Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 157
Filter
1.
Oncogene ; 31(25): 3039-50, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22020333

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive brain tumor for which there is no cure. Overexpression of wild-type epidermal growth factor receptor (EGFR) and loss of the tumor suppressor genes Ink4a/Arf and PTEN are salient features of this deadly cancer. Surprisingly, targeted inhibition of EGFR has been clinically disappointing, demonstrating an innate ability for GBM to develop resistance. Efforts at modeling GBM in mice using wild-type EGFR have proven unsuccessful to date, hampering endeavors at understanding molecular mechanisms of therapeutic resistance. Here, we describe a unique genetically engineered mouse model of EGFR-driven gliomagenesis that uses a somatic conditional overexpression and chronic activation of wild-type EGFR in cooperation with deletions in the Ink4a/Arf and PTEN genes in adult brains. Using this model, we establish that chronic activation of wild-type EGFR with a ligand is necessary for generating tumors with histopathological and molecular characteristics of GBMs. We show that these GBMs are resistant to EGFR kinase inhibition and we define this resistance molecularly. Inhibition of EGFR kinase activity using tyrosine kinase inhibitors in GBM tumor cells generates a cytostatic response characterized by a cell cycle arrest, which is accompanied by a substantial change in global gene expression levels. We demonstrate that an important component of this pattern is the transcriptional activation of the MET receptor tyrosine kinase and that pharmacological inhibition of MET overcomes the resistance to EGFR inhibition in these cells. These findings provide important new insights into mechanisms of resistance to EGFR inhibition and suggest that inhibition of multiple targets will be necessary to provide therapeutic benefit for GBM patients.


Subject(s)
Disease Models, Animal , ErbB Receptors/genetics , Glioblastoma/genetics , Mice , Proto-Oncogene Proteins c-met/genetics , Animals , ErbB Receptors/antagonists & inhibitors , Genes, Tumor Suppressor , Glioblastoma/physiopathology , Humans , Mice, Transgenic
2.
J Hum Hypertens ; 23(10): 636-44, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19194457

ABSTRACT

Increased arterial stiffness and wave reflection have been identified as cardiovascular disease risk factors. In light of significant sex differences and the moderate heritability of vascular function measures, we hypothesized that variation in the genes coding for oestrogen receptors alpha (ESR1) and beta (ESR2) and aromatase (CYP19A1) is associated with aortic stiffness and pressure wave reflection as measured by non-invasive arterial tonometry. In all, 1261 unrelated Framingham Offspring Study participants who attended the seventh examination cycle (mean age 62+/-10 years, 52% women) and had arterial tonometry and genotyping data were included in the study. Analysis of covariance was used to assess the association of polymorphisms with forward wave amplitude, augmented pressure, augmentation index (AI), carotid-femoral pulse wave velocity and mean arterial pressure with adjustment for potential confounders. In the sex-pooled analysis, those homozygous for the minor allele at any of four ESR1 variants that were in strong linkage disequilibrium ((TA)(n), rs2077647, rs2234693 and rs9340799) had on an average 18% higher augmented pressure and 16% greater AI compared with carriers of one or two major alleles (P=0.0002-0.01). A similar magnitude of association was detected in those homozygous for the common allele at two ESR2 single-nucleotide polymorphisms (P=0.007-0.02). Our results are consistent with the hypothesis that variation in ESR1 and ESR2, but not CYP19A1, is associated with an increased wave reflection that may contribute to associations between these variants and adverse clinical events demonstrated earlier. Our findings will need to be replicated in additional cohorts.


Subject(s)
Aromatase/genetics , Arteries/physiopathology , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Peripheral Vascular Diseases/genetics , Polymorphism, Single Nucleotide , Aged , Arteries/diagnostic imaging , Blood Pressure , Brachial Artery/physiopathology , Carotid Arteries/physiopathology , Elasticity , Female , Femoral Artery/physiopathology , Gene Frequency , Genetic Association Studies , Genetic Predisposition to Disease , Homozygote , Humans , Linkage Disequilibrium , Male , Manometry , Middle Aged , Peripheral Vascular Diseases/diagnostic imaging , Peripheral Vascular Diseases/physiopathology , Phenotype , Pulsatile Flow , Ultrasonography, Doppler
3.
Am J Med Genet B Neuropsychiatr Genet ; 150B(3): 425-9, 2009 Apr 05.
Article in English | MEDLINE | ID: mdl-18712713

