Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Sci Adv ; 7(18)2021 04.
Article in English | MEDLINE | ID: mdl-33910907

ABSTRACT

Relative brain size has long been considered a reflection of cognitive capacities and has played a fundamental role in developing core theories in the life sciences. Yet, the notion that relative brain size validly represents selection on brain size relies on the untested assumptions that brain-body allometry is restrained to a stable scaling relationship across species and that any deviation from this slope is due to selection on brain size. Using the largest fossil and extant dataset yet assembled, we find that shifts in allometric slope underpin major transitions in mammalian evolution and are often primarily characterized by marked changes in body size. Our results reveal that the largest-brained mammals achieved large relative brain sizes by highly divergent paths. These findings prompt a reevaluation of the traditional paradigm of relative brain size and open new opportunities to improve our understanding of the genetic and developmental mechanisms that influence brain size.

2.
J Clin Pathol ; 57(10): 1057-62, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15452160

ABSTRACT

BACKGROUND: Recent studies have shown that minichromosome maintenance (MCM) proteins (Mcm2-7) may be useful proliferation markers in dysplasia and cancer in various tissues. AIMS: To investigate the use of Mcm7 as a proliferation marker in 79 lymph node negative prostate cancers and compare it with Ki-67, a commonly used cell proliferation marker. METHODS: The percentage of proliferating cells (proliferation index; PI) was calculated for basal and luminal epithelial cells in benign prostate tissue, prostatic intraepithelial neoplasia (PIN), and epithelial cells in adenocarcinoma. The PI for each biomarker was correlated with the preoperative prostate specific antigen concentration, the Gleason score, surgical resection margin status, and the AJCC pT stage for each patient. RESULTS: The mean PIs for Ki-67 and Mcm7 were: benign luminal epithelium 0.7 and 1.2 and benign basal epithelium 0.8 and 8.2; PIN non-basal epithelium 4.9 and 10.6 and PIN basal epithelium 0.7 and 3.1; adenocarcinoma 9.8 and 22.7, respectively. Mcm7 had a significantly higher mean PI (p<0.0001) than Ki-67 for all cell categories except benign luminal epithelial cells. Mcm7 was a better discriminatory marker of proliferation between benign epithelium, PIN, and invasive adenocarcinoma (p<0.0001) than Ki-67. The drop in Mcm7 mean basal cell PI from benign epithelium to PIN epithelium was significantly larger than for Ki-67 (p<0.0001). Mcm7 had a significantly higher PI than Ki-67 at each risk level. CONCLUSION: Mcm7 may be a useful proliferation marker in prostatic neoplasia and warrants further evaluation as a complementary tool in the diagnosis of PIN and prostate carcinoma.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Cell Cycle Proteins/analysis , Cell Proliferation , DNA-Binding Proteins/analysis , Nuclear Proteins/analysis , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Aged , Analysis of Variance , Humans , Immunohistochemistry/methods , Ki-67 Antigen/analysis , Male , Middle Aged , Minichromosome Maintenance Complex Component 7 , Prostate/pathology , Prostatic Hyperplasia/pathology , Risk Assessment
3.
J Clin Pathol ; 57(6): 625-9, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15166269

ABSTRACT

BACKGROUND: Recent studies have reported CD10 expression in myoepithelial cells (MEC) of the breast, supporting its use as a marker to help distinguish invasive breast carcinoma (IC) from ductal carcinoma in situ (DCIS). AIM: To compare the effectiveness of CD10 with smooth muscle myosin heavy chain (SMMHC) in the detection of MEC in benign and malignant breast lesions. METHODS: Histological material from 25 patients with DCIS and 21 with IC were immunostained for CD10 and SMMHC. Staining was scored on a scale of 0 to 3+ (0, no staining; 3+, intense) and the staining distribution was documented as focal, partial, or circumferential. RESULTS: Uniform, 3+ circumferential CD10 and SMMHC staining of MEC was seen in normal breast ducts and lobules, and in ducts and acini involved in sclerosing adenosis and apocrine metaplasia. In an analysis of total ducts involved by DCIS, 3+ circumferential staining was seen in 65 of 366 ducts (17.7%) stained for CD10 versus 190 of 396 ducts (48%) stained for SMMHC. MEC were not detected immunohistochemically in 116 of 366 ducts (31.7%) with anti-CD10 and 50 of 396 (12.7%) with anti-SMMHC. In contrast, all ICs were negative for both CD10 and SMMHC. Focal background staining of stromal myofibroblasts was seen with both CD10 and SMMHC, but CD10 showed a higher rate of non-specific staining of epithelial cells. CONCLUSION: Although CD10 can aid in the distinction between IC and DCIS, SMMHC is a more sensitive and specific marker of MEC and shows less heterogeneity of immunostaining patterns.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Myosin Heavy Chains/analysis , Neprilysin/analysis , Smooth Muscle Myosins/analysis , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Breast/chemistry , Breast Neoplasms/chemistry , Carcinoma, Ductal, Breast/chemistry , Carcinoma, Ductal, Breast/pathology , Carcinoma, Intraductal, Noninfiltrating/chemistry , Carcinoma, Intraductal, Noninfiltrating/pathology , Diagnosis, Differential , Epithelial Cells/chemistry , Female , Humans , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/analysis
4.
Cancer Biother Radiopharm ; 16(2): 147-57, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11385961

