Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters











Publication year range
1.
Am J Med Genet A ; 152A(3): 732-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20186791

ABSTRACT

Dopamine beta-hydroxylase (DBH) deficiency is characterized by a lack of sympathetic noradrenergic function. Affected individuals exhibit profound deficits in autonomic regulation of cardiovascular function. The diagnosis of DBH deficiency is based on clinical findings, biochemical studies, and sequencing of DBH gene. We report here the characterization of a mosaic cytogenetic abnormality detected by array-CGH in a 16-year-old female with primary DBH deficiency together with dysmorphic features. These features could not be explained by DBH deficiency leading to further investigation. Karyotype was reported normal (46,XX), while a targeted genomic array-CGH revealed a mosaic loss for a segment of at least 1 Mb across 11p13. This segmental loss included the PAX6 and WT1 genes within the WAGR syndrome critical region. Interestingly, the derivative chromosome 11 was observed only in about 28% of cells analyzed. Utilizing a genome-wide oligonucleotide-based array, the deletion segment was estimated to encompass a segment of approximately 10 Mb. Mosaic deletions of 11p13 in WAGR are extremely uncommon. In this case it is distinctly possible that the patient's bilateral iris colobomata might be a manifestation, albeit abbreviated, of the haploinsufficiency for PAX6. This case highlights the importance of cytogenetic analysis when a mutation alone cannot account for the complete phenotype. It also emphasizes the enhanced ability of high-resolution array-CGH techniques in accurately detecting subtle rearrangements in a mosaic form. Finally, it demonstrates the possible phenotypic effects of low-level PAX6 haploinsufficiency in a dosage-sensitive manner.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 11/genetics , Dopamine beta-Hydroxylase/deficiency , Mosaicism , Abnormalities, Multiple/genetics , Adolescent , Coloboma/genetics , Comparative Genomic Hybridization , Eye Proteins/genetics , Female , Gene Dosage , Genes, Wilms Tumor , Homeodomain Proteins/genetics , Humans , Hypotension, Orthostatic/enzymology , Hypotension, Orthostatic/genetics , In Situ Hybridization, Fluorescence , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Repressor Proteins/genetics
2.
J Med Genet ; 46(12): 825-33, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19584063

ABSTRACT

BACKGROUND: Deletions in the 17p13.3 region are associated with abnormal neuronal migration. Point mutations or deletion copy number variants of the PAFAH1B1 gene in this genomic region cause lissencephaly, whereas extended deletions involving both PAFAH1B1 and YWHAE result in Miller-Dieker syndrome characterised by facial dysmorphisms and a more severe grade of lissencephaly. The phenotypic consequences of YWHAE deletion without deletion of PAFAH1B1 have not been studied systematically. METHODS: We performed a detailed clinical and molecular characterization of five patients with deletions involving YWHAE but not PAFAH1B1, two with deletion including PAFAH1B1 but not YWHAE, and one with deletion of YWHAE and mosaic for deletion of PAFAH1B1. RESULTS: Three deletions were terminal whereas five were interstitial. Patients with deletions including YWHAE but not PAFAH1B1 presented with significant growth restriction, cognitive impairment, shared craniofacial features, and variable structural abnormalities of the brain. Growth restriction was not observed in one patient with deletion of YWHAE and TUSC5, implying that other genes in the region may have a role in regulation of growth with CRK being the most likely candidate. Using array based comparative genomic hybridisation and long range polymerase chain reaction, we have delineated the breakpoints of these nonrecurrent deletions and show that the interstitial genomic rearrangements are likely generated by diverse mechanisms, including the recently described Fork Stalling and Template Switching (FoSTeS)/Microhomology Mediated Break Induced Replication (MMBIR). CONCLUSIONS: Microdeletions of chromosome 17p13.3 involving YWHAE present with growth restriction, craniofacial dysmorphisms, structural abnormalities of brain and cognitive impairment. The interstitial deletions are mediated by diverse molecular mechanisms.


