Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Clin Epigenetics ; 11(1): 156, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31685013

ABSTRACT

SETD1B is a component of a histone methyltransferase complex that specifically methylates Lys-4 of histone H3 (H3K4) and is responsible for the epigenetic control of chromatin structure and gene expression. De novo microdeletions encompassing this gene as well as de novo missense mutations were previously linked to syndromic intellectual disability (ID). Here, we identify a specific hypermethylation signature associated with loss of function mutations in the SETD1B gene which may be used as an epigenetic marker supporting the diagnosis of syndromic SETD1B-related diseases. We demonstrate the clinical utility of this unique epi-signature by reclassifying previously identified SETD1B VUS (variant of uncertain significance) in two patients.


Subject(s)
Anxiety/genetics , Autism Spectrum Disorder/genetics , DNA Methylation , Epilepsy/genetics , Histone-Lysine N-Methyltransferase/genetics , Intellectual Disability/genetics , Loss of Function Mutation , Adolescent , Adult , Child , Child, Preschool , CpG Islands , Epigenesis, Genetic , F-Box Proteins/genetics , Female , Genetic Markers , Humans , Infant, Newborn , Jumonji Domain-Containing Histone Demethylases/genetics , Male
2.
Clin Genet ; 83(2): 145-54, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22369279

ABSTRACT

Higher resolution whole-genome arrays facilitate the identification of smaller copy number variations (CNVs) and their integral genes contributing to autism and/or intellectual disability (ASD/ID). Our study describes the use of one of the highest resolution arrays, the Affymetrix(®) Cytogenetics 2.7M array, coupled with quantitative multiplex polymerase chain reaction (PCR) of short fluorescent fragments (QMPSF) for detection and validation of small CNVs. We studied 82 subjects with ASD and ID in total (30 in the validation and 52 in the application cohort) and detected putatively pathogenic CNVs in 6/52 cases from the application cohort. This included a 130-kb maternal duplication spanning exons 64-79 of the DMD gene which was found in a 3-year-old boy manifesting autism and mild neuromotor delays. Other pathogenic CNVs involved 4p14, 12q24.31, 14q32.31, 15q13.2-13.3, and 17p13.3. We established the optimal experimental conditions which, when applied to select small CNVs for QMPSF confirmation, reduced the false positive rate from 60% to 25%. Our work suggests that selection of small CNVs based on the function of integral genes, followed by review of array experimental parameters resulting in highest confirmation rate using multiplex PCR, may enhance the usefulness of higher resolution platforms for ASD and ID gene discovery.


Subject(s)
Autistic Disorder/genetics , Cytogenetic Analysis/methods , DNA Copy Number Variations , Intellectual Disability/genetics , Autistic Disorder/diagnosis , Cohort Studies , Genome, Human , Humans , Intellectual Disability/diagnosis , Oligonucleotide Array Sequence Analysis/methods , Polymerase Chain Reaction/methods
3.
Hum Reprod ; 25(11): 2913-22, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20847186

ABSTRACT

BACKGROUND: Recurrent pregnancy loss (RPL), defined as two or more miscarriages, affects 3-5% of couples trying to establish a family. Despite extensive evaluation, no factor is identified in ∼40% of cases. In this study, we investigated the possibility that submicroscopic chromosomal changes, not detectable by conventional cytogenetic analysis, exist in miscarriages with normal karyotypes (46,XY or 46,XX) from couples with idiopathic RPL. METHODS: Array comparative genomic hybridization (array-CGH) was used to assess for DNA copy number variants (CNVs) in 26 miscarriages with normal karyotypes. Parental array-CGH analysis was performed to determine if miscarriage CNVs were de novo or inherited. RESULTS: There were 11 unique (previously not described) CNVs, all inherited, identified in 13 miscarriages from 8 couples. The maternal origin of two CNVs was of interest as they involved the imprinted genes TIMP2 and CTNNA3, which are only normally expressed from the maternal copy in the placenta. Two additional cohorts, consisting of 282 women with recurrent miscarriage (RM) and 61 fertile women, were screened for these two CNVs using a Quantitative Multiplex Fluorescent PCR of Short Fragments assay. One woman with RM, but none of the fertile women, carried the CTNNA3-associated CNV. CONCLUSIONS: This preliminary study shows that array-CGH is useful for detecting CNVs in cases of RPL. Further investigations of CNVs, particularly those involving genes that are imprinted in placenta, in women with RPL could be worthwhile.