ABSTRACT

The instability of the CAG repeat size of the HD gene when transmitted intergenerationally has critical implications for genetic counseling practices. In particular, CAG repeats between 27 and 35 have been the subject of debate based on small samples. To address this issue, we analyzed allelic instability in the Venezuelan HD kindreds, the largest and most informative families ascertained for HD. We identified 647 transmissions. Our results indicate that repeats in the 27-35 CAG range are highly stable. Out of 69 transmitted alleles in this range, none expand into any penetrant ranges. Contrastingly, 14% of alleles transmitted from the incompletely penetrant range (36-39 CAGs) expand into the completely penetrant range, characterized by alleles with 40 or more CAG repeats. At least 12 of the 534 transmissions from the completely penetrant range contract into the incompletely penetrant range of 36-39 CAG repeats. In these kindreds, none of the individuals with 27-39 CAGs were symptomatic, even though they ranged in age from 11 to 82 years. We expect these findings to be helpful in updating genetic counseling practices.


Subject(s)
Family , Genetic Counseling , Huntington Disease/genetics , Trinucleotide Repeat Expansion , Adolescent , Adult , Age of Onset , Aged , Aged, 80 and over , Alleles , Child , Female , Humans , Huntingtin Protein , Male , Middle Aged , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Penetrance , Venezuela , Young Adult
4.
J Med Genet ; 44(1): 44-50, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17018562

ABSTRACT

BACKGROUND: The major determinant of age of onset in Huntington's disease is the length of the causative triplet CAG repeat. Significant variance remains, however, in residual age of onset even after repeat length is factored out. Many genetic polymorphisms have previously shown evidence of association with age of onset of Huntington's disease in several different populations. OBJECTIVE: To replicate these genetic association tests in 443 affected people from a large set of kindreds from Venezuela. METHODS: Previously tested polymorphisms were analysed in the HD gene itself (HD), the GluR6 kainate glutamate receptor (GRIK2), apolipoprotein E (APOE), the transcriptional coactivator CA150 (TCERG1), the ubiquitin carboxy-terminal hydrolase L1 (UCHL1), p53 (TP53), caspase-activated DNase (DFFB), and the NR2A and NR2B glutamate receptor subunits (GRIN2A, GRIN2B). RESULTS: The GRIN2A single-nucleotide polymorphism explains a small but considerable amount of additional variance in residual age of onset in our sample. The TCERG1 microsatellite shows a trend towards association but does not reach statistical significance, perhaps because of the uninformative nature of the polymorphism caused by extreme allele frequencies. We did not replicate the genetic association of any of the other genes. CONCLUSIONS: GRIN2A and TCERG1 may show true association with residual age of onset for Huntington's disease. The most surprising negative result is for the GRIK2 (TAA)(n) polymorphism, which has previously shown association with age of onset in four independent populations with Huntington's disease. The lack of association in the Venezuelan kindreds may be due to the extremely low frequency of the key (TAA)(16) allele in this population.


Subject(s)
Huntington Disease/epidemiology , Huntington Disease/genetics , Polymorphism, Single Nucleotide , Receptors, N-Methyl-D-Aspartate/genetics , Trans-Activators/genetics , Age of Onset , Apolipoproteins E/genetics , Deoxyribonucleases/genetics , Gene Frequency , Humans , Huntingtin Protein , Microsatellite Repeats , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Poly-ADP-Ribose Binding Proteins , Receptors, Kainic Acid/genetics , Transcriptional Elongation Factors , Trinucleotide Repeat Expansion/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin Thiolesterase/genetics , Venezuela , GluK2 Kainate Receptor
5.
Nature ; 413(6857): 739-43, 2001 Oct 18.
Article in English | MEDLINE | ID: mdl-11607033

ABSTRACT

Proteins with expanded polyglutamine repeats cause Huntington's disease and other neurodegenerative diseases. Transcriptional dysregulation and loss of function of transcriptional co-activator proteins have been implicated in the pathogenesis of these diseases. Huntington's disease is caused by expansion of a repeated sequence of the amino acid glutamine in the abnormal protein huntingtin (Htt). Here we show that the polyglutamine-containing domain of Htt, Htt exon 1 protein (Httex1p), directly binds the acetyltransferase domains of two distinct proteins: CREB-binding protein (CBP) and p300/CBP-associated factor (P/CAF). In cell-free assays, Httex1p also inhibits the acetyltransferase activity of at least three enzymes: p300, P/CAF and CBP. Expression of Httex1p in cultured cells reduces the level of the acetylated histones H3 and H4, and this reduction can be reversed by administering inhibitors of histone deacetylase (HDAC). In vivo, HDAC inhibitors arrest ongoing progressive neuronal degeneration induced by polyglutamine repeat expansion, and they reduce lethality in two Drosophila models of polyglutamine disease. These findings raise the possibility that therapy with HDAC inhibitors may slow or prevent the progressive neurodegeneration seen in Huntington's disease and other polyglutamine-repeat diseases, even after the onset of symptoms.