Subject(s)
14-3-3 Proteins/genetics , Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 17/genetics , Classical Lissencephalies and Subcortical Band Heterotopias/genetics , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Abnormalities, Multiple/pathology , Adolescent , Child , Child, Preschool , Chromosome Mapping , Classical Lissencephalies and Subcortical Band Heterotopias/pathology , DNA/genetics , Female , Humans , Male , Microtubule-Associated Proteins/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction
3.
Clin Genet ; 76(1): 54-62, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19558528

ABSTRACT

Array comparative genomic hybridization studies were performed to further characterize cytogenetic abnormalities found originally by karyotype and fluorescence in situ hybridization in five clinical cases of distal 10q deletions, including several with complex cytogenetic rearrangements and one with a partial male-to-female sex-reversal phenotype. These results have enabled us to narrow the previously proposed critical regions for the craniofacial, urogenital, and neuropsychiatric disease-related manifestations associated with distal 10q deletion syndrome. Furthermore, we propose that haploinsufficiency of the DOCK1 gene may play a crucial role in the pathogenesis of the 10q deletion syndrome. We hypothesize that alteration of DOCK1 and/or other genes involved in regulation and signaling of multiple pathways can explain the wide range of phenotypic variability between patients with similar or identical cytogenetic abnormalities.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 10/genetics , Adult , Child , Child, Preschool , Female , Humans , Infant, Newborn , Karyotyping , Male , Syndrome
4.
J Med Genet ; 46(9): 626-34, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19052029

ABSTRACT

BACKGROUND: Cornelia de Lange syndrome (CdLS) is a multisystem congenital anomaly disorder. Heterozygous point mutations in three genes (NIPBL, SMC3 and SMC1A), encoding components of the sister chromatid cohesion apparatus, are responsible for approximately 50-60% of CdLS cases. Recent studies have revealed a high degree of genomic rearrangements (for example, deletions and duplications) in the human genome, which result in gene copy number variations (CNVs). CNVs have been associated with a wide range of both Mendelian and complex traits including disease phenotypes such as Charcot-Marie-Tooth type 1A, Pelizaeus-Merzbacher, Parkinson, Alzheimer, autism and schizophrenia. Increased versus decreased copy number of the same gene can potentially cause either similar or different clinical features. METHODS AND RESULTS: This study identified duplications on chromosomes 5 or X using genome wide array comparative genomic hybridisation (aCGH). The duplicated regions contain either the NIPBL or the SMC1A genes. Junction sequences analyses revealed the involvement of three genomic rearrangement mechanisms. The patients share some common features including mental retardation, developmental delay, sleep abnormalities, and craniofacial and limb defects. The systems affected are the same as in CdLS, but clinical manifestations are distinct from CdLS; particularly the absence of the CdLS facial gestalt. CONCLUSIONS: The results confirm the notion that duplication CNV of genes can be a common mechanism for human genetic diseases. Defining the clinical consequences for a specific gene dosage alteration represents a new "reverse genomics" trend in medical genetics that is reciprocal to the traditional approach of delineation of the common clinical phenotype preceding the discovery of the genetic aetiology.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , De Lange Syndrome/genetics , Gene Dosage , Gene Duplication , Proteins/genetics , Adolescent , Adult , Base Sequence , Child , Child, Preschool , Chondroitin Sulfate Proteoglycans/genetics , Comparative Genomic Hybridization , Female , Humans , Infant , Male , Molecular Sequence Data , Phenotype , Sequence Alignment , Sister Chromatid Exchange
5.
Arch Dis Child ; 94(1): 55-8, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19103789

