Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Am J Hum Genet ; 108(9): 1725-1734, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34433009

ABSTRACT

Copy-number variations (CNVs) are a common cause of congenital limb malformations and are interpreted primarily on the basis of their effect on gene dosage. However, recent studies show that CNVs also influence the 3D genome chromatin organization. The functional interpretation of whether a phenotype is the result of gene dosage or a regulatory position effect remains challenging. Here, we report on two unrelated families with individuals affected by bilateral hypoplasia of the femoral bones, both harboring de novo duplications on chromosome 10q24.32. The ∼0.5 Mb duplications include FGF8, a key regulator of limb development and several limb enhancer elements. To functionally characterize these variants, we analyzed the local chromatin architecture in the affected individuals' cells and re-engineered the duplications in mice by using CRISPR-Cas9 genome editing. We found that the duplications were associated with ectopic chromatin contacts and increased FGF8 expression. Transgenic mice carrying the heterozygous tandem duplication including Fgf8 exhibited proximal shortening of the limbs, resembling the human phenotype. To evaluate whether the phenotype was a result of gene dosage, we generated another transgenic mice line, carrying the duplication on one allele and a concurrent Fgf8 deletion on the other allele, as a control. Surprisingly, the same malformations were observed. Capture Hi-C experiments revealed ectopic interaction with the duplicated region and Fgf8, indicating a position effect. In summary, we show that duplications at the FGF8 locus are associated with femoral hypoplasia and that the phenotype is most likely the result of position effects altering FGF8 expression rather than gene dosage effects.


Subject(s)
Chromosome Duplication , Chromosomes, Human, Pair 10/chemistry , DNA Copy Number Variations , Fibroblast Growth Factor 8/genetics , Lower Extremity Deformities, Congenital/genetics , Adolescent , Alleles , Animals , CRISPR-Cas Systems , Child, Preschool , Chromatin/chemistry , Chromatin/metabolism , Chromosomes, Human, Pair 10/metabolism , Enhancer Elements, Genetic , Family , Female , Femur/abnormalities , Femur/diagnostic imaging , Femur/metabolism , Fibroblast Growth Factor 8/metabolism , Gene Editing , Heterozygote , Humans , Infant , Lower Extremity Deformities, Congenital/diagnostic imaging , Lower Extremity Deformities, Congenital/metabolism , Lower Extremity Deformities, Congenital/pathology , Male , Mice , Mice, Transgenic , Pedigree , Phenotype
2.
Mol Genet Metab ; 120(3): 173-179, 2017 03.
Article in English | MEDLINE | ID: mdl-28087245

ABSTRACT

Heterozygous Fabry females usually have an attenuated form of Fabry disease, causing them to be symptomatic; however, in rare cases, they can present with a severe phenotype. In this study, we report on a 37-year-old woman with acroparesthesia, a dysmorphic face, left ventricular hypertrophy, and intellectual disability. Her father had Fabry disease and died due to chronic renal and congestive cardiac failure. Her paternal uncle had chronic renal failure and intellectual disability, and her paternal aunt was affected with congestive cardiac failure. The patient has two sisters with no significant medical illness. However, her nephew has acroparesthesia, anhidrosis, and school phobia, and her niece shows mild phenotypes. The patient's enzyme analysis showed very low α-galactosidase A (α-gal A) activity in dried blood spot (DBS), lymphocytes, and skin fibroblasts with massive excretion of Gb3 and Gb2 in urine and lyso-Gb3 in DBS and plasma. Electron microscopic examination showed a large accumulation of sphingolipids in vascular endothelial cells and keratinocytes. Chromosomal analysis and comparative genomic hybridization microarray showed 10q26 terminal deletion. Molecular data showed a novel heterozygous stop codon mutation in exon 1 of the GLA gene in her sisters and niece, and a hemizygous state in her nephew. When we checked the methylation status, we found her non-mutated allele in the GLA gene was methylated. However, the non-mutated alleles of her sisters were non-methylated, and those of her niece were partially methylated. The chromosomal and methylation study may speculate the severity of her clinical phenotypes.