Subject(s)
Enzyme Inhibitors/metabolism , Glutamine/metabolism , Histone Deacetylase Inhibitors , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Nuclear Proteins/metabolism , Peptides/metabolism , Saccharomyces cerevisiae Proteins , Trans-Activators/metabolism , Acetylation , Acetyltransferases/metabolism , Animals , Animals, Genetically Modified , CREB-Binding Protein , Disease Models, Animal , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , E1A-Associated p300 Protein , Gene Expression Regulation , Glutathione Transferase/metabolism , Histone Acetyltransferases , Histone Deacetylases/metabolism , Histones/metabolism , Huntingtin Protein , Huntington Disease/enzymology , Huntington Disease/metabolism , Huntington Disease/prevention & control , Nerve Degeneration , Nerve Tissue Proteins/chemistry , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/enzymology , Nuclear Proteins/chemistry , PC12 Cells , Protein Structure, Tertiary , Rats , Repetitive Sequences, Amino Acid , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sin3 Histone Deacetylase and Corepressor Complex
6.
J Comp Neurol ; 437(4): 398-407, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11503142

ABSTRACT

CalDAG-GEFI and CalDAG-GEFII (identical to RasGRP) are novel, brain-enriched guanine nucleotide exchange factors (GEFs) that can be stimulated by calcium and diacylglycerol and that can activate small GTPases, including Ras and Rap1, molecules increasingly recognized as having signaling functions in neurons. Here, we show that CalDAG-GEFI and CalDAG-GEFII mRNAs, detected by in situ hybridization analysis, have sharply contrasting forebrain-predominant distributions in the mature brain: CalDAG-GEFI is expressed mainly in the striatum and olfactory structures and deep cortical layers, whereas CalDAG-GEFII is expressed widely in the forebrain. Within the striatum, however, the two CalDAG-GEF mRNAs have nearly identical distributions: they are coexpressed in striatal projection neurons that give rise to the direct and indirect pathways of the basal ganglia. Subcellular fractionation analysis of the substantia nigra with monoclonal antibodies against CalDAG-GEFI suggests that CalDAG-GEFI protein is present not only in the cell bodies of striatal projection neurons but also in their axons and axon terminals. These results suggest that the CalDAG-GEFs may be key intracellular regulators whereby calcium and diacylglycerol signals can regulate cellular functions through small GTPases in the basal ganglia circuits.


Subject(s)
Corpus Striatum/cytology , Guanine Nucleotide Exchange Factors/analysis , Nerve Tissue Proteins/analysis , Neurons/chemistry , Age Factors , Animals , Basal Ganglia/metabolism , Blotting, Western , Corpus Striatum/embryology , Corpus Striatum/growth & development , Corpus Striatum/metabolism , Efferent Pathways/chemistry , Electrophoresis, Polyacrylamide Gel , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/biosynthesis , Guanine Nucleotide Exchange Factors/genetics , In Situ Hybridization , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Rats , Rats, Sprague-Dawley , Subcellular Fractions/chemistry , rap1 GTP-Binding Proteins/physiology , ras Guanine Nucleotide Exchange Factors , ras Proteins/physiology
7.
J Biol Chem ; 276(31): 29456-65, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11384996

ABSTRACT

PDZ domains are involved in the scaffolding and assembly of multi-protein complexes at various subcellular sites. We describe here the isolation and characterization of a novel PDZ domain-containing protein that localizes to the Golgi apparatus. Using an in silico cloning approach, we have identified and isolated a cDNA encoding a ubiquitously expressed 59-kDa protein that we call FIG. It is composed of two coiled coil regions, a leucine zipper, and a single PDZ domain. Cytological studies using indirect immunofluorescence microscopy revealed that FIG is a peripheral protein that uses one of its coiled coil domains to localize to the Golgi apparatus. To ascertain the modalities of this Golgi localization, the same coiled coil region was tested for its ability to interact with a panel of coiled coil domain-containing integral membrane Golgi proteins. Using a series of GST fusion protein binding assays, co-immunofluorescence and co-immunoprecipitation experiments, we show that FIG specifically binds to the coiled coil domain-containing Q-SNARE (Q-soluble NSF attachment protein receptor) protein syntaxin 6 both in vitro and in vivo. The structural features of FIG and its interaction with a SNARE protein suggest that FIG may play a role in membrane vesicle trafficking. This is the first example of a PDZ domain-containing peripheral protein that localizes to the Golgi through a coiled coil-mediated interaction with a resident membrane protein. Our results broaden the scope of PDZ domain-mediated functions.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Transcription, Genetic , Vesicular Transport Proteins , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Amino Acid Substitution , Animals , Base Sequence , Binding Sites , Carrier Proteins/chemistry , Cell Line , DNA, Complementary , Evolution, Molecular , Female , Golgi Matrix Proteins , Humans , Male , Membrane Proteins/chemistry , Membrane Transport Proteins , Molecular Sequence Data , Molecular Weight , Mutagenesis, Site-Directed , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Organ Specificity , Phylogeny , Protein Biosynthesis , Protein Conformation , Qa-SNARE Proteins , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , SNARE Proteins , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
8.
Cancer Epidemiol Biomarkers Prev ; 10(6): 711-4, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11401924