ABSTRACT

Intragenic exonic deletions, which cannot be detected by direct DNA sequencing, are a common cause of Mendelian disease. Array-based comparative genomic hybridisation (aCGH) is now widely used for the clinical diagnosis of large chromosomal deletions, but not small deletions or analysis of the mitochondrial genome. An oligonucleotide-based microarray that provides high-density coverage of the entire mitochondrial genome and nuclear genes related to mitochondrial disorders has been developed. In this report, the case of an infant referred with tyrosinaemia on newborn screening who developed liver failure is presented. DNA sequencing revealed a heterozygous missense mutation (c.679G>A, p.E227K) in the deoxyguanosine gene (DGUOK). Oligonucleotide aCGH allowed simultaneous detection of an intragenic heterozygous deletion of exon 4 of DGUOK and mitochondrial DNA depletion in blood and liver. Screening of the parents' DNA samples indicated that the patient was compound heterozygous for these mutations. An older sibling who had died from liver failure was then retrospectively diagnosed with the same mutations. This report shows the clinical utility of this oligoarray in the detection of changes in DNA copy number in both the mitochondrial and nuclear genomes, thus greatly improving the molecular diagnosis of mitochondrial disorders caused by nuclear genes involved in mitochondrial DNA biosynthesis.


Subject(s)
DNA, Mitochondrial/genetics , Deoxyguanine Nucleotides/genetics , Liver Failure/diagnosis , Mitochondrial Diseases/diagnosis , Mutation, Missense/genetics , Alanine/blood , Base Sequence , DNA Mutational Analysis , DNA, Mitochondrial/biosynthesis , Exons , Gene Deletion , Genetic Carrier Screening/methods , Humans , Infant , Infant, Newborn , Liver Failure/genetics , Male , Mitochondrial Diseases/genetics , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Tandem Mass Spectrometry , Treatment Outcome , Tyrosine/blood
6.
J Med Genet ; 46(3): 168-75, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18812404

ABSTRACT

BACKGROUND: Wolff-Parkinson-White syndrome (WPW) is a bypass re-entrant tachycardia that results from an abnormal connection between the atria and ventricles. Mutations in PRKAG2 have been described in patients with familial WPW syndrome and hypertrophic cardiomyopathy. Based on the role of bone morphogenetic protein (BMP) signalling in the development of annulus fibrosus in mice, it has been proposed that BMP signalling through the type 1a receptor and other downstream components may play a role in pre-excitation. METHODS AND RESULTS: Using the array comparative genomic hybridisation (CGH), we identified five individuals with non-recurrent deletions of 20p12.3. Four of these individuals had WPW syndrome with variable dysmorphisms and neurocognitive delay. With the exception of one maternally inherited deletion, all occurred de novo, and the smallest of these harboured a single gene, BMP2. In two individuals with additional features of Alagille syndrome, deletion of both JAG1 and BMP2 were identified. Deletion of this region has not been described as a copy number variant in the Database of Genomic Variants and has not been identified in 13 321 individuals from other cohort examined by array CGH in our laboratory. CONCLUSIONS: Our findings demonstrate a novel genomic disorder characterised by deletion of BMP2 with variable cognitive deficits and dysmorphic features and show that individuals bearing microdeletions in 20p12.3 often present with WPW syndrome.


Subject(s)
Bone Morphogenetic Protein 2/genetics , Cognition Disorders/genetics , Sequence Deletion , Wolff-Parkinson-White Syndrome/genetics , Adult , Alagille Syndrome/genetics , Animals , Calcium-Binding Proteins/genetics , Comparative Genomic Hybridization , Electrocardiography , Facies , Female , Gene Dosage , Humans , Infant , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Male , Membrane Proteins/genetics , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Serrate-Jagged Proteins , Wolff-Parkinson-White Syndrome/pathology
7.
Am J Med Genet A ; 146A(18): 2361-9, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18698622