Subject(s)
Abortion, Habitual/genetics , DNA Copy Number Variations/genetics , Comparative Genomic Hybridization , Female , Humans , Pregnancy , Tissue Inhibitor of Metalloproteinase-2/genetics , alpha Catenin/genetics
4.
Hum Genet ; 128(2): 179-94, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20512354

ABSTRACT

Array CGH enables the detection of pathogenic copy number variants (CNVs) in 5-15% of individuals with intellectual disability (ID), making it a promising tool for uncovering ID candidate genes. However, most CNVs encompass multiple genes, making it difficult to identify key disease gene(s) underlying ID etiology. Using array CGH we identified 47 previously unreported unique CNVs in 45/255 probands. We prioritized ID candidate genes using five bioinformatic gene prioritization web tools. Gene priority lists were created by comparing integral genes from each CNV from our ID cohort with sets of training genes specific either to ID or randomly selected. Our findings suggest that different training sets alter gene prioritization only moderately; however, only the ID gene training set resulted in significant enrichment of genes with nervous system function (19%) in prioritized versus non-prioritized genes from the same de novo CNVs (7%, p < 0.05). This enrichment further increased to 31% when the five web tools were used in concert and included genes within mitogen-activated protein kinase (MAPK) and neuroactive ligand-receptor interaction pathways. Gene prioritization web tools enrich for genes with relevant function in ID and more readily facilitate the selection of ID candidate genes for functional studies, particularly for large CNVs.


Subject(s)
Comparative Genomic Hybridization/methods , Intellectual Disability/genetics , Computational Biology , Genes , Humans
5.
Mol Hum Reprod ; 16(2): 125-34, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19778950

ABSTRACT

Developmental abnormalities of human embryos can be visualized in utero using embryoscopy. Our previous embryoscopic and genetic evaluations detected developmental abnormalities in the majority of both euploid (74%) and aneuploid or polyploid (90%) miscarriages. Since we found the pattern of morphological changes to be similar in euploid and non-euploid embryos, we proposed that lethal submicroscopic changes, not detected by standard chromosome testing, may be responsible for miscarriage of euploid embryos. Whole genome oligo and bacterial artificial chromosome array comparative genome hybridization (CGH) was used to screen for submicroscopic chromosomal changes (DNA copy number variants or CNVs) in 17 euploid embryonic miscarriages, with a range of developmental abnormalities documented by embryoscopy. The CNV breakpoints were refined using a custom array (Agilent) with high resolution coverage of the CNVs. Six unique CNVs, previously not reported, were identified in 5 of the 17 embryos (29% of all cases or 50% of cases studied with higher resolution arrays). All six unique CNVs were <250 kb in size. On the basis of parental array CGH analysis, a de novo origin of a CNV was determined for one embryo (at 13q32.1) and suspected for another (at 10p15.3). Three CNVs, at Xq28, 1q25.3 and 7p14.3, were inherited and a CNV at 17p13.1 was of unknown origin. The genes contained within these unique CNVs will be discussed, with specific reference to rearrangements of syntaxin and tryptophan-aspartic acid (WD) repeat genes. Our report describes for the first time, de novo and inherited unique CNVs in euploid human embryos with specific developmental defects.


Subject(s)
Comparative Genomic Hybridization/methods , Embryo, Mammalian/metabolism , Abortion, Spontaneous/genetics , Chromosomes, Artificial, Bacterial/genetics , DNA Copy Number Variations/genetics , Female , Humans , Pregnancy
6.
J Med Genet ; 46(10): 680-8, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19625284