Subject(s)
Codon, Nonsense , DNA Methylation , Fabry Disease/pathology , Learning Disabilities/pathology , alpha-Galactosidase/blood , Adult , Alleles , Chromosome Deletion , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Comparative Genomic Hybridization , Fabry Disease/genetics , Fabry Disease/metabolism , Facies , Female , Heterozygote , Humans , Learning Disabilities/genetics , Learning Disabilities/metabolism , Pedigree , Phenotype , Sequence Analysis, DNA , alpha-Galactosidase/genetics
3.
Tsitologiia ; 58(4): 267-71, 2016.
Article in English, Russian | MEDLINE | ID: mdl-30191692

ABSTRACT

The article is a review of the results of the study of the structural and functional organization of the human sphingomyelin synthase 1 gene (SGMS1) in Human Molecular Genetics Department of Institute of Molecular Genetics RAS. SGMS1 gene encodes an essential enzyme which is involved in the synthesis of sphingomyelin and diacylglycerol from phosphatidylcholine and ceramide, wich determines its participation in the regulation of intracellular vesicular transport, cholesterol metabolism, cell proliferation, apoptosis and other significant processes. Our research has shown that the SGMS1 gene is located on the chromosome 10, has a size of 320 kb and contains more than 20 exons. A detailed study of the SGMS1 gene's structure allowed us to identify the variety of its transcripts. mRNA isoforms with different fragments of 5R untranslated region (5R UTR) and encoding the full length protein, as well as transcripts resulting from alternative combinations of exons and containing the coding region of the gene and 3R UTR have been discovered. We have found new transcripts among the products of SGMS1 gene ­ circular RNAs, which mostly contained sequences of multi-exon 5R UTR of the gene. They are conservative and predominantly expressed in the brain. Circular RNAs of SGMS1 gene had a large number of binding sites for a microRNA that may determine the functional significance of these molecules. The review describes the latest information about the structural and functional organization of the human gene SGMS1 as well as the features of its expression.


Subject(s)
5' Untranslated Regions/physiology , Alternative Splicing/physiology , Chromosomes, Human, Pair 10 , Gene Expression Regulation, Enzymologic/physiology , Membrane Proteins , MicroRNAs , Nerve Tissue Proteins , Transferases (Other Substituted Phosphate Groups) , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Transferases (Other Substituted Phosphate Groups)/biosynthesis , Transferases (Other Substituted Phosphate Groups)/genetics
4.
Mol Oncol ; 8(2): 221-31, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24315414

ABSTRACT

The RET (REarranged during Transfection) receptor tyrosine kinase is targeted by oncogenic rearrangements in thyroid and lung adenocarcinoma. Recently, a RET (exon 12) rearrangement with FGFR1OP [fibroblast growth factor receptor 1 (FGFR1) oncogene partner] (exon 12) was identified in one chronic myelomonocytic leukemia (CMML) patient. We report the molecular cloning and functional characterization of a novel FGFR1OP (exon 11)-RET (exon 11) gene fusion event (named FGFR1OP-RET), mediated by a reciprocal translocation t(6; 10)(q27; q11), in a patient affected by primary myelofibrosis (PMF) with secondary acute myeloid leukemia (AML). The FGFR1OP-RET fusion protein displayed constitutive tyrosine kinase and transforming activity in NIH3T3 fibroblasts, and induced IL3-independent growth and activation of PI3K/STAT signaling in hematopoietic Ba/F3 cells. FGFR1OP-RET supported cytokine-independent growth, protection from stress and enhanced self-renewal of primary murine hematopoietic progenitor and stem cells in vitro. In vivo, FGFR1OP-RET caused a spectrum of disease phenotypes, with >50% of mice showing a fatal myeloproliferative disorder (MPD). Other phenotypes were leukemia transplantable in secondary recipients, dramatic expansion of the mast cell lineage, and reduction of repopulating activity upon lethal irradiation. In conclusion, FGFR1OP-RET chimeric oncogenes are endowed with leukemogenic potential and associated to myeloid neoplasms (CMML and PMF/AML).