ABSTRACT

Recent advances in molecular cytogenetics enable identification of small chromosomal aberrations that are undetectable by routine chromosome banding in 5-20% of patients with mental retardation/developmental delay (MR/DD) and dysmorphism. The aim of this study was to compare the clinical usefulness of two molecular cytogenetic techniques, metaphase high-resolution comparative genomic hybridization (HR-CGH) and targeted array CGH, also known as Chromosomal Microarray Analysis (CMA). A total of 116 patients with unexplained mild to severe MR and other features suggestive of a chromosomal abnormality with apparently normal or balanced karyotypes were analyzed using HR-CGH (43 patients) and/or CMA (91 patients). Metaphase HR-CGH detected seven interstitial deletions (16.3%). Rare deletions of chromosomes 16 (16p11.2p12.1) and 8 (8q21.11q21.2) were identified. Targeted CMA revealed copy-number changes in 19 of 91 patients (20.8%), among which 11 (11.8%) were clinically relevant, 6 (6.5%) were interpreted as polymorphic variants and 2 (2.1%) were of uncertain significance. The changes varied in size from 0.5 to 12.9 Mb. In summary, our results show that metaphase HR-CGH and array CGH techniques have become important components in cytogenetic diagnostics, particularly for detecting cryptic constitutional chromosome imbalances in patients with MR, in whom the underlying genetic defect is unknown. Additionally, application of both methods together increased the detection rates of genomic imbalances in the tested groups.


Subject(s)
Abnormalities, Multiple/genetics , Gene Deletion , Gene Duplication , Intellectual Disability/genetics , Oligonucleotide Array Sequence Analysis/methods , Child, Preschool , Developmental Disabilities/genetics , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Metaphase
8.
Clin Genet ; 72(5): 411-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17916097

ABSTRACT

High-resolution array-comparative genome hybridization (CGH) is a powerful tool for detection of submicroscopic chromosome deletions and duplications. We describe two patients with mild mental retardation (MR) and de novo microdeletions of 17q11.2q12. Although the deletions did not involve the neurofibromatosis type 1 (NF1) gene, they overlap with long-range deletions of the NF1 region which have been encountered in a small group of NF1 patients with more severe MR. Given the overlap of the deletions in our two patients with the large-sized NF1 microdeletions but not with the more frequent and smaller NF1 deletions, we hypothesize that more than one gene in the 17q11.2q12 region may be involved in MR. We discuss candidate genes for MR within this interval that was precisely defined through array-CGH analysis.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 17 , Developmental Disabilities/genetics , Nucleic Acid Hybridization , Child , Child, Preschool , Cytogenetic Analysis/methods , Female , Humans , Male
9.
Clin Genet ; 72(4): 329-38, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17850629

ABSTRACT

Monosomy 1p36 is the most common terminal deletion syndrome with an estimated occurrence of 1:5000 live births. Typically, the deletions span <10 Mb of 1pter-1p36.23 and result in mental retardation, developmental delay, sensorineural hearing loss, seizures, cardiomyopathy and cardiovascular malformations, and distinct facies including large anterior fontanel, deep-set eyes, straight eyebrows, flat nasal bridge, asymmetric ears, and pointed chin. We report five patients with 'atypical' proximal interstitial deletions from 1p36.23-1p36.11 using array-comparative genomic hybridization. Four patients carry large overlapping deletions of approximately 9.38-14.69 Mb in size, and one patient carries a small 2.97 Mb deletion. Interestingly, these patients manifest many clinical characteristics that are different from those seen in 'classical' monosomy 1p36 syndrome. The clinical presentation in our patients included: pre- and post-natal growth deficiency (mostly post-natal), feeding difficulties, seizures, developmental delay, cardiovascular malformations, microcephaly, limb anomalies, and dysmorphic features including frontal and parietal bossing, abnormally shaped and posteriorly rotated ears, hypertelorism, arched eyebrows, and prominent and broad nose. Most children also displayed hirsutism. Based on the analysis of the clinical and molecular data from our patients and those reported in the literature, we suggest that this chromosomal abnormality may constitute yet another deletion syndrome distinct from the classical distal 1p36 deletion syndrome.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1 , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis/methods , Cardiovascular Abnormalities/genetics , Child, Preschool , Craniofacial Abnormalities/genetics , Developmental Disabilities/genetics , Facies , Female , Hirsutism/genetics , Humans , Infant , Male , Syndrome
10.
Oncogene ; 20(46): 6707-17, 2001 Oct 11.
Article in English | MEDLINE | ID: mdl-11709705