ABSTRACT

BACKGROUND: Autism spectrum disorders (ASDs) are common, heritable neurobiologic conditions of unknown aetiology confounded by significant clinical and genetic heterogeneity. METHODS: This study evaluated a broad categorisation of phenotypic traits (or phenome) for 100 subjects with Autism Diagnostic Interview-Revised/Autism Diagnostic Observation Schedule-Generic (ADI-R/ADOS-G) confirmed idiopathic ASD undergoing 1 Mb bacterial artificial chromosome (BAC) array comparative genomic hybridisation (CGH). RESULTS AND CONCLUSIONS: Array CGH uncovered nine different pathogenic copy number variants (pCNVs) in 9/100 ASD subjects having complex phenotypes (ASD+/- intellectual disability (ID; IQ<70)) and/or physical anomalies), normal karyotype, fragile X analysis, and comprehensive evaluation by a clinical geneticist. Unique pCNVs in our cohort included del(5)(p15.2p15.31) (2.4 Mb), del(3)(p24.3) (0.1 Mb) and dup(18)(p11.3)(0.9 Mb). Five pCNVs were recurrent in our cohort or were previously described in subjects with ASD+/-ID: (dup(7)(q11.23)(1.5 Mb); del(2)(p15p16.1) (6.1 Mb and 7.9 Mb); del(14)(q11.2) (0.7 Mb) and dup(15)(q11q13) (10 Mb), including del(X)(p11.22) (470 Kb) in two autistic brothers. Male: female distribution in subjects with pCNVs was reduced to 1.25:1 from 3.2:1 in the original cohort. The authors stratified the study population according to a broad spectrum of clinical features and correlated specific phenotypes with respect to CNV load and pathogenicity. The findings indicate increased prevalence of pCNVs in subjects with microcephaly (<2nd centile; n = 2 of 4 ASD subjects with microcephaly; p = 0.04), and ID (n = 9 of 64 subjects with ASD and ID; p = 0.02). Interestingly, in the absence of ID co-morbidity with an ASD, no pCNVs were found. The relationship between parental ages at delivery and CNV load and pathogenicity was also explored.


Subject(s)
Child Development Disorders, Pervasive/genetics , Genetic Variation , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Female , Gene Dosage , Genotype , Humans , Male , Phenotype
7.
Am J Med Genet A ; 149A(3): 490-5, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19213034

ABSTRACT

We describe two males with intellectual disability (ID) and facial dysmorphism, both of whom have non-mosaic Y chromosome rearrangements resulting in deletions of large portions of the Y chromosome. Patient A, with ID, mild dysmorphism, speech delay, Duane anomaly of the eye, hypermetropia and conductive hearing loss, had two structurally rearranged Y chromosomes resulting in both p and q arm deletions in addition to a Yp duplication. Patient B, also with speech and language delay, developmental delay and short stature, had an interstitial deletion of Yq11.21-11.23. Array-CGH excluded the presence of additional submicroscopic rearrangements at the 1 Mb resolution level. A review of males with Y chromosome rearrangements and ID was performed. Our study provides a more detailed molecular cytogenetic assessment of Y rearrangements in individuals with ID than has been previously possible, and facilitates assessment and comparison of other individuals with a Y chromosome rearrangement.


Subject(s)
Chromosomes, Human, Y , Cytogenetic Analysis , Developmental Disabilities/genetics , Gene Rearrangement , Language Development Disorders/genetics , Child , Chromosomes, Artificial, Bacterial , Comparative Genomic Hybridization , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Young Adult
8.
Clin Genet ; 74(2): 134-44, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18498374

ABSTRACT

We describe two brothers with autistic disorder, intellectual disability (ID) and cleft lip/palate with a microdeletion of Xp11.22 detected through screening individuals with autism spectrum disorders (ASDs) for microdeletions and duplications using 1-Mb resolution array comparative genomic hybridization. The deletion was confirmed by fluorescence in situ hybridization/real-time quantitative polymerase chain reaction (RT-qPCR) and shown to be inherited from their unaffected mother who had skewed (100%) X inactivation of the aberrant chromosome. RT-qPCR characterization of the del(X)(p11.22) region ( approximately 53,887,000-54,359,000 bp) revealed complete deletion of the plant homeodomain finger protein 8 (PHF8) gene as well as deletions of the FAM120C and WNK lysine-deficient protein kinase 3 (WNK3) genes, for which a definitive phenotype has not been previously characterized. Xp11.2 is a gene-rich region within the critical linkage interval for several neurodevelopmental disorders. Rare interstitial microdeletions of Xp11.22 have been recognized with ID, craniofacial dysmorphism and/or cleft lip/palate and truncating mutations of the PHF8 gene within this region. Despite evidence implicating genes within Xp11.22 with language and cognitive development that could contribute to an ASD phenotype, their involvement with autism has not been systematically evaluated. Population screening of 481 (319 males/81 females) and 282 X chromosomes (90 males/96 females) in respective ASD and control cohorts did not identify additional subjects carrying this deletion. Our findings show that in addition to point mutations, a complete deletion of the PHF8 gene is associated with the X-linked mental retardation Siderius-Hamel syndrome (OMIM 300263) and further suggest that the larger size of the Xp11.22 deletion including genes FAM120C and WNK3 may be involved in the pathogenesis of autism.