ABSTRACT

A comparison was made between labeled antibody accumulations in nude mice having either single or multiple human xenografts. The LS174T tumors were implanted subcutaneously. All animals were given 2 micrograms of labeled murine anti-carcinoembryonic antigen (CEA) monoclonal antibody 111In-mT84.66. Some animals were also given specific antibody pretreatment (SAP) of 200 micrograms of unlabeled mT84.66 to reduce liver accumulation of activity. In order to represent these multiple tumor examples, a simple initial-phase pharmacokinetic model was first fitted to each of the two groups (SAP and PBS treated) of single-tumor animals. Using the resultant six non-adjustable parameters as constants, the n = 1 uptake model was then used to represent tumor, liver and blood accumulations (%injected dose/organ) in the multiple-tumor animals. The model was found to be a good representation; in particular, it had far better agreement than single tumor predictions in the PBS mice. Differences between the single-tumor accumulations and those seen in multiple tumor examples were generally between two- and three-fold. The model also demonstrated that the result of SAP was to essentially eliminate the effect of liver targeting of tumor-secreted CEA. We conclude that an initial-phase one-tumor model can describe the decrease of accumulation of activity in the case of multiple tumors in nude mice in both untreated (PBS) and pretreated conditions. Implications for clinical imaging and therapy with monoclonal agents are discussed.


Subject(s)
Antibodies, Monoclonal/pharmacokinetics , Carcinoembryonic Antigen/immunology , Liver Neoplasms/metabolism , Neoplasms, Experimental/immunology , Algorithms , Animals , Female , Humans , Indium Radioisotopes , Liver Neoplasms/diagnostic imaging , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/pathology , Radionuclide Imaging
5.
Mol Diagn ; 5(1): 23-32, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10837086

ABSTRACT

BACKGROUND: Currently, prostate cancer (CaP) cytogenetics is not well defined, largely because of technical difficulties in obtaining primary tumor metaphases. METHODS AND RESULTS: We examined three CaP cell lines (LNCaP, DU145, PC-3) using sequential Giemsa banding and spectral karyotyping (SKY) to search for a common structural aberration or translocation breakpoint. No consistent rearrangement common to all three cell lines was detected. A clustering of centromeric translocation breakpoints was detected in chromosomes 4, 5, 6, 8, 11, 12, 14, and 15 in DU145 and PC-3. Both these lines were found to have karyotypes with a greater level of complexity than LNCaP. CONCLUSION: The large number of structural aberrations present in DU145 and PC-3 implicate an underlying chromosomal instability and subsequent accumulation of cytogenetic alterations that confer a selective growth advantage. The high frequency of centromeric rearrangements in these lines indicates a potential role for mitotic irregularities associated with the centromere in CaP tumorigenesis.


Subject(s)
Centromere , Chromosome Aberrations , Chromosome Banding/methods , Chromosome Disorders , Karyotyping/methods , Prostatic Neoplasms/genetics , Azure Stains/metabolism , Humans , Male , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
6.
Cytogenet Cell Genet ; 88(3-4): 249-52, 2000.
Article in English | MEDLINE | ID: mdl-10828601