ABSTRACT

Numerous LOH and mutation analysis studies in different tumor tissues, including prostate, indicate that there are multiple tumor suppressor genes (TSGs) present within the human chromosome 8p21-22 and 10q23-24 regions. Recently, we showed that LZTS1 (or FEZ1), a putative TSG located on 8p22, has the potential to function as a cell growth modulator. We report here the cloning, gene organization, cDNA sequence characterization and expression analysis of LAPSER1, an LZTS1-related gene. This gene maps within a subregion of human chromosome 10q24.3 that has been reported to be deleted in various cancers, including prostate tumors, as frequently as the neighboring PTEN locus. The complete LAPSER1 cDNA sequence encodes a predicted protein containing various domains resembling those typically found in transcription factors (P-Box, Q-rich and multiple leucine zippers). LAPSER1 is expressed at the highest levels in normal prostate and testis, where multiple isoforms are seen, some of which are either undetectable or differentially expressed in some prostate tumor tissues and cell lines. Over-expression of LAPSER1 cDNA strongly inhibited cell growth and colony-forming efficiencies of most cancer cells assessed. Together these data suggest that LAPSER1 is another gene involved in the regulation of cell growth whose loss of function may contribute to the development of cancer.


Subject(s)
Chromosomes, Human, Pair 10 , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Genes, Tumor Suppressor , Tumor Suppressor Proteins , Amino Acid Sequence , Animals , Blotting, Northern , Cell Cycle Proteins , Cell Division , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/metabolism , Databases as Topic , Gene Deletion , Gene Expression Regulation, Neoplastic , Humans , Loss of Heterozygosity , Male , Models, Genetic , Molecular Sequence Data , Prostate/metabolism , Prostatic Neoplasms/metabolism , Protein Structure, Tertiary , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Testis/metabolism , Tissue Distribution , Transfection
11.
Oncogene ; 20(31): 4169-79, 2001 Jul 12.
Article in English | MEDLINE | ID: mdl-11464283

ABSTRACT

Deletions in the 8p21-22 region of the human genome are among the most common genetic alterations in prostate carcinomas. Several studies in different tumor tissues, including prostate, indicate that there are probably multiple tumor suppressor genes (TSGs) present in this region. To identify candidate TSGs on 8p22 a YAC contig spanning this region was assembled and YAC clones retrofitted with a selectable marker (neo) were transferred into rat prostate AT6.2 cells. Two overlapping YAC clones showed greatly reduced colony-forming efficiency, indicating they may carry a TSG. Two BAC clones encompassing the overlapping region also appeared to exert suppressive effects on the growth of AT6.2 cells. Database searches for genes mapped to the critical region identified a gene known as FEZ1 (LZTS1) as a potential candidate suppressor gene. Subsequent experiments showed that over-expression of LZTS1 cDNA inhibited stable colony-forming efficiencies of AT6.2, HEK-293 and LNCaP cells. In contrast, LZTS1-transfected Rat-1 and RM1 cells were growth-stimulated. Database searches also identified additional isoforms of the LZTS1 mRNA, as well as LZTS1 protein domains reminiscent of those found in transcription factors. Together these data suggest that the LZTS1 gene is involved in the regulation of cell growth and its loss of function may contribute to the development of prostatic carcinomas, as well as other cancers.


Subject(s)
Chromosomes, Human, Pair 8 , Genes, Tumor Suppressor , Oncogenes , Prostatic Neoplasms/genetics , Animals , Cell Division , Cell Line , Chromosome Mapping , Chromosomes, Artificial, Bacterial , Chromosomes, Artificial, Yeast , Exons , Humans , Male , Plasmids , Prostate/cytology , Prostatic Neoplasms/pathology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transfection
12.
Curr Protoc Hum Genet ; Chapter 5: Unit 5.5, 2001 May.
Article in English | MEDLINE | ID: mdl-18428293

ABSTRACT

This unit describes procedures for screening large-insert libraries by multistep polymerase chain reaction (PCR) analysis of DNA samples from clone pools. In the basic protocol, PCR amplification and agarose gel electrophoresis are used to identify successively smaller pools of DNA or colonies that contain clones with the appropriate-sized amplification product. In the last screening step, individual clones are identified. The first and second support protocols describe the preparation of DNA and yeast-cell pools for screening, and the first alternate protocol describes the preparation of crude lysates suitable for PCR from individual clones or small-scale pools.