Subject(s)
Chromosomes, Human, Pair 10 , Chromosomes, Human, Pair 6 , Hematologic Neoplasms , Oncogene Proteins, Fusion , Proto-Oncogene Proteins c-ret , Proto-Oncogene Proteins , Translocation, Genetic , Animals , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Chromosomes, Human, Pair 6/genetics , Chromosomes, Human, Pair 6/metabolism , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Mice , NIH 3T3 Cells , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism
6.
J Biol Chem ; 287(6): 3808-22, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22184110

ABSTRACT

Tuberous sclerosis complex 2 (TSC2) and phosphatase and tensin homolog deleted on chromosome 10 (PTEN) function to block growth factor-induced mammalian target of rapamycin (mTOR) signaling and are mutated in autosomal dominant hamartoma syndromes. mTOR binds to a spectrum of common and different proteins to form TOR complex 1 (TORC1) and TORC2, which regulate cell growth, division, and metabolism. TSC2 deficiency induces constitutive activation of mTOR, leading to a state of insulin resistance due to a negative feedback regulation, resulting in reduced Akt phosphorylation. We have recently described an alternative mechanism showing that in TSC2 deficiency, enhanced PTEN expression contributes to reduced Akt phosphorylation. To explore the mechanism of PTEN regulation, we used rapamycin and constitutively active mTOR to show that TORC1 increases the expression of PTEN mRNA and protein. We found that in TSC2(-/-) mouse embryonic fibroblasts expression of a kinase-dead mutant of mTOR, which inhibits both TORC1 and TORC2, decreases the expression of PTEN via transcriptional mechanism. Furthermore, kinase-dead mTOR increased and decreased phosphorylation of Akt at catalytic loop site Thr-308 and hydrophobic motif site Ser-473, respectively. Moreover, inhibition of deregulated TORC1 in TSC2-null mouse embryonic fibroblasts or in 293 cells by down-regulation of raptor decreased the levels of the transcription factor Hif1α and blocked PTEN expression, resulting in enhanced phosphorylation of Akt at Thr-308 and Ser-473. Finally, knockdown of rictor or mSin1 attenuated the expression of Hif1α, which decreased transcription of PTEN. These results unravel a previously unrecognized cell-autonomous function of TORC1 and TORC2 in the up-regulation of PTEN, which prevents phosphorylation of Akt and may shield against the development of malignancy in TSC patients.


Subject(s)
PTEN Phosphohydrolase/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Up-Regulation , Animals , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Deletion , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Tuberous Sclerosis/genetics , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
Methods Mol Biol ; 767: 177-90, 2011.
Article in English | MEDLINE | ID: mdl-21822875

ABSTRACT

Classical cytogenetics by karyotyping has been utilized in clinical research laboratories for more than 50 years and remains the key method used in the stem cell laboratory to assess the genetic stability of stem cell cultures. It is currently the most readily accessible method for detecting chromosomal abnormalities in pluripotent stem cell cultures. This chapter will describe (1) how to prepare a culture to maximize the number of metaphase cells, (2) how to prepare slides containing chromosome spreads (3) methods used to stain chromosomes, and (4) how to interpret the cytogenetic report.


Subject(s)
Cytogenetics/methods , Karyotyping/methods , Azure Stains/metabolism , Chromosome Banding , Chromosomes, Human, Pair 10/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Indoles/metabolism , Metaphase , Trypsin/metabolism
8.
Cell Cycle ; 10(17): 2967-77, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21857152