Subject(s)
Autistic Disorder/genetics , Chromosome Deletion , Chromosomes, Human, X/genetics , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Autistic Disorder/etiology , Case-Control Studies , Cleft Lip/genetics , Cleft Palate/genetics , Family Health , Female , Gene Deletion , Histone Demethylases , Humans , Male , Mass Screening , Membrane Proteins/deficiency , Mental Disorders/genetics , Mental Retardation, X-Linked/genetics , Pedigree , Protein Serine-Threonine Kinases/deficiency , Reverse Transcriptase Polymerase Chain Reaction , Siblings , Transcription Factors/deficiency , X Chromosome Inactivation
9.
Cytogenet Genome Res ; 123(1-4): 79-87, 2008.
Article in English | MEDLINE | ID: mdl-19287141

ABSTRACT

Putatively benign copy number variants (bCNVs) can be broadly defined as DNA copy number gains or losses that do not lead to a recognizable clinical phenotype. Detection of bCNVs in genomes of clinically healthy individuals is increasing with the widespread use of whole genome arrays of different resolutions and the use of sequence comparison methods. However, the role of bCNVs in human disease susceptibility and phenotype diversity is mostly unknown. In order to explore a potential role of bCNVs in the susceptibility to and/or pathogenesis of human neurodevelopmental disorders we examined the frequency and type of common bCNVs (detected in >/=2 independent control studies) amongst 221 subjects with an autism spectrum disorder (ASD) and/or intellectual disability (ID) in comparison to 40 controls using three array platforms of increasing resolution (Spectral Genomics (1 Mb), Agilent (0.03 Mb) and NimbleGen (0.01 Mb)). We determined that the number of bCNVs/subject, type and frequency of most common bCNVs were similar for both the test and control cohorts when the same array platform was used. The comparison of the 'load' of bCNVs (i.e. number/subject) to a standardized metric of phenotypic features (see de Vries et al., 2001) in 91 ASD subjects revealed that a phenotype score >/=4 is significantly more common (P < 0.05) in persons with an ASD having one or more bCNVs via 1 Mb array-CGH, whereas individuals without any recognizable bCNVs are significantly more likely to have a less complex phenotype and a score

Subject(s)
Autistic Disorder/genetics , Cognition Disorders/genetics , Gene Dosage/genetics , Humans , Oligonucleotide Array Sequence Analysis
10.
J Med Genet ; 44(4): 269-76, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16963482

ABSTRACT

BACKGROUND: During whole genome microarray-based comparative genomic hybridisation (array CGH) screening of subjects with idiopathic intellectual disability, we identified two unrelated individuals with a similar de novo interstitial microdeletion at 2p15-2p16.1. Both individuals share a similar clinical phenotype including moderate to severe intellectual disability, autism/autistic features, microcephaly, structural brain anomalies including cortical dysplasia/pachygyria, renal anomalies (multicystic kidney, hydronephrosis), digital camptodactyly, visual impairment, strabismus, neuromotor deficits, communication and attention impairments, and a distinctive pattern of craniofacial features. Dysmorphic craniofacial features include progressive microcephaly, flat occiput, widened inner canthal distance, small palpebral fissures, ptosis, long and straight eyelashes, broad and high nasal root extending to a widened, prominent nasal tip with elongated, smooth philtrum, rounding of the upper vermillion border and everted lower lips. METHODS: Clinical assessments, and cytogenetic, array CGH and fluorescence in situ hybridisation (FISH) analyses were performed. RESULTS: The microdeletions discovered in each individual measured 4.5 Mb and 5.7 Mb, spanning the chromosome 2p region from 57.2 to 61.7 Mb and from 56 to 61.7 Mb, respectively. Each deleted clone in this range demonstrated a dosage reduction from two to one copy in each proband except for clone RP11-79K21, which was present in three copies in each proband and in four copies in their respective parents (two per each chromosome 2 homologue). DISCUSSION: The common constellation of features found in the two affected subjects indicates that they have a newly recognised microdeletion syndrome involving haploinsufficiency of one or more genes deleted within at least a 4.5-Mb segment of the 2p15-16.1 region.