Subject(s)
Gene Library , Polymerase Chain Reaction/methods , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, P1 Bacteriophage/genetics , Chromosomes, Artificial, Yeast/genetics , DNA/genetics , DNA/isolation & purification , Electrophoresis, Agar Gel , Genetic Techniques , Genetics, Medical , Humans
13.
Genomics ; 69(1): 139-42, 2000 Oct 01.
Article in English | MEDLINE | ID: mdl-11013085

ABSTRACT

Even with the completion of a draft version of the human genome sequence only a fraction of the genes identified from this sequence have known functions. Chromosomal engineering in mouse cells, in concert with gene replacement assays to prove the functional significance of a given genomic region or gene, represents a rapid and productive means for understanding the role of a given set of genes. Both techniques rely heavily on detailed maps of chromosomal regions, initially to understand the scope of the regions being modified and finally to provide the cloned resources necessary to allow both finished sequencing and large insert complementation. This report describes the creation of a BAC clone contig on mouse chromosome 11 in a region showing conservation of synteny with sequences on human chromosome 17. We have created a detailed map of an approximately 3-cM region containing at least 33 genes through the use of multiple BAC mapping strategies, including chromosome walking and multiplex oligonucleotide hybridization and gap filling. The region described is one of the targets of a large effort to create a series of mice with regional deletions on mouse chromosome 11 (33-80 cM) that can subsequently be subjected to further mutagenesis.


Subject(s)
BRCA1 Protein/genetics , Chromosomes/genetics , Contig Mapping , Animals , Chromosomes, Bacterial/genetics , Expressed Sequence Tags , Mice , Mice, Inbred C57BL , Microsatellite Repeats
14.
Chromosoma ; 108(8): 485-90, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10794570

ABSTRACT

One of the prominent cell cycle-related modifications of histone proteins whose function remains unresolved is the phosphorylation of linker histone H1. In this work we have used indirect immunofluorescence on human cells with antibodies that are specific for phosphorylated histone H1 to examine the cellular distribution and chromosome association patterns of this protein. With confocal microscopy on whole cells, strong immunofluorescence was seen in association with mitotic chromosomes as well as a prominent punctate pattern of labeling throughout the mitotic cell, whereas interphase cells showed very little, if any, specific fluorescence. Multiple patterns of fluorescence distribution were detected with metaphase chromosomes, ranging from apparent tight colocalization with the DNA to expanded "puffy" mitotic figures to an amorphous network of staining. It was also shown that the ability to label chromosomes could vary drastically with different fixation procedures, adding further complications to interpretation of the potentially complex role of phosphorylated histone H1 in chromatin condensation or decondensation.


Subject(s)
Chromosomes, Human , Histones/immunology , Acetylation , Antibodies/immunology , Fluorescent Antibody Technique, Indirect , HeLa Cells , Histones/metabolism , Humans , Metaphase , Microscopy, Confocal , Phosphorylation
15.
Chromosoma ; 107(2): 105-12, 1998 May.
Article in English | MEDLINE | ID: mdl-9601979

ABSTRACT

The establishment of human chromosomal regions as distinct and characteristic domains has been demonstrated by the reproducible banding patterns observed on metaphase chromosomes as a result of various staining techniques. Although the exact molecular properties responsible for the patterns are not well understood, a general correlation has been established between the time of replication of a particular region of DNA and its banding characteristics. Using a replication timing assay based on fluorescence in situ hybridization patterns, we investigated replication timing properties across chromosomal regions with potentially distinct chromatin properties. Relative replication timing values were determined using cosmid DNA probes around the pseudoautosomal region boundary in Xp22.3 and the cytogenetic band boundary regions surrounding Xp22.2. Although we observed replication timing domains that were generally consistent with cytogenetic banding patterns, we did not find sharp replication timing boundaries at either the pseudoautosomal region boundary or at the cytogenetic band boundaries.


