 23.
Article in English | MEDLINE | ID: mdl-26493046

ABSTRACT

TUBA1A mutations cause a wide spectrum of lissencephaly and brain malformations. Here, we report two patients with severe cortical dysgeneses, one with an extremely thin cerebral parenchyma apparently looking like hydranencephaly and the other with lissencephaly accompanied by marked hydrocephalus, both harbouring novel de novo missense mutations of TUBA1A. To elucidate how the various TUBA1A mutations affect the severity of the phenotype, we examined the capacity of the mutant protein to incorporate into the endogenous microtubule network in transfected COS7 cells by measuring line density using line extraction in an immunofluorescence study. The mutants responsible for severe phenotypes were found to incorporate extensively into the network. To determine how each mutant alters the microtubule stability, we examined cold-induced microtubule depolymerisation in fibroblasts. The depolymerisation of patients' fibroblasts occurred earlier than that of control fibroblasts, suggesting that microtubules bearing mutated tubulins are unstable. Both mutations are predicted to participate in lateral interactions of microtubules. Our data suggest that the TUBA1A mutations disrupting lateral interactions have pronounced dominant-negative effects on microtubule dynamics that are associated with the severe end of the lissencephaly spectrum.


Subject(s)
Hydranencephaly/genetics , Malformations of Cortical Development/genetics , Mutation , Tubulin/genetics , Child, Preschool , Exome , Female , Humans , Male , Sequence Analysis
17.
Hum Genome Var ; 2: 15003, 2015.
Article in English | MEDLINE | ID: mdl-27081519

ABSTRACT

Ichthyosis prematurity syndrome (IPS) is a rare autosomal recessive disorder characterized by prematurity, a thick caseous scale at birth and lifelong atopic diathesis. Here, we describe the first Japanese case of IPS and report novel compound heterozygous mutations (p.C403Y and p.R510H) in fatty acid transport protein 4 (FATP4). She is the first reported patient of Asian origin, entirely distinct from the Scandinavian population, in whom the heterozygote carrier frequency is very high.

18.
Pediatr Int ; 56(4): 462-6, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24980921

ABSTRACT

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.


Subject(s)
Chromosome Disorders/epidemiology , Cleft Palate/epidemiology , Heart Defects, Congenital/epidemiology , Intellectual Disability/epidemiology , Muscle Hypotonia/epidemiology , Computational Biology , Epidemiological Monitoring , Female , Humans , Japan/epidemiology , Male , Prevalence , Surveys and Questionnaires
19.
PLoS One ; 9(5): e96710, 2014.
Article in English | MEDLINE | ID: mdl-24806359

ABSTRACT

Aneuploidy in fetal chromosomes is one of the causes of pregnancy loss and of congenital birth defects. It is known that the frequency of oocyte aneuploidy increases with the human maternal age. Recent data have highlighted the contribution of cohesin complexes in the correct segregation of meiotic chromosomes. In mammalian oocytes, cohesion is established during the fetal stages and meiosis-specific cohesin subunits are not replenished after birth, raising the possibility that the long meiotic arrest of oocytes facilitates a deterioration of cohesion that leads to age-related increases in aneuploidy. We here examined the cohesin levels in dictyate oocytes from different age groups of humans and mice by immunofluorescence analyses of ovarian sections. The meiosis-specific cohesin subunits, REC8 and SMC1B, were found to be decreased in women aged 40 and over compared with those aged around 20 years (P<0.01). Age-related decreases in meiotic cohesins were also evident in mice. Interestingly, SMC1A, the mitotic counterpart of SMC1B, was substantially detectable in human oocytes, but little expressed in mice. Further, the amount of mitotic cohesins of mice slightly increased with age. These results suggest that, mitotic and meiotic cohesins may operate in a coordinated way to maintain cohesions over a sustained period in humans and that age-related decreases in meiotic cohesin subunits impair sister chromatid cohesion leading to increased segregation errors.


Subject(s)
Aging/metabolism , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Meiosis/physiology , Oocytes/metabolism , Adult , Age Factors , Aneuploidy , Animals , Female , Humans , Mice , Middle Aged , Young Adult , Cohesins
20.
Nat Commun ; 4: 1592, 2013.
Article in English | MEDLINE | ID: mdl-23481400

ABSTRACT

Gross chromosomal rearrangements (GCRs), such as translocations, deletions or inversions, are often generated by illegitimate repair between two DNA breakages at regions with nucleotide sequences that might potentially adopt a non-B DNA conformation. We previously established a plasmid-based model system that recapitulates palindrome-mediated recurrent chromosomal translocations in humans, and demonstrated that cruciform DNA conformation is required for the translocation-like rearrangements. Here we show that two sequential reactions that cleave the cruciform structures give rise to the translocation: GEN1-mediated resolution that cleaves diagonally at the four-way junction of the cruciform and Artemis-mediated opening of the subsequently formed hairpin ends. Indeed, translocation products in human sperm reveal the remnants of this two-step mechanism. These two intrinsic pathways that normally fulfil vital functions independently, Holliday-junction resolution in homologous recombination and coding joint formation in rearrangement of antigen-receptor genes, act upon the unusual DNA conformation in concert and lead to a subset of recurrent GCRs in humans.


Subject(s)
DNA, Cruciform/metabolism , Inverted Repeat Sequences/genetics , Translocation, Genetic/genetics , AT Rich Sequence , Base Sequence , Blotting, Southern , Chromosome Breakage , DNA, Cruciform/chemistry , DNA, Cruciform/genetics , DNA-Binding Proteins , Endonucleases , Gene Knockdown Techniques , Gene Rearrangement/genetics , HEK293 Cells , Holliday Junction Resolvases/metabolism , Humans , Male , Models, Biological , Molecular Sequence Data , Nuclear Proteins/metabolism , Nucleotides/genetics , Spermatozoa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...