ABSTRACT

Primary autosomal recessive microcephaly (MCPH) is a congenital disorder characterized by a pronounced reduction of brain size and mental retardation. We present here a consanguineous Turkish family clinically diagnosed with MCPH and without linkage to any of the known loci (MCPH1-MCPH7). Autozygosity mapping identified a homozygous region of 15.8 Mb on chromosome 10q11.23-21.3, most likely representing a new locus for MCPH. Although we were unable to identify the underlying genetic defect after extensive molecular screening, we could delineate a possible molecular function in chromosome segregation by the characterization of mitosis in the patients' cells. Analyses of chromosome nondisjunction in T-lymphocytes and fibroblasts revealed a significantly elevated rate of nondisjunction in the patients' cells as compared to controls. Mitotic progression was further explored by immunofluorescence analyses of several chromosome and spindle associated proteins. We detected a remarkable alteration in the anaphase distribution of Aurora B and INCENP, which are key regulators of chromosome segregation. In particular, a fraction of both proteins remained abnormally loaded on chromosomes during anaphase in MCPH patients' cells while in cells of normal control subjects both proteins are completely transferred to the spindle midzone. We did not observe any other alterations regarding cell cycle progression, chromosome structure, or response to DNA damage. Our observations point towards a molecular role of the underlying gene product in the regulation of anaphase/telophase progression possibly through interaction with chromosomal passenger proteins. In addition, our findings represent further evidence for the proposed role of MCPH genes in the regulation of mitotic progression.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Microcephaly/genetics , Protein Serine-Threonine Kinases/metabolism , Adolescent , Anaphase , Aurora Kinase B , Aurora Kinases , Brain/abnormalities , Child , Child, Preschool , Chromosome Mapping , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Computational Biology , Congenital Abnormalities/pathology , Consanguinity , Female , Fluorescent Antibody Technique , Genome, Human , Humans , Male , Microcephaly/pathology , Mitosis , Pedigree , Sequence Alignment , Sequence Analysis, DNA , Turkey
9.
Int J Mol Med ; 23(3): 307-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212647

ABSTRACT

FGFR2 gene at human chromosome 10q26 encodes FGFR2b and FGFR2c isoforms functioning as FGF receptors with distinct expression domain and ligand specificity. FGFR2 plays oncogenic and anti-oncogenic roles in a context-dependent manner. Single nucleotide polymorphisms (SNPs) within intron 2 of FGFR2 gene are associated with breast cancer through allelic FGFR2 upregulation. Missense mutations or copy number gains of FGFR2 gene occur in breast cancer and gastric cancer to activate FGFR2 signaling. Aberrant FGFR2 signaling activation induces proliferation and survival of tumor cells. The class switch from FGFR2b to FGFR2c occurs during progression of prostate cancer and bladder cancer because of spliceosome dysregulation. In addition, epidermal Fgfr2b knockout mice show increased sensitivity to chemical carcinogenesis partly due to the failure of Nfe2l2 (Nrf2)-mediated detoxification of reactive oxygen species (ROS). Loss of FGFR2b signaling induces epithelial-to-mesenchymal transition (EMT) and unruly ROS. FGFR2 signaling dysregulation due to the accumulation of epigenetic modifications and genetic alterations during chronic inflammation, smoking, increased caloric uptake, and decreased exercise leads to carcinogenesis. PD173074, SU5402, AZD2171, and Ki23057 are small-molecule FGFR inhibitors. Human antibody, peptide mimetic, RNA aptamer, siRNA, and synthetic microRNA (miRNA) are emerging technologies to be applied for cancer therapeutics targeted to FGFR2. Because novel sequence technology and peta-scale super-computer are opening up the sequence era following the genome era, personalized medicine prescribing targeted drugs based on germline and/or somatic genomic information is coming reality. Application of FGFR2 inhibitors for cancer treatment in patients with FGFR2 mutation or gene amplification is beneficial; however, that for cancer prevention in people with FGFR2 risk allele might be disadvantageous due to the impediment of a cytoprotective mechanism against oxidative stress.