Subject(s)
Chromosome Banding , Replicon/genetics , X Chromosome/genetics , Cell Line, Transformed , Cytogenetics , DNA Probes , DNA Replication , Humans , In Situ Hybridization, Fluorescence , Male , X Chromosome/chemistry
16.
Genome Res ; 8(2): 146-57, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9477342

ABSTRACT

Prader-Willi syndrome and Angelman syndrome are associated with parent-of-origin-specific abnormalities of chromosome 15q11-q13, most frequently a deletion of an approximately 4-Mb region. Because of genomic imprinting, paternal deficiency of this region leads to PWS and maternal deficiency to AS. Additionally, this region is frequently involved in other chromosomal rearrangements including duplications, triplications, or supernumerary marker formation. A detailed physical map of this region is important for elucidating the genes and mechanisms involved in genomic imprinting, as well as for understanding the mechanism of recurrent chromosomal rearrangments. An initial YAC contig extended from D15S18 to D15S12 and was comprised of 23 YACs and 21 STSs providing an average resolution of about one STS per 200 kb. To close two gaps in this contig, YAC screening was performed using two STSs that flank the gap between D15S18 and 254B5R and three STSs located distal to the GABRA5-149A9L gap. Additionally, we developed 11 new STSs, including seven polymorphic markers. Although several groups have developed whole-genome genetic and radiation hybrid maps, the depth of coverage for 15q11-q13 has been somewhat limited and discrepancies in marker order exist between the maps. To resolve the inconsistencies and to provide a more detailed map order of STSs in this region, we have constructed an integrated YAC STS-based physical map of chromosome 15q11-q13 containing 118 YACs and 118 STSs, including 38 STRs and 49 genes/ESTs. Using an estimate of 4 Mb for the size of this region, the map provides an average STS spacing of 35 kb. This map provides a valuable resource for identification of disease genes localized to this region as well as a framework for complete DNA sequencing.


Subject(s)
Angelman Syndrome/genetics , Chromosomes, Artificial, Yeast/genetics , Chromosomes, Human, Pair 15/genetics , Prader-Willi Syndrome/genetics , Sequence Tagged Sites , Base Composition , Chromosome Mapping/methods , Genetic Markers , Humans , Polymorphism, Genetic
17.
Genome Res ; 8(1): 29-40, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9445485

ABSTRACT

The Human Genome Project has created a formidable challenge: the extraction of biological information from extensive amounts of raw sequence. With the increasing availability of genomic sequence from other species, one approach to extracting coding and regulatory element information is through cross-species sequence comparison. To assess the strengths and weaknesses of this methodology for large-scale sequence analysis, 227 kb of mouse sequence syntenic to a gene-rich cluster on human chromosome 12p13 was obtained. Primarily through percent identity plots (PIPs) of SIM comparative sequence alignments, the sequence of coding regions, putative alternative exons, conserved noncoding regions, and correlation in repetitive element insertions were easily determined. The analysis demonstrated that the number, order, and orientation of all 17 genes are conserved between the two species, whereas two human pseudogenes are absent in mouse. In addition, apart from MIRs, no direct correlation of distribution or position of the majority of repetitive elements between the two species is seen. Finally, in examining the synonymous and nonsynonymous substitution rates in the conserved genes, a large variation in nonsynonymous rates is observed indicating that the genes in this region are diverging at different rates. This study indicates the utility and strength of large-scale cross-species sequence comparisons in the extraction of biological information from raw sequence, especially when combined with other computational tools such as GRAIL and BLAST.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Chromosomes/genetics , Multigene Family , Amino Acid Sequence/genetics , Animals , Chromosome Mapping , Conserved Sequence , Humans , Mice , Molecular Sequence Data , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Sequence Analysis, DNA
18.
Nat Genet ; 17(2): 154-63, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9326934