Subject(s)
Abnormalities, Multiple/genetics , Autistic Disorder/genetics , Brain/abnormalities , Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 2/genetics , Craniofacial Abnormalities/genetics , Kidney/abnormalities , Attention Deficit Disorder with Hyperactivity/genetics , Child , Chromosome Disorders/pathology , Chromosomes, Human, Pair 2/ultrastructure , Female , Humans , In Situ Hybridization, Fluorescence , Infant, Newborn , Male , Nucleic Acid Hybridization , Phenotype , Syndrome
11.
Clin Genet ; 69(2): 124-34, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16433693

ABSTRACT

Autism spectrum disorders (ASDs) are a group of neurodevelopmental disorders with a strong genetic aetiology. In approximately 1% of cases, duplication of the 15q11-13 region has been reported. We report the clinical, array-comparative genomic hybridization (CGH) and cytogenetic evaluation of two individuals from a multiplex family demonstrating autism due to a maternally inherited gain of 15q11-13. Our findings indicate that unlike most 15q11-13 gains, which are caused by interstitial duplication of this region or supernumerary marker chromosomes deriving from proximal 15q, the 15q gain in this family is the result of abnormal segregation of a cryptic familial translocation with breakpoints at 14q11.2 and 15q13.3. The affected members of this family were found to have a normal karyotype at >550 band resolution. This translocation was identified using the 1-Mb resolution whole genome array (Spectral Genomics). The affected individuals have a gain of seven clones from proximal 15q, a loss of two clones from proximal 14q and a gain of two clones from 6q. Fluorescent in situ hybridization (FISH) analysis with clones from chromosomes 14 and 15, combined with DAPI reverse banding, showed an abnormal karyotype with one normal chromosome 15 and the der(15) t(14;15)(q11.2.;q13.3), resulting in the gain of proximal 15q and the loss of proximal 14q in affected individuals. The duplication of two clones from 6q in the affected subjects was also found in unaffected members of the family. Our findings suggest that the gain of 15q in autism may in some cases be due to cryptic translocations with breakpoints in the pericentromic regions of chromosome 15 and a different acrocentric chromosome. Variation in the size of pericentromic regions of any acrocentric chromosome may justify karyotype and FISH studies of autistic probands and their parents using probes from the 15q proximal region to determine recurrence risk for autism in some families.


Subject(s)
Autistic Disorder/genetics , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 15/genetics , Gene Duplication , Translocation, Genetic/genetics , Adolescent , Adult , Child , Chromosome Banding , Family Health , Female , Genome, Human , Genomics , Humans , In Situ Hybridization, Fluorescence , Infant , Infant, Newborn , Male , Nucleic Acid Hybridization , Pedigree
12.
Am J Med Genet A ; 139(3): 173-85, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16283669

ABSTRACT

Intellectual disability (ID) affects about 3% of the population (IQ < 70), and in about 40% of moderate (IQ 35-49) to severe ID (IQ < 34), and 70% of cases of mild ID (IQ 50-70), the etiology of the disease remains unknown. It has long been suspected that chromosomal gains and losses undetectable by routine cytogenetic analysis (i.e., less than 5-10 Mb in size) are implicated in ID of unknown etiology. Array CGH has recently been used to perform a genome-wide screen for submicroscopic gains and losses in individuals with a normal karyotype but with features suggestive of a chromosome abnormality. In two recent studies, the technique has demonstrated a approximately 15% detection rate for de novo copy number changes of individual clones or groups of clones. Here, we describe a study of 22 individuals with mild to moderate ID and nonsyndromic pattern of dysmorphic features suspicious of an underlying chromosome abnormality, using the 3 Mb and 1 Mb commercial arrays (Spectral Genomics). Deletions and duplications of 16 clones, previously described to show copy number variability in normal individuals [Iafrate et al., 2004; Lapierre et al., 2004; Schoumans et al., 2004; Vermeesch et al., 2005] were seen in 21/22 subjects and were considered polymorphisms. In addition, three subjects showed submicroscopic deletions and duplications not previously reported as normal variants. Two of these submicroscopic changes were of de novo origin (microdeletions at 7q36.3 and a microduplication at 11q12.3-13.1) and one was of unknown origin as parental testing of origin could not be performed (microduplication of Xp22.3). The clinical description of the three subjects with submicroscopic chromosomal changes at 7q36.3, 11q12.3-13.1, Xp22.3 is provided.