Subject(s)
Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Alleles , Animals , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Female , Gene Dosage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/genetics , Polymorphism, Single Nucleotide , Reactive Oxygen Species/metabolism , Risk Factors , Signal Transduction/drug effects , Signal Transduction/genetics , Spliceosomes/genetics , Spliceosomes/metabolism , Up-Regulation/drug effects
10.
Eur J Hum Genet ; 16(11): 1289-300, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18781191

ABSTRACT

The PTEN hamartoma tumor syndromes (PHTS) are a collection of rare clinical syndromes characterized by germline mutations of the tumor suppressor PTEN. These syndromes are driven by cellular overgrowth, leading to benign hamartomas in virtually any organ. Cowden syndrome (CS), the prototypic PHTS syndrome, is associated with increased susceptibility to breast, thyroid, and endometrial cancer. PTEN is located on chromosome 10q22-23 and negatively regulates the prosurvival PI3K/Akt/mTOR pathway through its lipid phosphatase activity. Loss of PTEN activates this pathway and leads to increased cellular growth, migration, proliferation, and survival. Clinical management of patients with PHTS, particularly those with CS, should include early and frequent screening, surveillance, and preventive care for associated malignancies. Concomitant with improved understanding of the biology of PTEN and the PI3K/Akt/mTOR pathway, inhibitors of this pathway are being developed as anticancer agents. These medications could have applications for patients with PHTS, for whom no medical options currently exist.


Subject(s)
Antineoplastic Agents/therapeutic use , Hamartoma Syndrome, Multiple , Mutation , PTEN Phosphohydrolase/metabolism , Cell Movement , Cell Proliferation , Cell Survival , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Female , Hamartoma Syndrome, Multiple/drug therapy , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/metabolism , Humans , Male , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases
11.
Oncogene ; 25(29): 4099-109, 2006 Jul 06.
Article in English | MEDLINE | ID: mdl-16491119

ABSTRACT

The Clathrin Assembly Lymphoid Myeloid leukemia gene (CALM or PICALM) was first identified as the fusion partner of AF10 in the t(10;11)(p13;q14) translocation, which is observed in acute myeloid leukemia (AML), acute lymphoblastic leukemia (ALL) and malignant lymphoma. The CALM/AF10 fusion protein plays a crucial role in t(10;11)(p13;q14) associated leukemogenesis. Using the N-terminal half of CALM as a bait in a yeast two-hybrid screen, a novel protein named CATS (CALM interacting protein expressed in thymus and spleen) was identified. Multiple tissue Northern blot analysis showed predominant expression of CATS in thymus, spleen and colon. CATS codes for two protein isoforms of 238 and 248 amino acids (aa). The interaction between CALM and CATS could be confirmed using pull down assays, co-immunoprecipitation and colocalization experiments. The CATS interaction domain of CALM was mapped to aa 221-335 of CALM. This domain is contained in the CALM/AF10 fusion protein. CATS localizes to the nucleus and shows a preference for nucleoli. Expression of CATS was able to markedly increase the nuclear localization of CALM and of the leukemogenic fusion protein CALM/AF10. The possible implications of these findings for CALM/AF10-mediated leukemogenesis are discussed.


Subject(s)
Active Transport, Cell Nucleus , Carrier Proteins/metabolism , Cell Nucleolus/metabolism , Monomeric Clathrin Assembly Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , 3T3 Cells , Active Transport, Cell Nucleus/genetics , Animals , Base Sequence , Carrier Proteins/genetics , Cell Nucleolus/genetics , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 10/metabolism , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 13/metabolism , Gene Expression Regulation/genetics , Humans , Intracellular Signaling Peptides and Proteins , Mice , Molecular Sequence Data , Monomeric Clathrin Assembly Proteins/genetics , Nuclear Proteins , Oncogene Proteins, Fusion/genetics , Organ Specificity , Protein Binding/genetics , Translocation, Genetic/genetics
12.
Dev Cell ; 8(6): 949-61, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15935783

ABSTRACT

REC8 is a key component of the meiotic cohesin complex. During meiosis, cohesin is required for the establishment and maintenance of sister-chromatid cohesion, for the formation of the synaptonemal complex, and for recombination between homologous chromosomes. We show that REC8 has an essential role in mammalian meiosis, in that Rec8 null mice of both sexes have germ cell failure and are sterile. In the absence of REC8, early chromosome pairing events appear normal, but synapsis occurs in a novel fashion: between sister chromatids. This implies that a major role for REC8 in mammalian meiosis is to limit synapsis to between homologous chromosomes. In all other eukaryotic species studied to date, REC8 phenotypes have been restricted to meiosis. Unexpectedly, Rec8 null mice are born in sub-Mendelian frequencies and fail to thrive. These findings illuminate hitherto unknown REC8 functions in chromosome dynamics during mammalian meiosis and possibly in somatic development.