ABSTRACT

Activation of the ERK mitogen-activated protein (MAP) kinase pathway has been implicated in the regulation of cell growth, differentiation and senescence. In this pathway, the MAP kinases ERK1/ERK2 are phosphorylated and activated by the dual-specificity kinases MEK1 and MEK2, which in turn are activated by serine phosphorylation by a number of MAP kinase kinase kinases. We report here the chromosomal localization of the human genes encoding the MAP kinase kinase isoforms MEK1 and MEK2. Using a combination of fluorescence in situ hybridization, somatic cell hybrid analysis, DNA sequencing and yeast artificial chromosome (YAC) clone analysis, we have mapped the MEK1 gene (MAP2K1) to chromosome 15q21. We also present evidence for the presence of a MEK1 pseudogene on chromosome 8p21. The MEK2 gene (MAP2K2) was mapped to chromosome 7q32 by fluorescence in situ hybridization and YAC clone analysis.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 7/genetics , Mitogen-Activated Protein Kinase Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Blotting, Southern , Chromosomes, Artificial, Yeast/genetics , Chromosomes, Human, Pair 8/genetics , Cloning, Molecular , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Isoenzymes/genetics , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Molecular Sequence Data , Physical Chromosome Mapping , Pseudogenes/genetics , Sequence Analysis, DNA
7.
Health Educ Behav ; 27(2): 241-53, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10768805

ABSTRACT

The North Carolina Black Churches United for Better Health project was a 4-year intervention trial that successfully increased fruit and vegetable (F&V) consumption among rural African American adults, for cancer and chronic disease prevention. The multicomponent intervention was based on an ecological model of change. A process evaluation that included participant surveys, church reports, and qualitative interviews was conducted to assess exposure to, and relative impact of, interventions. Participants were 1,198 members of 24 intervention churches who responded to the 2-year follow-up survey. In addition, reports and interviews were obtained from 23 and 22 churches, respectively. Serving more F&V at church functions was the most frequently reported activity and had the highest perceived impact, followed by the personalized tailored bulletins, pastor sermons, and printed materials. Women, older individuals, and members of smaller churches reported higher impact of certain activities. Exposure to interventions was associated with greater F&V intake. A major limitation was reliance on church volunteers to collect process data.


Subject(s)
Black or African American , Christianity , Diet , Health Promotion/methods , Nutritional Sciences/education , Adult , Demography , Female , Fruit , Health Surveys , Humans , Interviews as Topic , Male , North Carolina , Program Evaluation/methods , Vegetables
8.
J Mol Biol ; 295(3): 489-504, 2000 Jan 21.
Article in English | MEDLINE | ID: mdl-10623541

ABSTRACT

Glucuronidation is a major pathway of androgen metabolism and is catalyzed by UDP-glucuronosyltransferase (UGT) enzymes. UGT2B15 and UGT2B17 are 95% identical in primary structure, and are expressed in steroid target tissues where they conjugate C19 steroids. Despite the similarities, their regulation of expression are different; however, the promoter region and genomic structure of only the UGT2B17 gene have been characterizedX to date. To isolate the UGT2B15 gene and other novel steroid-conjugating UGT2B genes, eight P-1-derived artificial chromosomes (PAC) clones varying in length from 30 kb to 165 kb were isolated. The entire UGT2B15 gene was isolated and characterized from the PAC clone 21598 of 165 kb. The UGT2B15 and UGT2B17 genes are highly conserved, are both composed of six exons spanning approximately 25 kb, have identical exon sizes and have identical exon-intron boundaries. The homology between the two genes extend into the 5'-flanking region, and contain several conserved putative cis-acting elements including Pbx-1, C/EBP, AP-1, Oct-1 and NF/kappaB. However, transfection studies revealed differences in basal promoter activity between the two genes, which correspond to regions containing non-conserved potential elements. The high degree of homology in the 5'-flanking region between the two genes is lost upstream of -1662 in UGT2B15, and suggests a site of genetic recombination involved in duplication of UGT2B genes. Fluorescence in situ hybridization mapped the UGT2B15 gene to chromosome 4q13.3-21.1. The other PAC clones isolated contain exons from the UGT2B4, UGT2B11 and UGT2B17 genes. Five novel exons, which are highly homologous to the exon 1 of known UGT2B genes, were also identified; however, these exons contain premature stop codons and represent the first recognized pseudogenes of the UGT2B family. The localization of highly homologous UGT2B genes and pseudogenes as a cluster on chromosome 4q13 reveals the complex nature of this gene locus, and other novel homologous UGT2B genes encoding steroid conjugating enzymes are likely to be found in this region of the genome.