ABSTRACT

Smith-Magenis syndrome (SMS), caused by del(17)p11.2, represents one of the most frequently observed human microdeletion syndromes. We have identified three copies of a low-copy-number repeat (SMS-REPs) located within and flanking the SMS common deletion region and show that SMS-REP represents a repeated gene cluster. We have isolated a corresponding cDNA clone that identifies a novel junction fragment from 29 unrelated SMS patients and a different-sized junction fragment from a patient with dup(17)p11.2. Our results suggest that homologous recombination of a flanking repeat gene cluster is a mechanism for this common microdeletion syndrome.


Subject(s)
Gene Deletion , Multigene Family , Recombination, Genetic , Repetitive Sequences, Nucleic Acid , Abnormalities, Multiple/genetics , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Chromosomes, Artificial, Yeast/genetics , Chromosomes, Human, Pair 17/genetics , DNA Primers/genetics , Humans , Intellectual Disability/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Syndrome
19.
Genomics ; 42(3): 436-45, 1997 Jun 15.
Article in English | MEDLINE | ID: mdl-9205115

ABSTRACT

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder that results in parathyroid, anterior pituitary, and pancreatic and duodenal endocrine tumors in affected individuals. The MEN1 locus is tightly linked to the marker PYGM on chromosome 11q13, and linkage analysis has placed the MEN1 gene within a 2-Mb interval flanked by D11S1883 and D11S449. As a step toward cloning the MEN1 gene, we have constructed a 2.8-Mb clone contig consisting of YAC and bacterial clones (PAC, BAC, and P1) for the D11S480 to D11S913 region. The bacterial clones alone represent nearly all of the 2.8-Mb contig. The contig was assembled based on a high-density STS-content analysis of 79 genomic clones (YAC, PAC, BAC, and P1) with 118 STSs. The STSs included 22 polymorphic markers and 20 transcripts, with the remainder primarily derived from the end sequences of the genomic clones. An independent cosmid contig for the 1-Mb PYGM-SEA region was also generated. Support for correctness of the 2.8-Mb contig map comes from an independent ordering of the clones by fiber-FISH. This sequence-ready contig will be a useful resource for positional cloning of MEN1 and other disease genes whose loci fall within this region.


Subject(s)
Chromosomes, Human, Pair 11 , Multiple Endocrine Neoplasia/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Proteins , Chromosomes, Artificial, Yeast , Cloning, Molecular , Cosmids , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Sequence Tagged Sites
20.
Somat Cell Mol Genet ; 23(2): 97-109, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9330638

ABSTRACT

X chromosome inactivation is associated with a highly asynchronous pattern of DNA replication at most X-linked loci in females. We studied the human HPRT locus, which is subject to X inactivation and expressed from only the active homolog, with the goal of comparing replication properties between the active and inactive homologs in this region using a fluorescence in situ hybridization approach. We found that in normal female lymphoblasts this locus is replicated in a highly asynchronous manner across a broad, discrete 500-600 kb zone with earliest replication appearing at the gene coding sequence. This general timing profile is maintained in normal male lymphoblasts, as well as in hamster x human hybrid cells containing the active human X chromosome. However, the inactive human X chromosome in the hamster cell background does not appear to function in a fully equivalent manner to the normal inactive X chromosome in female cells. Furthermore, reactivation of the inactive human X chromosome in a hamster x human hybrid system by 5-azacytidine treatment and HAT selection restores early replication at the HPRT gene itself, but does not change the overall domain behavior.


Subject(s)
DNA Replication , Hypoxanthine Phosphoribosyltransferase/genetics , X Chromosome/enzymology , Animals , Azacitidine/pharmacology , Cell Line, Transformed , Chromatin/chemistry , Cloning, Molecular , Cricetinae , DNA Replication/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Female , Gene Expression Regulation/drug effects , Humans , Hybrid Cells , Hypoxanthine Phosphoribosyltransferase/drug effects , Male , Time Factors , X Chromosome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL