Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Mol Biol ; 433(6): 166700, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33160979

ABSTRACT

Significant efforts have been recently made to obtain the three-dimensional (3D) structure of the genome with the goal of understanding how structures may affect gene regulation and expression. Chromosome conformational capture techniques such as Hi-C, have been key in uncovering the quantitative information needed to determine chromatin organization. Complementing these experimental tools, co-polymers theoretical methods are necessary to determine the ensemble of three-dimensional structures associated to the experimental data provided by Hi-C maps. Going beyond just structural information, these theoretical advances also start to provide an understanding of the underlying mechanisms governing genome assembly and function. Recent theoretical work, however, has been focused on single chromosome structures, missing the fact that, in the full nucleus, interactions between chromosomes play a central role in their organization. To overcome this limitation, MiChroM (Minimal Chromatin Model) has been modified to become capable of performing these multi-chromosome simulations. It has been upgraded into a fast and scalable software version, which is able to perform chromosome simulations using GPUs via OpenMM Python API, called Open-MiChroM. To validate the efficiency of this new version, analyses for GM12878 individual autosomes were performed and compared to earlier studies. This validation was followed by multi-chain simulations including the four largest human chromosomes (C1-C4). These simulations demonstrated the full power of this new approach. Comparison to Hi-C data shows that these multiple chromosome interactions are essential for a more accurate agreement with experimental results. Without any changes to the original MiChroM potential, it is now possible to predict experimentally observed inter-chromosome contacts. This scalability of Open-MiChroM allow for more audacious investigations, looking at interactions of multiple chains as well as moving towards higher resolution chromosomes models.


Subject(s)
Chromatin/chemistry , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 2/chemistry , Chromosomes, Human, Pair 3/chemistry , Chromosomes, Human, Pair 4/chemistry , Molecular Dynamics Simulation , Software , Animals , Cell Line, Tumor , Chromatin/metabolism , Chromatin/ultrastructure , Chromosomes, Human, Pair 1/metabolism , Chromosomes, Human, Pair 1/ultrastructure , Chromosomes, Human, Pair 2/metabolism , Chromosomes, Human, Pair 2/ultrastructure , Chromosomes, Human, Pair 3/metabolism , Chromosomes, Human, Pair 3/ultrastructure , Chromosomes, Human, Pair 4/metabolism , Chromosomes, Human, Pair 4/ultrastructure , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Lymphocytes/cytology , Lymphocytes/metabolism , Saccharum/genetics , Saccharum/metabolism , Thermodynamics , Triticum/genetics , Triticum/metabolism
2.
Sci Rep ; 9(1): 4915, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30894657

ABSTRACT

The PBX1 homeodomain transcription factor is converted by t(1;19) chromosomal translocations in acute leukemia into the chimeric E2A-PBX1 oncoprotein. Fusion with E2A confers potent transcriptional activation and constitutive nuclear localization, bypassing the need for dimerization with protein partners that normally stabilize and regulate import of PBX1 into the nucleus, but the mechanisms underlying its oncogenic activation are incompletely defined. We demonstrate here that E2A-PBX1 self-associates through the PBX1 PBC-B domain of the chimeric protein to form higher-order oligomers in t(1;19) human leukemia cells, and that this property is required for oncogenic activity. Structural and functional studies indicate that self-association facilitates the binding of E2A-PBX1 to DNA. Mutants unable to self-associate are transformation defective, however their oncogenic activity is rescued by the synthetic oligomerization domain of FKBP, which confers conditional transformation properties on E2A-PBX1. In contrast to self-association, PBX1 protein domains that mediate interactions with HOX DNA-binding partners are dispensable. These studies suggest that oligomeric self-association may compensate for the inability of monomeric E2A-PBX1 to stably bind DNA and circumvents protein interactions that otherwise modulate PBX1 stability, nuclear localization, DNA binding, and transcriptional activity. The unique dependence on self-association for E2A-PBX1 oncogenic activity suggests potential approaches for mechanism-based targeted therapies.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinogenesis/genetics , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Oncogene Proteins, Fusion/genetics , Pre-B-Cell Leukemia Transcription Factor 1/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 19/chemistry , DNA, Neoplasm/metabolism , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Oncogene Proteins, Fusion/metabolism , Pre-B-Cell Leukemia Transcription Factor 1/metabolism , Protein Binding , Protein Multimerization , Protein Stability , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Transcription, Genetic , Translocation, Genetic
3.
Mol Vis ; 25: 1-11, 2019.
Article in English | MEDLINE | ID: mdl-30820140