Subject(s)
Chromosomes, Human, Pair 4 , Glucuronosyltransferase/genetics , Isoenzymes/genetics , Multigene Family , Pseudogenes , Amino Acid Sequence , Base Sequence , Cell Line , Chromosome Mapping , Cloning, Molecular , DNA, Complementary , Exons , Humans , In Situ Hybridization, Fluorescence , Introns , Molecular Sequence Data , Open Reading Frames , Promoter Regions, Genetic
11.
Gene Ther ; 6(4): 482-97, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10476208

ABSTRACT

Inefficient nuclear delivery of plasmid DNA is thought to be one of the daunting hurdles to gene transfer, utilizing a nonviral delivery system such as polycation-DNA complex. Following its internalization by endocytosis, plasmid DNA has to be released into the cytosol before its nuclear entry can occur. However, the stability of plasmid DNA in the cytoplasm, that may play a determinant role in the transfection efficiency, is not known. The turnover of plasmid DNA, delivered by microinjection into the cytosol, was determined by fluorescence in situ hybridization (FISH) and quantitative single-cell fluorescence video-image analysis. Both single- and double-stranded circular plasmid DNA disappeared with an apparent half-life of 50-90 min from the cytoplasm of HeLa and COS cells, while the amount of co-injected dextran (MW 70,000) remained unaltered. We propose that cytosolic nuclease(s) are responsible for the rapid-degradation of plasmid DNA, since (1) elimination of plasmid DNA cannot be attributed to cell division or to the activity of apoptotic and lysosomal nucleases; (2) disposal of microinjected plasmid DNA was inhibited in cytosol-depleted cells or following the encapsulation of DNA in phospholipid vesicles; (3) generation and subsequent elimination of free 3'-OH ends could be detected by the terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling assay (TUNEL), reflecting the fragmentation of the injected DNA; and finally (4) isolated cytosol, obtained by selective permeabilization of the plasma membrane, exhibits divalent cation-dependent, thermolabile nuclease activity, determined by Southern blotting and 32P-release from end-labeled DNA. Collectively, these findings suggest that the metabolic instability of plasmid DNA, caused by cytosolic nuclease, may constitute a previously unrecognized impediment for DNA translocation into the nucleus and a possible target to enhance the efficiency of gene delivery.


Subject(s)
Cytosol/metabolism , Gene Transfer Techniques , Plasmids/metabolism , Transfection , Blotting, Southern , Calcium/metabolism , Deoxyribonucleases/metabolism , Digitonin/metabolism , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Microinjections , Microscopy, Fluorescence
12.
Biochem Biophys Res Commun ; 261(1): 163-8, 1999 Jul 22.
Article in English | MEDLINE | ID: mdl-10405340

ABSTRACT

The human MEP1A gene encodes the meprin alpha subunit that consists of a protease domain conserved in the astacin family of metalloendopeptidases and several C-terminal interaction domains present in other proteins. Using the alpha subunit cDNA, we identified two clones from a human P1-derived artificial chromosome (PAC) library. Fluorescence in situ hybridization (FISH) mapped both PACs (1e12, 65a14) to chromosome 6p21, confirming the MEP1A location. FISH also mapped PAC 65a14 to chromosome 13cen, and to chromosome 9 in three different regions, 9p12-13, 9q21, and 9q22. Southern blot analysis showed that sequences of PAC 65a14 and MEP1A were similar in the 3' end but different in the 5' end, revealing for the first time that the human genome may encode multiple interaction domains highly similar to those of the meprin alpha subunit. The symbols of MEP1AL1, MEP1AL2, MEP1AL3, and MEP1AL4 have been designated for MEP1A-like sequences on 9p12-13, 9q21, 9q22, and 13cen, respectively.


Subject(s)
Chromosomes, Human/genetics , Genes/genetics , Metalloendopeptidases/genetics , Sequence Homology, Nucleic Acid , Animals , Blotting, Southern , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 6/genetics , Chromosomes, Human, Pair 9/genetics , Cloning, Molecular , Gene Dosage , Genome, Human , Genomic Library , Humans , In Situ Hybridization, Fluorescence , Metalloendopeptidases/chemistry , Mice
13.
Genetics ; 152(2): 685-97, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10353910

ABSTRACT

In the present study, we investigated intrachromosomal homologous recombination in a murine hybridoma in which the recipient for recombination, the haploid, endogenous chromosomal immunoglobulin mu-gene bearing a mutation in the constant (Cmu) region, was separated from the integrated single copy wild-type donor Cmu region by approximately 1 Mb along the hybridoma chromosome. Homologous recombination between the donor and recipient Cmu region occurred with high frequency, correcting the mutant chromosomal mu-gene in the hybridoma. This enabled recombinant hybridomas to synthesize normal IgM and to be detected as plaque-forming cells (PFC). Characterization of the recombinants revealed that they could be placed into three distinct classes. The generation of the class I recombinants was consistent with a simple unequal sister chromatid exchange (USCE) between the donor and recipient Cmu region, as they contained the three Cmu-bearing fragments expected from this recombination, the original donor Cmu region along with both products of the single reciprocal crossover. However, a simple mechanism of homologous recombination was not sufficient in explaining the more complex Cmu region structures characterizing the class II and class III recombinants. To explain these recombinants, a model is proposed in which unequal pairing between the donor and recipient Cmu regions located on sister chromatids resulted in two crossover events. One crossover resulted in the deletion of sequences from one chromatid forming a DNA circle, which then integrated into the sister chromatid by a second reciprocal crossover.