Subject(s)
Acetaminophen/analogs & derivatives , Chromatids/metabolism , Chromosome Pairing/physiology , Meiosis/physiology , Nuclear Proteins/deficiency , Nuclear Proteins/physiology , Phosphoproteins/deficiency , Phosphoproteins/physiology , Saccharin/analogs & derivatives , Acetaminophen/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Death/physiology , Chondroitin Sulfate Proteoglycans/metabolism , Chromatids/ultrastructure , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Painting/methods , Chromosomes/metabolism , Chromosomes/ultrastructure , Chromosomes, Human, Pair 10/metabolism , Cloning, Molecular/methods , DNA-Binding Proteins/metabolism , Electroporation/methods , Female , Humans , Immunohistochemistry/methods , In Situ Nick-End Labeling/methods , Indoles/metabolism , Male , Meiotic Prophase I/physiology , Mice , Mice, Knockout , Microscopy, Electron, Transmission/methods , Models, Biological , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oncorhynchus kisutch/metabolism , Ovary/metabolism , Pachytene Stage/physiology , Phosphate-Binding Proteins , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rad51 Recombinase , Saccharin/metabolism , Spermatogenesis/genetics , Testis/metabolism , Testis/ultrastructure , Trans-Activators/metabolism
13.
Hum Mol Genet ; 9(19): 2879-84, 2000 Nov 22.
Article in English | MEDLINE | ID: mdl-11092764

ABSTRACT

Chromosomal rearrangements occur more frequently in subtelomeric domains than in other regions of the genome and are often associated with human pathology. To further elucidate the plasticity of subtelomeric domains, we examined the 3.3 kb D4Z4 repeat array on chromosome 4 and its homologue on chromosome 10 in 208 Dutch blood donors by pulsed field gel electrophoresis. These subtelomeric repeats are known to rearrange and partial deletions of this polymorphic array on chromosome 4 are associated with facioscapulohumeral muscular dystrophy (FSHD), an autosomal dominant myopathy. Our results show that mitotic rearrangements occur frequently as 3% of individuals display somatic mosaicism for a repeat expansion or contraction explaining the high variability of subtelomeric repeat array sizes. Translocated 4-type repeat arrays on chromosome 10 and the reverse configuration of 10-type repeat arrays on chromosome 4 are observed in 21% of individuals. The translocated repeat arrays on chromosome 4 tend to be more heterogeneous than 4-type repeats on chromosome 10. The repeat length on chromosome 4 is on average larger than on chromosome 10. But on both chromosomes we observe a multi-modal repeat length distribution with equidistant peaks at intervals of 65 kb, possibly reflecting a higher-order chromatin structure. Interestingly, in as many as six random blood donors (3%) we identified FSHD-sized 4-type repeat arrays. Assuming that these individuals are clinically unaffected, these results imply an incomplete penetrance in the upper range of FSHD alleles. Overall, the observed dynamic characteristics of these homologous domains may serve as a model for subtelomeric plasticity.