ABSTRACT

Purpose: To identify the mutation for Volkmann cataract (CTRCT8) at 1p36.33. Methods: The genes in the candidate region 1p36.33 were Sanger and parallel deep sequenced, and informative single nucleotide polymorphisms (SNPs) were identified for linkage analysis. Expression analysis with reverse transcription polymerase chain reaction (RT-PCR) of the candidate gene was performed using RNA from different human tissues. Quantitative transcription polymerase chain reaction (qRT-PCR) analysis of the GNB1 gene was performed in affected and healthy individuals. Bioinformatic analysis of the linkage regions including the candidate gene was performed. Results: Linkage analysis of the 1p36.33 CCV locus applying new marker systems obtained with Sanger and deep sequencing reduced the candidate locus from 2.1 Mb to 0.389 Mb flanked by the markers STS-22AC and rs549772338 and resulted in an logarithm of the odds (LOD) score of Z = 21.67. The identified mutation, rs763295804, affects the donor splice site in the long non-coding RNA gene RP1-140A9.1 (ENSG00000231050). The gene including splice-site junctions is conserved in primates but not in other mammalian genomes, and two alternative transcripts were shown with RT-PCR. One of these transcripts represented a lens cell-specific transcript. Meta-analysis of the Cross-Linking-Immuno-Precipitation sequencing (CLIP-Seq) data suggested the RNA binding protein (RBP) eIF4AIII is an active counterpart for RP1-140A9.1, and several miRNA and transcription factors binding sites were predicted in the proximity of the mutation. ENCODE DNase I hypersensitivity and histone methylation and acetylation data suggest the genomic region may have regulatory functions. Conclusions: The mutation in RP1-140A9.1 suggests the long non-coding RNA as the candidate cataract gene associated with the autosomal dominant inherited congenital cataract from CCV. The mutation has the potential to destroy exon/intron splicing of both transcripts of RP1-140A9.1. Sanger and massive deep resequencing of the linkage region failed to identify alternative candidates suggesting the mutation in RP1-140A9.1 is causative for the CCV phenotype.


Subject(s)
Cataract/congenital , Chromosomes, Human, Pair 1/chemistry , Mutation , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Acetylation , Adult , Base Sequence , Binding Sites , Cataract/diagnosis , Cataract/genetics , Cataract/pathology , Eukaryotic Initiation Factor-4A/genetics , Eukaryotic Initiation Factor-4A/metabolism , Exons , Family , Female , Genes, Dominant , Genetic Loci , Genetic Markers , High-Throughput Nucleotide Sequencing , Histones/genetics , Histones/metabolism , Humans , Introns , Male , Methylation , Middle Aged , Pedigree , RNA Splice Sites , RNA Splicing , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism
4.
Mol Hum Reprod ; 25(4): 194-205, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30770928

ABSTRACT

Endometriotic lesions are composed in part of endometrial-like stromal cells, however, there is a shortage of immortalized human endometrial stromal cultures available for research. As genetic factors play a role in endometriosis risk, it is important that genotype is also incorporated into analysis of pathological mechanisms. Human telomerase reverse transcriptase (hTERT) immortalization (using Lenti-hTERT-green fluorescent protein virus) took place following genotype selection; 13 patients homozygous for either the risk or non-risk 'other' allele for one or more important endometriosis risk single nucleotide polymorphism on chromosome 1p36.12 (rs3820282, rs56318008, rs55938609, rs12037376, rs7521902 or rs12061255). Short tandem repeat DNA profiling validated that donor tissue matched that of the immortalized cell lines and confirmed that cultures were genetically novel. Expression of morphological markers (vimentin and cytokeratin) and key genes of interest (telomerase, estrogen and progesterone receptors and LINC00339) were examined and functional assays for cell proliferation, steroid hormone and inflammatory responses were performed for 7/13 cultures. All endometrial stromal cell lines maintained their fibroblast-like morphology (vimentin-positive) and homozygous endometriosis-risk genotype following introduction of hTERT. Furthermore, the new stromal cultures demonstrated positive and diverse responses to hormones (proliferation and decidualisation changes) and inflammation (dose-dependent response), while maintaining hormone receptor expression. In conclusion, we successfully developed a range of human endometrial stromal cell lines that carry important endometriosis-risk alleles. The wider implications of this approach go beyond advancing endometriosis research; these cell lines will be valuable tools for multiple endometrial pathologies offering a level of genetic and phenotypic diversity not previously available.