Subject(s)
Chromosome Aberrations , Chromosome Disorders/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, X/genetics , Intellectual Disability/genetics , DNA Primers , Female , Humans , In Situ Hybridization, Fluorescence , Male , Microsatellite Repeats/genetics , Nucleic Acid Hybridization/genetics , Oligonucleotide Array Sequence Analysis/methods , Pedigree , Polymerase Chain Reaction
13.
Clin Genet ; 67(4): 341-51, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15733271

ABSTRACT

Cri du Chat syndrome (CdCs) is a well-defined clinical entity, with an incidence of 1/15,000 to 1/50,000. The critical region for CdCs has been mapped to 5p15, with the hallmark cat-like cry sublocalized to 5p15.3 and the remaining clinical features to 5p15.2. We report findings in a subject with a de novo t(5;7)(p15.2;p12.2) and an inv(3)(p24q24), who was found to have a cryptic microdeletion in the critical region for CdCs detected using a 1-Mb genomic microarray. In addition to 5p deletion, the proband had a de novo single clone loss at the 3p breakpoint of inv(3)(p24q24) and a familial single clone deletion at 18q12. Deletions were confirmed using microsatellite analysis and fluorescence in situ hybridization. The 5p deletion encompasses approximately 3 Mb, mapping to the border between bands 5p15.2 and 5p15.31. The single clone deletion on chromosome 3 maps to 3p24.3-3p25, for which there is no known phenotype. The clinical features of our proband differ from the characteristic CdC phenotype, which may reflect the combined effect of the two de novo microdeletions and/or may further refine the critical region for CdCs. Typical features of CdCs that are present in the proband include moderate intellectual disability, speech, and motor delay as well as dysmorphic features (e.g. broad and high nasal root, hypertelorism, and coarse facies). Expected CdCs features that are not present are growth delay, microcephaly, round facies, micrognathia, epicanthal folds, and the signature high-pitched cry. Behavioral traits in this subject included autism spectrum disorder, attention-deficit hyperactivity disorder, and unmanageable behavior including aggression, tantrums, irritability, and self-destructive behavior. Several of these behaviors have been previously reported in patients with 5p deletion syndrome. Although most agree on the cat-cry critical region (5p15.3), there is discrepancy in the precise location and size of the region associated with the more severe manifestations of CdCs. The clinical description of this proband and the characterization of his 5p deletion may help to further refine the phenotype-genotype associations in CdCs and autism spectrum disorder.


Subject(s)
Autistic Disorder/genetics , Chromosome Deletion , Cri-du-Chat Syndrome/genetics , Adolescent , Autistic Disorder/complications , Child , Child, Preschool , Chromosome Banding , Chromosomes, Human, Pair 3/genetics , Chromosomes, Human, Pair 5/genetics , Cri-du-Chat Syndrome/complications , Female , Genotype , Humans , In Situ Hybridization, Fluorescence , Male , Microsatellite Repeats/genetics , Phenotype
15.
Genomics ; 77(3): 181-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11597143

ABSTRACT

Here we report the genomic organization and mapping of the X-linked inhibitor of apoptosis gene (BIRC4, also known as XIAP and hILP) and the identification of a closely related transcript. BIRC4 is located on Xq25 and is composed of seven exons. The intron/exon structure is highly conserved between the mouse homologue and its human counterpart. Four bands cross-react with a BIRC4 coding region probe on a genomic Southern blot. One of these cross-reactive bands encodes an intronless gene that expresses a 2.2-kb transcript solely in the testis. This testis-specific transcript contains a putative open reading frame (ORF) that is homologous to the carboxy-terminal end of BIRC4; overexpression of this ORF shows protective effects against BAX-induced apoptosis.