Subject(s)
Chromosomes, Human, Pair 10/metabolism , Chromosomes, Human, Pair 4/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Repetitive Sequences, Nucleic Acid/genetics , Telomere/genetics , Translocation, Genetic/genetics , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 4/genetics , Electrophoresis, Gel, Pulsed-Field , Female , Humans , Male , Mitosis/genetics , Mosaicism/genetics , Netherlands , Nucleic Acid Hybridization
14.
Hum Mol Genet ; 9(2): 175-85, 2000 Jan 22.
Article in English | MEDLINE | ID: mdl-10607828

ABSTRACT

Using combined immunofluorescence and fluorescence in situ hybridization (FISH) analysis we have extensively characterized the proteins associating with two different homologue human neocentromeres at interphase and prometaphase/metaphase, and compared these directly with those found with normal human centromeres. Antisera to CENP-A, CENP-B, CENP-C, CENP-E, CENP-F, INCENP, CLIP-170, dynein, dynactin subunits p150 (Glued) and Arp1, MCAK, Tsg24, p55CDC, HZW10, HBUB1, HBUBR1, BUB3, MAD2, ERK1, 3F3/2, topoisomerase II and a murine HP1 homologue, M31, were used in immuno-fluorescence experiments in conjunction with FISH employing specific DNA probes to clearly identify neocentromeric DNA. We found that except for the total absence of CENP-B binding, neocentromeres are indistinguishable from their alpha satellite-containing counterparts in terms of protein composition and distribution. This suggests that the DNA base of a potential centromeric locus is of minimal importance in determining the overall structure of a functional kinetochore and that, once seeded, the events leading to functional kinetochore formation occur independently of primary DNA sequence.


Subject(s)
Centromere/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Kinetochores/metabolism , Anaphase/genetics , Animals , CHO Cells , Cell Cycle/genetics , Cell Line, Transformed , Centromere/immunology , Centromere/physiology , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/immunology , Chromosomal Proteins, Non-Histone/physiology , Chromosomes, Human, Pair 10/metabolism , Chromosomes, Human, Pair 20/metabolism , Cricetinae , Humans , Immune Sera/metabolism , Kinetochores/physiology , Metaphase/genetics , Microtubule-Associated Proteins/metabolism , Protein Binding/genetics , Tumor Cells, Cultured
15.
Cancer ; 81(5): 299-308, 1997 Oct 25.
Article in English | MEDLINE | ID: mdl-9349518

ABSTRACT

BACKGROUND: Benign body cavity effusions sometimes cannot be distinguished from malignant ones by conventional cytology. The authors performed fluorescence in situ hybridization (FISH) on ThinPrep slides using chromosome specific probes to see if hyperdiploid malignant cells could be detected in 20 body cavity effusions. The results were then compared with those of conventional cytology. METHODS: A total of 20 body cavity effusions from 19 patients were studied using conventional cytology and FISH. Probes specific for chromosomes 3, 8, 10, and 12 were used to detect hyperdiploidy on ThinPrep slides (Cytyc Corporation, Boxborough, MA). RESULTS: A total of 13 patients had malignant conditions (either prior history of malignancy or the presence of malignancy anywhere in the body). Conventional cytology and FISH were both positive in 5 of these patients (6 samples) and negative in 2 patients. The results for one sample were inconclusive by both methods. There were 5 discrepant cytology-FISH results in patients with malignant conditions. One sample was positive by FISH and negative by cytology, one was positive by FISH and "atypical" by cytology, and three were inconclusive by FISH and negative by cytology. FISH results were either negative (in 4 samples) or inconclusive (in 2 samples) in the 6 patients with benign conditions. CONCLUSIONS: FISH can detect hyperdiploid malignant cells in body cavity effusions and is especially useful when the major cell population consists of malignant cells that cannot be differentiated from mesothelial or "atypical" cells. It is less useful in detecting a small population of malignant cells hidden in an inflammatory or reactive cell background. More studies are needed to establish diagnostic criteria further and to assess the clinical usefulness of this procedure.


Subject(s)
Ascitic Fluid/pathology , Diploidy , Neoplasms/pathology , Pericardial Effusion/pathology , Pleural Effusion, Malignant/pathology , Adult , Aged , Aged, 80 and over , Ascitic Fluid/metabolism , Chromosomes, Human, Pair 10/metabolism , Chromosomes, Human, Pair 12/metabolism , Chromosomes, Human, Pair 3/metabolism , Chromosomes, Human, Pair 8/metabolism , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Neoplasms/metabolism , Pericardial Effusion/metabolism , Pleural Effusion, Malignant/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...