Subject(s)
Endometriosis/genetics , Founder Effect , Genotype , Stromal Cells/metabolism , Telomerase/genetics , Adult , Biomarkers/metabolism , Cell Line, Transformed , Cell Proliferation , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 1/metabolism , Endometriosis/metabolism , Endometriosis/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Gene Expression , Homozygote , Humans , Keratins/genetics , Keratins/metabolism , Microsatellite Repeats , Polymorphism, Single Nucleotide , RNA, Long Noncoding , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Risk , Stromal Cells/pathology , Telomerase/metabolism , Vimentin/genetics , Vimentin/metabolism
6.
Am J Med Genet A ; 173(4): 972-977, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28328126

ABSTRACT

As genome wide techniques become more common, an increasing proportion of patients with intellectual disability (ID) are found to have genetic defects allowing genotype-phenotype correlations. Previously, AKT3 deletion was suggested to be responsible for microcephaly in patients with 1q43-q44 deletion syndrome, but this does not correspond to all cases. We report a case of a de novo 1q44 deletion in an 8-year-old boy with microcephaly in whom AKT3 is not deleted. We used a systematic review of the literature, our patient, and network analysis to gain a better understanding of the genetic basis of microcephaly in 1q deletion patients. Our analysis showed that while AKT3 deletion is associated with more severe (≤3 SD) microcephaly in 1q43-q44 deletion patients, other genes may contribute to microcephaly in AKT3 intact patients with microcephaly and 1q43-44 deletion syndrome. We identified a potential role for HNRNPU, SMYD3, NLRP3, and KIF26B in microcephaly. Overall, our study highlights the need for network analysis and quantitative measures reporting in the phenotypic analysis of a complex genetic syndrome related to copy number variation.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1/chemistry , Gene Regulatory Networks , Intellectual Disability/genetics , Microcephaly/genetics , Proto-Oncogene Proteins c-akt/genetics , Child , Computational Biology , DNA Copy Number Variations , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Intellectual Disability/diagnosis , Intellectual Disability/pathology , Kinesins/genetics , Male , Microcephaly/diagnosis , Microcephaly/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phenotype
7.
Nat Commun ; 8: 14759, 2017 03 13.
Article in English | MEDLINE | ID: mdl-28287101

ABSTRACT

Genome-wide association studies (GWAS) do not distinguish between single nucleotide polymorphisms (SNPs) that are causal and those that are merely in linkage-disequilibrium with causal mutations. Here we describe a versatile, functional pipeline and apply it to SNPs at 1p22, a locus identified in several GWAS for non-syndromic cleft lip with or without cleft palate (NS CL/P). First we amplified DNA elements containing the ten most-highly risk-associated SNPs and tested their enhancer activity in vitro, identifying three SNPs with allele-dependent effects on such activity. We then used in vivo reporter assays to test the tissue-specificity of these enhancers, chromatin configuration capture to test enhancer-promoter interactions, and genome editing in vitro to show allele-specific effects on ARHGAP29 expression and cell migration. Our results further indicate that two SNPs affect binding of CL/P-associated transcription factors, and one affects chromatin configuration. These results translate risk into potential mechanisms of pathogenesis.


Subject(s)
Chromosomes, Human, Pair 1/chemistry , Cleft Lip/genetics , Cleft Palate/genetics , GTPase-Activating Proteins/genetics , Genetic Predisposition to Disease , Transcription Factors/genetics , Alleles , Animals , Animals, Genetically Modified , Biological Assay , Chromatin/chemistry , Cleft Lip/diagnosis , Cleft Lip/pathology , Cleft Palate/diagnosis , Cleft Palate/pathology , Enhancer Elements, Genetic , GTPase-Activating Proteins/metabolism , Gene Expression , Genes, Reporter , Genetic Loci , Genome-Wide Association Study , Humans , Linkage Disequilibrium , Luciferases/genetics , Luciferases/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Risk Factors , Transcription Factors/metabolism , Zebrafish
8.
Kaohsiung J Med Sci ; 33(1): 1-10, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28088267

ABSTRACT

The aim of this meta-analysis was to detect whether three identified single nucleotide polymorphisms (SNPs) (rs646776, rs599839, and rs17465637) at 1p13.3 and 1q41 are associated with lipid levels and the risk of coronary artery disease (CAD). Databases of MEDLINE, EMBASE, the Cochrane Library, and BIOSIS were systematically searched. The pooled effects were expressed as odds ratio or standardized mean difference or mean difference with 95% confidence intervals. A total of 14 studies with 57,916 patients were included in the meta-analysis. Pooled effects showed that the AA group of 1p13.3 rs599839 had higher total cholesterol (TC) and low-density lipoprotein cholesterol (LDLC), and lower high-density lipoprotein cholesterol (HDLC) levels than the GA/GG group, and the CAD group had higher AA genotype frequency than the control group. The TT group of 1p13.3 rs646776 had higher TC and LDLC levels and lower HDLC levels than the CT/CC group. The CAD group also had higher CC genotype frequency of 1q41 rs17465637 than the control group. The SNPs of 1p13 rs599839 and rs646776 were associated with serum lipid levels. The genetic variants of 1p13 rs599839 and 1q41 rs17465637 SNPs were prominently related to CAD, and the genetic variants of chromosome 1p13 promote the risk of CAD by increased TC and LDLC levels and decreased HDLC levels.


Subject(s)
Chromosomes, Human, Pair 1/chemistry , Coronary Artery Disease/genetics , Genetic Loci , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Aged , Alleles , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Coronary Artery Disease/pathology , Female , Gene Frequency , Genotype , Humans , Male , Middle Aged , Odds Ratio , Risk
10.
Tsitol Genet ; 50(4): 74-8, 2016.
Article in English | MEDLINE | ID: mdl-30480420

ABSTRACT

Complex chromosomal rearrangements are rarely observed prenatally. Genetic counceling of CCR carriers is complicated, especially in cases of de novo origin of the rearrangement. Here we present a new case of a de novo CCR involving four chromosomes observed in amniotic fluid cells of the fetus at 17 weeks of gestation. The rearrangement was characterized as an apparently balanced four-way translocation t(1;11;7;13)(~p21;~q13.5;~q32;~q22)dn by conventional cytogenetic studies. However, array-based comparative genomic hybridization revealed 5 submicroscopic heterozygous interstitial deletions on chromosome 1, 11, 7, 13 with a total loss of 21.1 Mb of genetic material in regions close to those, designated as breakpoints by conventional cytogenetic analysis. The described case clearly illustrates that high-resolution molecular genetic analysis should be combined with conventional cytogenetic techniques to exclude subtle chromosomal abnormalities in CCR cases detected prenatally.


Subject(s)
Amniotic Fluid/cytology , Chromosomes, Human, Pair 11/chemistry , Chromosomes, Human, Pair 13/chemistry , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 7/chemistry , Translocation, Genetic , Amniocentesis , Comparative Genomic Hybridization , Female , Fetus , Gestational Age , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Male , Pregnancy , Pregnancy Trimester, Second , Young Adult
11.
Clin Chim Acta ; 445: 34-40, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25797897

ABSTRACT

Several recurrent copy number variants (CNVs) increasing risk to neuropsychiatric diseases have been identified in recent years. They show variable clinical expressivity, being associated with different disorders, and incomplete penetrance. However, due to its very low frequency, the full variety of clinical outcomes associated with each one of these CNVs is unknown. Current methods for detection of CNVs are labor intensive, expensive or not suitable for high throughput analysis. Quantitative interspecies competitive PCR linked to variant minisequencing and detection by mass-spectrometry may overcome these limitations. Here, we present two multiplex assays based on this method to screen for eleven psychiatric risk CNVs, such as 1q21, 16p11.2, 3q29, or 16p13.11 regions, among others. The assays were tested in our collection of 514 schizophrenia patients. Results were compared with MLPA at two CNVs. Additional positive results were confirmed by exome sequencing. A total of fourteen patients were CNV carriers. The method presents high sensitivity and specificity, showing its utility as a cheap, accurate, high throughput screening tool for recurrent CNVs. The method may be very useful for management of psychiatric patients as well as screening of different collections of samples to better identify the full spectrum of clinical variability.


Subject(s)
DNA Copy Number Variations , Genome, Human , High-Throughput Screening Assays , Schizophrenia/genetics , Animals , Cell Line , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 16/chemistry , Chromosomes, Human, Pair 3/chemistry , Exome , Genetic Predisposition to Disease , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Oligonucleotide Array Sequence Analysis , Pan troglodytes/genetics , Penetrance , Schizophrenia/diagnosis
12.
Bioinformatics ; 30(14): 2070-2, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24642064

ABSTRACT

MOTIVATION: Converting nucleotide sequences into short overlapping fragments of uniform length, k-mers, is a common step in many bioinformatics applications. While existing software packages count k-mers, few are optimized for speed, offer an application programming interface (API), a graphical interface or contain features that make it extensible and maintainable. We designed KAnalyze to compete with the fastest k-mer counters, to produce reliable output and to support future development efforts through well-architected, documented and testable code. Currently, KAnalyze can output k-mer counts in a sorted tab-delimited file or stream k-mers as they are read. KAnalyze can process large datasets with 2 GB of memory. This project is implemented in Java 7, and the command line interface (CLI) is designed to integrate into pipelines written in any language. RESULTS: As a k-mer counter, KAnalyze outperforms Jellyfish, DSK and a pipeline built on Perl and Linux utilities. Through extensive unit and system testing, we have verified that KAnalyze produces the correct k-mer counts over multiple datasets and k-mer sizes. AVAILABILITY AND IMPLEMENTATION: KAnalyze is available on SourceForge: https://sourceforge.net/projects/kanalyze/.


Subject(s)
Sequence Analysis, DNA/methods , Software , Algorithms , Chromosomes, Human, Pair 1/chemistry , Humans
13.
Nucleic Acids Res ; 42(2): 893-905, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24137003

ABSTRACT

Repair of DNA lesions must occur within the chromatin landscape and is associated with alterations in histone modifications and nucleosome rearrangement. To directly associate these chromatin features with DNA damage and repair, it is necessary to be able to map DNA adducts. We have developed a cyclobutane pyrimidine dimer (CPD)-specific immunoprecipitation method and mapped ultraviolet damage hotspots across human chromosomes 1 and 6. CPD hotspots occur almost equally in genic and intergenic regions. However, these hotspots are significantly more prevalent adjacent to repeat elements, especially Alu repeats. Nucleosome mapping studies indicate that nucleosomes are consistently positioned at Alu elements where CPD hotspots form, but by 2 h post-irradiation, these same regions are significantly depleted of nucleosomes. These results indicate that nucleosomes associated with hotspots of CPD formation are readily rearranged, potentially making them accessible to DNA repair machinery. Our results represent the first chromosome scale map of ultraviolet-induced DNA lesions in the human genome, and reveal the sequence features and dynamic chromatin changes associated with CPD hotspots.


Subject(s)
DNA Adducts/analysis , Pyrimidine Dimers/analysis , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 6/chemistry , DNA/chemistry , DNA/isolation & purification , DNA Adducts/isolation & purification , Fibroblasts/chemistry , Humans , Immunoprecipitation , Nucleosomes/metabolism , Poly T/chemistry , Pyrimidine Dimers/isolation & purification , Repetitive Sequences, Nucleic Acid
14.
Mol Vis ; 17: 1449-56, 2011.
Article in English | MEDLINE | ID: mdl-21677793

ABSTRACT

PURPOSE: To investigate the genotype and phenotype of juvenile-onset open angle glaucoma (JOAG) in a Chinese family (PN pedigree). METHODS: Each family member was comprehensively examined by an experienced ophthalmologist. The clinical characteristics of the family patients with JOAG were documented. Blood samples were obtained from 22 available participants from the PN pedigree. Linkage analysis was performed to identify the possible chromosome loci. The presence of gene mutation was ascertained by polymerase chain reaction amplification and subsequent direct sequencing. RESULTS: The affected members in the PN pedigree are characterized by early age of onset (mean age at diagnosis is 17 years old), severe clinical presentations, high intraocular pressure (mean IOP of 34.18±2.97 mmHg), and poor response to pharmacological treatment (87.5% of the patients required filtering surgery). The region on chromosome 1 between D1S3464 and D1S1619 was identified in this pedigree by linkage analysis. A Pro370Leu myocilin mutation resulting from a heterozygous C→T transition at the 1,109th nucleotide in exon 3 was detected by gene sequencing. The Pro370Leu mutation co-segregated among all affected individuals of PN pedigree. CONCLUSIONS: The GLC1A Pro370Leu mutation is firmly correlated with a severe POAG phenotype. These data provide clues for the severe disease-causing nature of the Pro370Leu allele. Gene screening may be a useful method for pre-symptom diagnosis and a forewarning to detect the at-risk individuals in familial open-angle glaucoma patients, especially in pedigrees of early-onset.


Subject(s)
Asian People/genetics , Cytoskeletal Proteins/genetics , Eye Proteins/genetics , Genetic Association Studies , Glaucoma, Open-Angle/genetics , Glycoproteins/genetics , Ocular Hypertension/genetics , Adolescent , Adult , Age of Onset , Base Sequence , Child , Chromosomes, Human, Pair 1/chemistry , Female , Genetic Linkage , Genetic Loci , Genetic Testing , Genotype , Glaucoma, Open-Angle/complications , Glaucoma, Open-Angle/diagnosis , Glaucoma, Open-Angle/surgery , Heterozygote , Humans , Male , Molecular Sequence Data , Mutation , Ocular Hypertension/complications , Ocular Hypertension/diagnosis , Pedigree , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Tomography, Optical Coherence , Visual Acuity
15.
Ultramicroscopy ; 107(4-5): 308-12, 2007.
Article in English | MEDLINE | ID: mdl-17034949

ABSTRACT

Scanning near-field optical microscopy (SNOM) has been successfully employed to generate high resolution (<100nm) fluorescence images of directly tagged human chromosomes. Direct tagging, fluorescence in-situ hybridisation processes (with and without amplification) are investigated and their fluorescence response to near-field excitation are compared. Using the simultaneous topography mode of SNOM, chromosome morphology was seen to differ as a result of the two processes; with chromatin collapse more extensive when the amplified direct tagging procedure was used. The results are discussed in the context of developing locus specific direct tags together with high resolution SNOM imaging for the observation of chromosome aberrations.


Subject(s)
Chromosomes, Human, Pair 1/chemistry , Fluorescein-5-isothiocyanate/chemistry , Microscopy, Fluorescence/methods , Chromosome Painting , Humans , Lymphocytes/ultrastructure
16.
Cancer Biol Ther ; 4(4): 440-8, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15846090

ABSTRACT

Hypermethylation at certain CpG-rich promoters and hypomethylation at repeated DNA sequences are very frequently found in cancers. We provide the first report that a DNA sequence (NBL2) can be either extensively hypermethylated or hypomethylated in cancer. Previously, it was shown that NBL2, a complex tandem DNA repeat in the acrocentric chromosomes, is hypomethylated at NotI sites in >70% of neuroblastomas and hepatocellular carcinomas and in cells from ICF syndrome (DNMT3B-deficiency) patients. Unexpectedly, by Southern blot analysis of 18 ovarian carcinomas, 51 Wilms tumors, and various somatic control tissues, we found that >70% of the cancers exhibited large increases in methylation at HhaI sites in NBL2 compared with all the controls. In contrast, 17% of the carcinomas showed major decreases in methylation at HhaI and NotI sites. The intermediate levels of methylation at HhaI sites in somatic controls enabled this discovery of cancer-linked hypermethylation and hypomethylation in NBL2. In a comparison of ovarian epithelial carcinomas, low malignant potential tumors, and cystadenomas, NBL2 hypermethylation at HhaI sites was significantly related to the degree of malignancy, and hypomethylation was seen only in the carcinomas. By RT-PCR, we found NBL2 transcripts at low levels in a few cancers and undetectable in various normal tissues. In the tumors there was no association of NBL2 hypomethylation and transcription, but this may reflect NBL2's lack of identifiable promoter elements and our evidence for run-through transcription from adjacent sequences into NBL2. The propensity of NBL2 sequences to become either hypermethylated or hypomethylated in cancer suggests that these opposite epigenetic changes share an early step during carcinogenesis and that cancer-linked hypermethylation might be spontaneously reversible.