ABSTRACT

UDP-glucuronosyltransferases (UGTs) catalyze the detoxification and the elimination of a large number of endogenous and exogenous compounds in the liver and extrahepatic tissues. One of the UGT1A family members, UGT1A1, is involved in estradiol metabolism and, therefore, represents a candidate gene in breast carcinogenesis. A common insertion/deletion polymorphism in the TATA-box of the promoter region of UGT1A1 results in decreased initiation of transcription. In a previous study, we found a positive association between the UGT1A1 low-transcriptional alleles and premenopausal breast cancer risk in an African-American population. In the present study, we sought to determine whether the low-transcription UGT1A1 promoter allele, UGT1A1*28 [A(TA)(7)TAA], was associated with increased breast cancer risk among primarily Caucasian women in a nested case-control study within the Nurses' Health Study cohort. No significant association between the UGT1A1*28 [A(TA)(7)TAA] allele and breast cancer was observed. Compared with women homozygous for the UGT1A1*1 [A(TA)(6)TAA] allele, the relative risk was 0.80 (confidence interval, 0.49-1.29) for women homozygous for the UGT1A1*28 allele. The effect of the UGT1A1 genotype on plasma hormone levels in postmenopausal women not using hormone replacement was also evaluated, and overall, no significant differences in hormone levels by genotypes were observed. When restricted to women who had at least one UGT1A1*28 allele and a body mass index at blood draw of >27 kg/m(2), particularly in combination with the cytochrome p450c17alpha genotype, estrone and estradiol levels tended to vary by UGT1A1 genotypes. The results presented do not support a strong association between the UGT1A1 promoter polymorphism and the risk of breast cancer.


Subject(s)
Breast Neoplasms/genetics , Estradiol/metabolism , Glucuronosyltransferase/genetics , Polymorphism, Genetic , Adult , Breast Neoplasms/pathology , Case-Control Studies , Estradiol/blood , Female , Genotype , Humans , Middle Aged , Promoter Regions, Genetic , Risk Factors , TATA Box/genetics , Transcriptional Activation
9.
Proc Natl Acad Sci U S A ; 98(8): 4764-9, 2001 Apr 10.
Article in English | MEDLINE | ID: mdl-11296304

ABSTRACT

This investigation was pursued to test the use of intracellular antibodies (intrabodies) as a means of blocking the pathogenesis of Huntington's disease (HD). HD is characterized by abnormally elongated polyglutamine near the N terminus of the huntingtin protein, which induces pathological protein-protein interactions and aggregate formation by huntingtin or its exon 1-containing fragments. Selection from a large human phage display library yielded a single-chain Fv (sFv) antibody specific for the 17 N-terminal residues of huntingtin, adjacent to the polyglutamine in HD exon 1. This anti-huntingtin sFv intrabody was tested in a cellular model of the disease in which huntingtin exon 1 had been fused to green fluorescent protein (GFP). Expression of expanded repeat HD-polyQ-GFP in transfected cells shows perinuclear aggregation similar to human HD pathology, which worsens with increasing polyglutamine length; the number of aggregates in these transfected cells provided a quantifiable model of HD for this study. Coexpression of anti-huntingtin sFv intrabodies with the abnormal huntingtin-GFP fusion protein dramatically reduced the number of aggregates, compared with controls lacking the intrabody. Anti-huntingtin sFv fused with a nuclear localization signal retargeted huntingtin analogues to cell nuclei, providing further evidence of the anti-huntingtin sFv specificity and of its capacity to redirect the subcellular localization of exon 1. This study suggests that intrabody-mediated modulation of abnormal neuronal proteins may contribute to the treatment of neurodegenerative diseases such as HD, Alzheimer's, Parkinson's, prion disease, and the spinocerebellar ataxias.