Subject(s)
Apoptosis , Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , X Chromosome/genetics , Animals , Base Sequence , Blotting, Southern , Blotting, Western , Cell Line , Cloning, Molecular , Conserved Sequence , Exons , Female , Gene Library , HeLa Cells , Humans , Introns , Male , Mice , Molecular Sequence Data , Open Reading Frames , Proteins/analysis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Testis/metabolism , Transfection , X-Linked Inhibitor of Apoptosis Protein , bcl-2-Associated X Protein
16.
Blood ; 98(4): 1003-11, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11493445

ABSTRACT

Several lines of evidence point to an abnormality in the response of Fanconi anemia cells to reactive oxygen species. To investigate the potential pathologic consequences of an in vivo alteration of redox state in mice lacking one of the Fanconi anemia genes, animals were generated having combined deficiencies of the cytosolic Cu/Zn superoxide dismutase (Sod1) and Fanconi anemia complementation group C (Fancc) genes. Interestingly, hepatocytes of Fancc(-/-)Sod1(-/-) mice exhibited a zonal pattern of microvesicular steatosis, possibly as a result of oxidative stress-induced injury to hepatocyte membranes. Consistent with this idea, freshly explanted Fancc(-/-)Sod1(-/-) hepatocytes demonstrated increased spontaneous production of superoxide in vitro. The second phenotypic feature of Fancc(-/-) Sod1(-/-) mice was that of bone marrow hypocellularity accompanied by significant decreases in peripheral blood erythrocyte and leukocyte numbers as compared with wild-type controls. Although flow cytometry analysis with monoclonal antibodies against cell surface antigens revealed normal numbers of primitive hematopoietic progenitor populations in Fancc(-/-)Sod1(-/-) marrow, lineage-positive progenitor numbers were significantly reduced in these mice. Furthermore, the in vitro clonogenic growth of Fancc(-/-)Sod1(-/-) erythroid, myeloid, and early B-lymphoid colonies in semisolid media was profoundly compromised. These results suggested that the altered redox state likely present in Fancc(-/-) Sod1(-/-) hematopoietic progenitors was responsible for an impairment of cell proliferation or survival. (Blood. 2001;98:1003-1011)


Subject(s)
Cell Cycle Proteins , DNA-Binding Proteins , Fatty Liver/genetics , Hematopoiesis/genetics , Nuclear Proteins , Proteins/genetics , Superoxide Dismutase/genetics , Animals , Bone Marrow Cells/pathology , Bone Marrow Diseases/pathology , Disease Models, Animal , Erythrocyte Count , Erythrocytes/pathology , Fanconi Anemia/complications , Fanconi Anemia/pathology , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Fatty Liver/complications , Leukopenia/etiology , Leukopenia/genetics , Mice , Mice, Knockout , Oxidation-Reduction , Proteins/pharmacology , Superoxide Dismutase/deficiency , Superoxide Dismutase/pharmacology , Superoxides/metabolism
17.
Am J Med Genet ; 99(4): 320-4, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11252000

ABSTRACT

We report on a female carrier of der(15) t(Y;15)(q12;p13) who had two pregnancy losses with trisomy 15 and one with tetraploidy. Molecular analysis showed that both non-disjunction events resulting in the trisomy 15 pregnancies occurred in maternal meiosis I. This finding raises the possibility that there may be an increased risk for trisomy 15 in some carriers of unbalanced t(Y;15) which, if followed by trisomic zygote rescue, may lead to uniparental disomy (UPD).


Subject(s)
Abortion, Habitual/genetics , Chromosomes, Human, Pair 15/genetics , Polyploidy , Translocation, Genetic , Abortion, Habitual/etiology , Adult , Cytogenetic Analysis , Female , Genetic Markers , Heterozygote , Humans , In Situ Hybridization, Fluorescence , Pregnancy , Trisomy , Y Chromosome
18.
Genomics ; 70(1): 113-22, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11087668