Subject(s)
DNA Methylation , DNA, Satellite/genetics , Ovarian Neoplasms/genetics , Wilms Tumor/genetics , Chromosome Mapping , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 1/genetics , CpG Islands/genetics , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Female , Humans , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction
17.
Mol Diagn ; 8(2): 93-100, 2004.
Article in English | MEDLINE | ID: mdl-15527323

ABSTRACT

BACKGROUND: In the last decade, microarray technology has been extensively used to evaluate gene expression profiles and genome imbalances. We have developed a microarray-based comparative genomic hybridization (CGH) approach to identify MYCN gene amplification and 1p36 chromosome loss, two markers of tumor aggressiveness in neuroblastoma. AIM: The aim was to use microarray CGH technology to detect the two major prognostic markers for neuroblastoma, MYCN amplification and 1p36 chromosome deletion, in neuroblastoma patients and, therefore, confirm the usefulness of this approach in this cancer. METHODS: DNA was purified from 16 tumors containing at least 90% malignant neuroblasts and collected at the onset of disease. Pooled fluorescent-labeled reference and neuroblastoma tumor genomic DNA was hybridized to epoxide-coated glass slides on laboratory-made complementary DNA microarray. The microarray contained cDNA mapped at the 1p36.33-36.1 chromosomal region and MYCN gene. cDNA from the 2q33-q34 and 12p13 chromosomes was used as a control and Arabidopsis thaliana DNA was spotted to control unspecific hybridization. Fluorescence in situ hybridization analysis was also performed to validate results from the microarray CGH. RESULTS: Both MYCN amplification and 1p36 chromosome deletion were detected by microarray CGH. The sensitivity and specificity for 1p36 loss detection were 66.7% and 90.0%, respectively. The method had a sensitivity of 66.7% and specificity of 90.9% to detect MYCN amplification. DISCUSSION: Our results demonstrated that the microarray CGH can be efficiently applied to study DNA gain and loss of specific chromosome regions.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Gene Amplification , Neuroblastoma/diagnosis , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Oncogene Proteins/genetics , Cell Nucleus/chemistry , Chromosomes, Human, Pair 1/chemistry , Fluorescent Dyes/chemistry , Genetic Markers , Humans , Indoles/chemistry , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Nuclear Proteins/chemistry , Oncogene Proteins/chemistry , Prognosis
18.
Neuro Oncol ; 6(2): 96-103, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15134623

ABSTRACT

Survival periods vary considerably for patients with high-grade astrocytomas, and reliable prognostic markers are not currently available. We therefore investigated whether genetic losses from chromosomes 1p, 19q, 9p, or 10q were associated with survival in 89 high-grade astrocytomas using tissue microarrays (TMAs) derived from Radiation Therapy Oncology Group clinical trials. Cases included 15 anaplastic astrocytomas (AAs) and 74 glioblastomas (GBMs) selected on the basis of survival times significantly shorter or longer than the expected median. Genetic analysis was performed by TMA-fluorescence in situ hybridization (FISH) on array sections using 8 DNA probes, including those directed at 1p32, 19q13.4, 9p21 (p16/CDKN2A), and 10q (PTEN and DMBT1). Genetic status for each locus was correlated with patient survival group, and data were analyzed by using Fisher's exact test of association (adjusted P = 0.025). Losses of chromosome 1p, either alone or in combination with 19q, were encountered in only 2 cases, both AAs. This contrasts with oligodendrogliomas, in which combined 1p and 19q losses are frequent and predictive of prolonged survival. Solitary 19q loss was noted in 3/15 AAs and in 7/70 GBMs and was more frequent in the long-term survival group (P = 0.041, AA and GBM combined). Chromosome 9p loss was seen in 5/8 AAs and 39/57 GBMs, whereas chromosome 10q loss was detected in 4/15 AAs and 48/68 GBMs. The 9p and 10q deletions were slightly more frequent in short-term survivors, though none of the comparisons achieved statistical significance. Long-term and short-term survival groups of high-grade astrocytomas appear to have dissimilar frequencies of 19q, 9p, and 10q deletions. TMA-FISH is a rapid and efficient way of evaluating genetic alterations in such tumors.