Subject(s)
Huntington Disease/pathology , Immunoglobulin Variable Region/immunology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , COS Cells , Green Fluorescent Proteins , Humans , Huntingtin Protein , Luminescent Proteins/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/immunology , Nuclear Proteins/immunology , Protein Binding
10.
Proc Natl Acad Sci U S A ; 97(26): 14530-5, 2000 Dec 19.
Article in English | MEDLINE | ID: mdl-11114193

ABSTRACT

Reactive oxygen species (ROS) are toxic compounds produced by normal metabolic processes. Their reactivity with cellular components is a major stress for aerobic cells that results in lipid, protein, and DNA damage. ROS-mediated DNA damage contributes to spontaneous mutagenesis, and cells deficient in repair and protective mechanisms have elevated levels of spontaneous mutations. In Escherichia coli a large number of genes are involved in the repair of oxidative DNA damage and its prevention by detoxification of ROS. In humans, the genes required for these processes are not well defined. In this report we describe the human OXR1 (oxidation resistance) gene discovered in a search for human genes that function in protection against oxidative damage. OXR1 is a member of a conserved family of genes found in eukaryotes but not in prokaryotes. We also outline the procedures developed to identify human genes involved in the prevention and repair of oxidative damage that were used to identify the human OXR1 gene. This procedure makes use of the spontaneous mutator phenotype of E. coli oxidative repair-deficient mutants and identifies genes of interest by screening for antimutator activity resulting from cDNA expression.


Subject(s)
Proteins/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary , Eukaryotic Cells , Humans , Hydrogen Peroxide/pharmacology , Mitochondrial Proteins , Molecular Sequence Data , Oxidation-Reduction , Proteins/physiology , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Sequence Analysis, DNA
11.
Mol Psychiatry ; 5(6): 650-3, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11126395

ABSTRACT

Despite considerable effort to identify susceptibility loci for schizophrenia, none have been localized. Multiple genome scans and collaborative efforts have shown evidence for linkage to regions on chromosomes 1q, 5q, 6q, 8p, 13q, 10p and 22q.(1-9) Heterogeneity is likely. We previously mapped schizophrenia susceptibility loci (SSL) to chromosomes 13q32 (P = 0.00002) and 8p21-22 (P= 0.0001) using 54 multiplex pedigrees and suggested linkage heterogeneity. We have now stratified these families based on co-segregating phenotypes in non-schizophrenic first degree relatives (schizophrenia spectrum personality disorders (SSPD); psychotic affective disorders (PAD)). Genome scans were conducted for these phenotypic subgroups of families and broadened affected phenotypes were tested. The SSPD group provided its strongest genome-wide linkage support for the chromosome 8p21 region (D8S1771) using either narrow (non-parametric lod (NPL) P= 0.000002) or broadened phenotypes (NPL P = 0.0000008) and a new region of interest on 1p was identified (P = 0.006). For PAD families, the peak NPL in the genome scan occurred on chromosome 3p26-p24 (P = 0.008). The identification of multiple susceptibility loci for schizophrenia may be enhanced by stratification of families using psychiatric diagnoses of the non-schizophrenic relatives.


Subject(s)
Genetic Heterogeneity , Genetic Linkage , Schizophrenia/genetics , Family Health , Genome, Human , Humans , Phenotype
12.
Pharmacogenetics ; 10(7): 629-44, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11037804

ABSTRACT

One of the most important mechanisms involved in host defense against xenobiotic chemicals and endogenous toxins is the glucuronidation catalysed by UDP-glucuronosyltransferase enzymes (UGT). The role of genetic factors in determining variable rates of glucuronidation is not well understood, but phenotypic evidence in support of such variation has been reported. In the present study, six single nucleotide polymorphisms were discovered in the first exon of the UGT1A7 gene, which codes for the putative substrate-binding domain, revealing a high structural heterogeneity at the UGT1 gene locus. The new UGT1A7 proteins differ in their primary structure at amino acid positions 129, 131 and 208, creating four distinct UGT1A7 allelic variants in the human population: UGT1A7*1 (N129 R131 W208), *2 (K129 K131 W208), *3 (K129 K131 R208), and *4 (N129 R131 R208). In functional studies, HEK cells stably transfected to express the four allelic UGT1A7 variants exhibited significant differences in catalytic activity towards 3-, 7-, and 9-hydroxy-benzo(a)pyrene. UGT1A7*3 exhibited a 5.8-fold lower relative Vmax compared to wild-type *1, whereas *2 and *4 had a 2.6- and 2.8-fold lower relative Vmax than *1, respectively, suggesting that these mutations confer slow glucuronidation phenotype. Kinetic characterization suggested that these differences were primarily attributable to altered Vmax. Additionally, it suggested that each amino acid substitutions can independently affect the UGT1A7 catalytic activity, and that their effects are additive. The expression pattern of UGT1A7 studied herein and its catalytic activity profile suggest a possible role of UGT1A7 in the detoxification and elimination of carcinogenic products in lung. A population study demonstrated that a considerable proportion of the population (15.3%) was found homozygous for the low activity allele containing all three missense mutations, UGT1A7*3. These findings suggest that further studies are needed to investigate the impact of the low UGT1A7 conjugator genotype on individual susceptibility to chemical-induced diseases and responses to therapeutic drugs.


Subject(s)
Glucuronosyltransferase/genetics , Mutation, Missense , Base Sequence , Cell Line, Transformed , DNA Primers , Female , Genotype , Glucuronosyltransferase/metabolism , Humans , Kinetics , Male , Pedigree , Polymerase Chain Reaction
13.
Nat Biotechnol ; 18(7): 760-3, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10888845

ABSTRACT

We have implemented a method for multiplexed detection of polymorphic sites and direct determination of haplotypes in 10-kilobase-size DNA fragments using single-walled carbon nanotube (SWNT) atomic force microscopy (AFM) probes. Labeled oligonucleotides are hybridized specifically to complementary target sequences in template DNA, and the positions of the tagged sequences are detected by direct SWNT tip imaging. We demonstrated this concept by detecting streptavidin and IRD800 labels at two different sequences in M13mp18. Our approach also permits haplotype determination from simple visual inspection of AFM images of individual DNA molecules, which we have done on UGT1A7, a gene under study as a cancer risk factor. The haplotypes of individuals heterozygous at two critical loci, which together influence cancer risk, can be easily and directly distinguished from AFM images. The application of this technique to haplotyping in population-based genetic disease studies and other genomic screening problems is discussed.


Subject(s)
Haplotypes , Microscopy, Atomic Force/methods , Sequence Analysis, DNA/methods , Alleles , Bacteriophage M13/genetics , Genetic Markers , Genotype , Glucuronosyltransferase/genetics , Heterozygote , Humans , Microscopy, Atomic Force/instrumentation , Nucleic Acid Hybridization , Physical Chromosome Mapping , Plasmids/genetics , Polymorphism, Single Nucleotide , Streptavidin/genetics
14.
Proc Natl Acad Sci U S A ; 97(12): 6763-8, 2000 Jun 06.
Article in English | MEDLINE | ID: mdl-10823891

ABSTRACT

Huntington's Disease (HD) is caused by an expansion of a polyglutamine tract within the huntingtin (htt) protein. Pathogenesis in HD appears to include the cytoplasmic cleavage of htt and release of an amino-terminal fragment capable of nuclear localization. We have investigated potential consequences to nuclear function of a pathogenic amino-terminal region of htt (httex1p) including aggregation, protein-protein interactions, and transcription. httex1p was found to coaggregate with p53 in inclusions generated in cell culture and to interact with p53 in vitro and in cell culture. Expanded httex1p represses transcription of the p53-regulated promoters, p21(WAF1/CIP1) and MDR-1. httex1p was also found to interact in vitro with CREB-binding protein (CBP) and mSin3a, and CBP to localize to neuronal intranuclear inclusions in a transgenic mouse model of HD. These results raise the possibility that expanded repeat htt causes aberrant transcriptional regulation through its interaction with cellular transcription factors which may result in neuronal dysfunction and cell death in HD.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Huntington Disease/genetics , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Tumor Suppressor Protein p53/physiology , Animals , Cells, Cultured , Humans , Huntingtin Protein , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic
15.
Hum Mol Genet ; 9(9): 1315-20, 2000 May 22.
Article in English | MEDLINE | ID: mdl-10814713

ABSTRACT

Some studies show that plasma triglyceride (TG) levels are a significant independent risk factor for cardiovascular disease (CVD). TG levels are inversely correlated with high density lipoprotein cholesterol (HDL-C) levels, and their metabolism may be closely interrelated. Therefore, the TG/HDL-C ratio may be a relevant CVD risk factor. Our analysis of families in the Framingham Heart Study gave a genetic heritability estimate for log(TG) of 0.40 and for log(TG/HDL-C) of 0.49, demonstrating an important genetic component for both. A 10 cM genome-wide scan for log(TG) level and log(TG/HDL-C) was carried out for the largest 332 extended families of the Framingham Heart Study (1702 genotyped individuals). The highest multipoint variance component LOD scores obtained for both log(TG) and log(TG/HDL-C) were on chromosome 7 (at 155 cM), where the results for the two phenotypes were 1.8 and 2.5, respectively. The 7q32.3-qter region contains several candidate genes. Four other regions with multipoint LOD scores greater than one were identified on chromosome 3 [LOD score for log(TG/HDL-C) = 1.8 at 140 cM], chromosome 11 [LOD score for log(TG/HDL-C) = 1.1 at 125 cM], chromosome 16 [LOD score for log(TG) = 1.5 at 70 cM, LOD score for log(TG/HDL-C) = 1.1 at 75 cM] and chromosome 20 [LOD score for log(TG/HDL-C) = 1.7 at 35 cM, LOD score for log(TG) = 1.3 at 40 cM]. These results identify loci worthy of further study.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , Cholesterol, HDL/blood , Cholesterol, HDL/genetics , Chromosomes, Human, Pair 7 , Triglycerides/blood , Triglycerides/genetics , Aged , Aged, 80 and over , Cohort Studies , Female , Genetic Linkage , Genome, Human , Humans , Lod Score , Male , Middle Aged , Phenotype
16.
Cancer Res ; 60(4): 950-6, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706110

ABSTRACT

We examined the role of constitutional genetic variation at the UDP-glucuronosyltransferase (UGT) 1A1 locus in breast cancer susceptibility. The UGT1A1 enzyme is a major UGT involved in estradiol glucuronidation. To date, four UGT1A1 variant alleles characterized by a variation in the number of TA from five through eight repeats in the atypical TATA box region have been described in the African-American population. Functional analyses of the transcriptional activity in breast and liver cells revealed that the transcription activation of a reporter gene is inversely correlated with the number of repeats. Reverse transcription-PCR analysis confirmed the expression of UGT1A1 in human liver in the hepatocarcinoma cell line HepG2 and provided evidence of the expression of UGT1A1 in breast cancer tissue, where a positive signal was observed in 11 of 12 breast cancer cell lines tested. The population-based case-control study involved 200 women with breast cancer and 200 female controls of African ancestry. We postulated that breast cancer cases might have a higher prevalence of low activity allele-containing genotypes than controls (alleles presenting seven and eight repeats in the A(TA)nTAA motif of the TATA box). The age-adjusted odds ratio (OR) for breast cancer comparing women with seven and eight allele-containing genotypes versus 5/5, 5/6, and 6/6 genotypes was 1.8 [95% confidence interval (CI), 1.0-3.1; P = 0.06] in premenopausal women and 1.0 (95% CI, 0.5-1.7; P = 0.9) in postmenopausal women. The observed 1.8-fold elevated risk in premenopausal women with invasive breast cancer is highly suggestive of a possible interaction between UGT genotype and hormones. Additional analyses suggested a stronger association of UGT1A1 genotype with estrogen receptor (ER)-negative breast cancer. Among premenopausal women, the association was stronger for ER- breast cancer (OR, 2.1; 95% CI, 1.0-4.2; P = 0.04) than ER+ breast cancer (OR, 1.3; 95% CI, 0.6-3.0; P = 0.5). The OR was slightly stronger among women who used oral contraceptives, and the association remained null in postmenopausal women, regardless of whether they took hormone replacement therapy. Our current findings suggest that further investigations are warranted to elucidate the role of UGT1A1 in breast cancer risk.


Subject(s)
Black People/genetics , Breast Neoplasms/ethnology , Glucuronosyltransferase/genetics , Adult , Aged , Breast Neoplasms/genetics , Female , Genotype , Humans , Middle Aged , Polymorphism, Genetic , Promoter Regions, Genetic , Receptors, Estrogen/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
17.
Mol Pharmacol ; 56(2): 359-69, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10419555

ABSTRACT

Most drugs for cancer therapy are targeted to relative differences in the biological characteristics of cancer cells and normal cells. The therapeutic index of such drugs is theoretically limited by the magnitude of such differences, and most anticancer drugs have considerable toxicity to normal cells. Here we describe a new approach for developing anticancer drugs. This approach, termed variagenic targeting, exploits the absolute difference in the genotype of normal cells and cancer cells arising from normal gene sequence variation in essential genes and loss of heterozygosity (LOH) occurring during oncogenesis. The technology involves identifying genes that are: 1) essential for cell survival; 2) are expressed as multiple alleles in the normal population because of the presence of one or more nucleotide polymorphisms; and 3) are frequently subject to LOH in several common cancers. An allele-specific drug inhibiting the essential gene remaining in cancer cells would be lethal to the malignant cell and would have minimal toxicity to the normal heterozygous cell that retains the drug-insensitive allele. With antisense oligonucleotides designed to target two alternative alleles of replication protein A, 70-kDa subunit (RPA70) we demonstrate in vitro selective killing of cancer cells that contain only the sensitive allele of the target gene without killing cells expressing the alternative RPA70 allele. Additionally, we identify several other candidate genes for variagenic targeting. This technology represents a new approach for the discovery of agents with high therapeutics indices for treating cancer and other proliferative disorders.