ABSTRACT

X-linked inhibitor of apoptosis protein (XIAP) is a potent modulator of programmed cell death. XIAP specifically binds and inhibits the function of caspase-3, -7, and -9, key effector proteases of apoptosis. We recently isolated, by yeast two-hybrid screening, a novel 34-kDa zinc finger protein, XIAP-associated factor 1 (XAF1). Both the caspase inhibiting and the anti-apoptotic abilities of XIAP were found to be blocked by overexpressed XAF1. Here, we report the isolation and characterization of the human XAF1 gene. The xaf1 gene consists of seven exons spanning 18 kb. Fluorescence in situ hybridization analysis localized the xaf1 locus at 17p13.2, telomeric to the p53 gene. The xaf1 locus was further refined to YAC 746C10, approximately 3 cM distal to TP53. Microsatellite analysis of the xaf1 locus using the NCI 60 cell line panel revealed significantly decreased heterozygosity at all three polymorphic markers tested, suggesting that allelic loss of the xaf1 gene is prevalent in cancer cell lines. Examination of the same NCI cell line panel for xaf1 RNA expression demonstrated that cancer cell lines exhibited very low levels of mRNA relative to normal human liver. In contrast, XIAP mRNA levels were relatively high in the majority of cancer cell lines tested. We propose that a high level of XIAP to XAF1 expression in cancer cells may provide a survival advantage through the relative increase of XIAP anti-apoptotic function.


Subject(s)
Neoplasm Proteins/genetics , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Alternative Splicing , Apoptosis , Apoptosis Regulatory Proteins , Biomarkers, Tumor , Chromosome Mapping , Female , Gene Deletion , Gene Duplication , Genomic Library , Humans , In Situ Hybridization, Fluorescence , Intracellular Signaling Peptides and Proteins , Loss of Heterozygosity , Male , Neoplasm Proteins/metabolism , Placenta , Pregnancy , Protein Binding , Tumor Cells, Cultured , Two-Hybrid System Techniques , X-Linked Inhibitor of Apoptosis Protein
19.
Cancer Genet Cytogenet ; 114(1): 71-4, 1999 Oct 01.
Article in English | MEDLINE | ID: mdl-10526539

ABSTRACT

Two different complex translocations from newly diagnosed cases of Philadelphia chromosome-positive chronic myelogenous leukemia (CML) were characterized by G-banding and fluorescence in situ hybridization (FISH) analysis. In one case, a unique balanced t(9;22;9;11) (q34;q11;p22;q23) was identified by G-banding, and confirmed by FISH using MBCR/ABL and painting probes. In the second case, an apparently balanced t(19;22) was identified by G-banding analysis. FISH using MBCR/ABL probe detected the fusion gene on the derivative chromosome 22, indicating the involvement of chromosome 9. Further FISH analysis with selected painting probes showed that the t(19;22) was a result of a complex translocation involving chromosomes 9, 19, 21, and 22.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 22 , Chromosomes, Human, Pair 9 , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Translocation, Genetic , Adult , Aged , Humans , Karyotyping , Male
20.
Cytogenet Cell Genet ; 82(3-4): 247-50, 1998.
Article in English | MEDLINE | ID: mdl-9858828

ABSTRACT

Myotonic dystrophy (DM) is caused by an expansion of a CTG repeat sequence in the 3' noncoding region of a protein kinase gene (DMPK) at 19q13.3. We used in situ hybridization to analyse the replication timing of the genomic region containing DMPK in fibroblasts and myoblasts from controls and myotonic dystrophy patients. In this method the relative proportion of singlet to doublet hybridization signals is used to infer the relative time of replication of specific loci or regions. Our results show that in cells from normal individuals approximately 65% of signals appear as doublets, indicating early replication. In DM patients with a number of CTG repeats ranging from about 600-1800 we observed a significant increase of singlet-doublets compared to the background level. These results suggest the existence of replication alternations and/or structural differences between the normal and mutant alleles induced by the presence of the DM mutation.


Subject(s)
DNA Replication , Myotonic Dystrophy/genetics , Protein Serine-Threonine Kinases/genetics , Trinucleotide Repeats , Alleles , Cells, Cultured , Cosmids , DNA Mutational Analysis , Fibroblasts/cytology , Humans , In Situ Hybridization, Fluorescence , Muscle Fibers, Skeletal/cytology , Myotonin-Protein Kinase , Skin/cytology
SELECTION OF CITATIONS
SEARCH DETAIL