Subject(s)
Astrocytoma/diagnosis , Astrocytoma/genetics , Chromosomes, Human, Pair 10/chemistry , Chromosomes, Human, Pair 19/chemistry , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 9/chemistry , Adult , Aged , Astrocytoma/pathology , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 9/genetics , Clinical Trials as Topic/methods , Genetic Markers/genetics , Humans , In Situ Hybridization, Fluorescence/methods , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis
19.
J Neurosci Res ; 56(2): 146-59, 1999 Apr 15.
Article in English | MEDLINE | ID: mdl-10494103

ABSTRACT

Rxt1, a transporter-like protein structurally related to the large family of Na(+)/Cl(-)-dependent carriers, was isolated from the rat brain. In the present study, Hxt1, the homologue of Rxt1, was isolated from human cortex cDNA. Comparison of their respective nucleotidic sequences revealed a 96% conservation between Hxt1 and Rxt1. Genetic mapping with human genome radiation hybrids allowed the location of the gene coding for Hxt1 between 323ya5 and 084xb3 AFM markers, on a portion of chromosome 1p which spans over 7 cM or 118 cRay. Northern blot analyses demonstrated that Hxt1 mRNA ( approximately 7.5 Kb) is expressed in the human brain but not in peripheral tissues. The immunodistribution of Hxt1 was determined with antibodies raised against the C-terminus of Rxt1. Hxt1 is concentrated in the cerebral cortex, caudate-putamen, substantia nigra, hippocampus, and cerebellum, appearing as a diffuse or a punctate labeling at the light microscope level. This regional and cellular distribution suggests that Hxt1, as its rat homologue, could be present in axon terminals of glutamatergic neurons. The high pressure of selection exerted upon this protein, its strategic anatomical and subcellular distributions suggest that this orphan transporter could be involved in critical functions in the central nervous system.


Subject(s)
Brain Chemistry/genetics , Carrier Proteins/genetics , Cerebral Cortex/chemistry , Chromosomes, Human, Pair 1/genetics , Symporters , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/chemistry , Chromosomes, Human, Pair 1/chemistry , Humans , Molecular Sequence Data , Rats , Sodium Chloride Symporters
20.
Mutat Res ; 423(1-2): 91-101, 1999 Jan 25.
Article in English | MEDLINE | ID: mdl-10029684

ABSTRACT

Rearrangements in heterochromatin in the vicinity of the centromeres of chromosomes 1 and 16 are frequent in many types of cancer, including ovarian epithelial carcinomas. Satellite 2 DNA is the main sequence in the unusually long heterochromatin region adjacent to the centromere of each of these chromosomes. Rearrangements in these regions and hypomethylation of satellite 2 DNA are a characteristic feature of patients with a rare recessive genetic disease, ICF (immunodeficiency, centromeric region instability, and facial anomalies). In all normal tissues of postnatal somatic origin, satellite 2 DNA is highly methylated. We examined satellite 2 DNA methylation in ovarian tumors of different malignant potential, namely, ovarian cystadenomas, low malignant potential (LMP) tumors, and epithelial carcinomas. Most of the carcinomas and LMP tumors exhibited hypomethylation in satellite 2 DNA of both chromosomes 1 and 16. A comparison of methylation of these sequences in the three types of ovarian neoplasms demonstrated that there was a statistically significant correlation between the extent of this satellite DNA hypomethylation and the degree of malignancy (P<0.01). Also, there was a statistically significant association (P<0.005) between genome-wide hypomethylation and undermethylation of satellite 2 DNA among these 17 tumors. In addition, we found abnormal hypomethylation of satellite alpha DNA in the centromere of chromosome 1 in many of these tumors. Our findings are consistent with the hypothesis that one of the ways that genome-wide hypomethylation facilitates tumor development is that it often includes satellite hypomethylation which might predispose cells to structural and numerical chromosomal aberrations. Several of the proteins that bind to pericentromeric heterochromatin are known to be sensitive to the methylation status of their target sequences and so could be among the sensors for detecting abnormal demethylation and mediating effects on chromosome structure and stability.


Subject(s)
Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , DNA Methylation , DNA, Satellite/metabolism , Ovarian Neoplasms/genetics , 5-Methylcytosine , Carcinoma/chemistry , Centromere/chemistry , Centromere/genetics , Chromosomes, Human, Pair 1/chemistry , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 16/chemistry , Chromosomes, Human, Pair 16/genetics , Cytosine/analogs & derivatives , Cytosine/analysis , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , Female , Humans , Ovarian Neoplasms/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...