Subject(s)
Antineoplastic Agents/therapeutic use , Loss of Heterozygosity , Neoplasms/drug therapy , Oligoribonucleotides, Antisense/therapeutic use , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drug Design , Feasibility Studies , Gene Targeting , Genetic Variation , Genome, Human , HeLa Cells , Humans , Neoplasms/genetics , Oligoribonucleotides, Antisense/pharmacology , Replication Protein A , Suppression, Genetic , Tumor Cells, Cultured
18.
Am J Med Genet ; 88(4): 348-51, 1999 Aug 20.
Article in English | MEDLINE | ID: mdl-10402501

ABSTRACT

To determine the importance of a candidate gene KCNN3 (formerly named hSKCa3) in the susceptibility to schizophrenia, we have studied the genotypes of a (CAG)n polymorphism within this gene in the DNAs of the members of 54 multiplex families with this disease. Parametric and nonparametric linkage analysis did not provide evidence for linkage between KCNN3 (that we mapped to chromosome 1q21) and schizophrenia. Furthermore, we observed no difference in the distribution of the (CAG)n alleles between affected and normal individuals. These results do not support the hypothesis that larger KCNN3 alleles are preferentially associated with schizophrenia [Chandy et al. 1998 Mol Psychiatr 3:32-37] in individuals from multiply affected families.


Subject(s)
Chromosomes, Human, Pair 1 , Genetic Linkage , Polymorphism, Genetic , Potassium Channels, Calcium-Activated , Potassium Channels/genetics , Schizophrenia/genetics , Trinucleotide Repeats , Chromosome Mapping , Female , Genetic Markers , Genotype , Humans , Male , Small-Conductance Calcium-Activated Potassium Channels
19.
Biochem Biophys Res Commun ; 255(1): 59-63, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-10082655

ABSTRACT

The tumor suppressor gene MEN1 and several oncogenes including CCND1/cyclin D1/PRAD1 map to chromosome 11q13. However, molecular and cytogenetic analysis suggests the presence of a second tumor suppressor locus at this chromosome region. We have identified a novel gene from chromosome 11q13, which encodes a protein of 126 amino acids sharing an overall 57% identity with the p12(DOC-1) protein encoded by the DOC-1 gene, the human homolog of hamster putative tumor suppressor doc-1 (deleted in oral cancer-1). We therefore designated the novel gene as DOC-1R for DOC-1-related. The cytogenetic location was confirmed by chromosome fluorescent in situ hybridization. Northern blot analysis indicated that it was expressed in all the tissues examined. DOC-1R protein showed heterogeneous subcellular localization. RT-PCR-SSCP analysis failed to detect deleterious mutations of the DOC-1R transcript in four premalignant oral keratinocyte lines and 20 different cancer cell lines from tumor types which frequently harbor LOH at chromosome 11q13.


Subject(s)
Chromosomes, Human, Pair 11 , Genes, Tumor Suppressor , Oncogene Proteins/genetics , Proteins/genetics , Tumor Suppressor Proteins , Amino Acid Sequence , Animals , Cell Line , Chromosome Mapping , Cricetinae , DNA Mutational Analysis , Humans , Molecular Sequence Data , Sequence Alignment
20.
Science ; 282(5397): 2275-9, 1998 Dec 18.
Article in English | MEDLINE | ID: mdl-9856955

ABSTRACT

cAMP (3',5' cyclic adenosine monophosphate) is a second messenger that in eukaryotic cells induces physiological responses ranging from growth, differentiation, and gene expression to secretion and neurotransmission. Most of these effects have been attributed to the binding of cAMP to cAMP-dependent protein kinase A (PKA). Here, a family of cAMP-binding proteins that are differentially distributed in the mammalian brain and body organs and that exhibit both cAMP-binding and guanine nucleotide exchange factor (GEF) domains is reported. These cAMP-regulated GEFs (cAMP-GEFs) bind cAMP and selectively activate the Ras superfamily guanine nucleotide binding protein Rap1A in a cAMP-dependent but PKA-independent manner. Our findings suggest the need to reformulate concepts of cAMP-mediated signaling to include direct coupling to Ras superfamily signaling.


Subject(s)
Cyclic AMP/metabolism , GTP-Binding Proteins/metabolism , Proteins/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Adrenal Glands/metabolism , Adult , Amino Acid Sequence , Animals , Brain/metabolism , Cell Line , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Fetus/metabolism , Gene Expression , Guanine Nucleotide Exchange Factors , Humans , In Situ Hybridization , Molecular Sequence Data , Phosphorylation , Proteins/chemistry , Proteins/genetics , Rats , Second Messenger Systems , Sequence Deletion , Signal Transduction , rap GTP-Binding Proteins , ras Guanine Nucleotide Exchange Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...