Subject(s)
DNA/genetics , Recombination, Genetic , Animals , Cell Line , Chromosomes/genetics , Hybridomas , Immunoglobulin mu-Chains/genetics , In Situ Hybridization, Fluorescence , Mammals , Mutation
15.
Genomics ; 55(2): 185-93, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9933565

ABSTRACT

The mouse looptail (Lp) mutation is an established model for neural tube defects with homozygous Lp embryos showing an open neural tube from the caudal midbrain to the tip of the tail. Heterozygous Lp mice are characterized by a "looped-tail" and wobbly head movements. The Lp gene has been mapped to a 0.6-cM interval on mouse chromosome 1 delineated by two clusters of markers, Fcer1gamma/Usf1/D1Mit113/D1Wsu1 on the proximal side and Fcer1alpha/Spna1/D1Mit149 distally. In the present study, we have created a high-resolution physical map of the Lp genetic interval that is based on long-range restriction mapping by PFGE, fluorescence in situ hybridization analysis of interphase nuclei and extended chromatid fibers, and the assembly of a cloned contig. This contig consists of 25 independent and overlapping BAC clones and 3 YAC clones. The combined analysis indicates that the 0.6-cM genetic interval for Lp corresponds to a minimal physical interval of 700 kb that is delineated by D1Mit113 proximally (two crossovers) and Fcer1alpha distally (one crossover). The overall gene order and intergene distances for the region were determined to be D1Mit113-<150 kb-Nhlh1-250 kb-Atp1alpha2-280 kb-Fcer1alpha. Partial sequencing of BAC clones from the contig yielded 42 new STS markers for this region of mouse chromosome 1. Sequence analysis of the BAC clones and assignment of ESTs from the human transcript map to the cloned contig allowed the placement of four new transcription units within this region: Pc326, Kiaa0253, and Pea15 were positioned in the Nhlh1/Atp1alpha2 nonrecombinant interval, while Girk3 was located distal to Atp1alpha2.


Subject(s)
Mutation , Neural Tube Defects/genetics , Physical Chromosome Mapping , Animals , Chromosomes, Artificial, Yeast , Chromosomes, Bacterial , Contig Mapping , Electrophoresis, Polyacrylamide Gel , Humans , In Situ Hybridization, Fluorescence , Mice , Molecular Sequence Data , Restriction Mapping , Sequence Tagged Sites
16.
Genomics ; 62(3): 529-32, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10644453

ABSTRACT

A murine Ets2 target gene isolated by differential display cloning was identified as the phospholipase A2 activating protein (PLAA) gene. A 2.7-kb human cDNA demonstrating high homology to mouse and rat Plaa genes was then isolated and characterized. Human PLAA contains six WD-40 repeat motifs and three different protein kinase consensus domains. Fluorescence in situ hybridization (FISH) mapping placed PLAA on chromosome 9p21, a region frequently deleted in various cancers. A comprehensive mapping strategy was employed to define further the chromosomal localization of PLAA relative to CDKN2A within the 9p21 locus. Radiation hybrid mapping placed the gene 7.69 cR from WI-5735 (LOD >3.0), a marker in close proximity to CDKN2A and CDKN2B. Yeast artificial chromosome (YAC) mapping localized PLAA proximal to the CDKN2A/CDKN2B genes and to a region flanked by D9S171 and INFA commonly deleted in many neoplasms. Two YACs contained both PLAA and D9S259, a marker present in a second more proximal minimal deleted region observed in cutaneous melanoma and squamous cell lung carcinoma. Double-color fiber FISH mapping confirmed the location of PLAA centromeric to D9S171 and CDKN2A/CDKN2B. The mapping data suggest a possible tumor suppressor role for this gene.


Subject(s)
Chromosomes, Human, Pair 9/genetics , DNA-Binding Proteins , Genes, Tumor Suppressor/genetics , Physical Chromosome Mapping , Proteins/genetics , Proto-Oncogene Proteins/physiology , Repressor Proteins , Trans-Activators/physiology , Transcription Factors , Animals , Bacteriophage P1/genetics , Carcinoma, Squamous Cell/genetics , Chromosomes, Artificial, Yeast/genetics , Humans , Hybrid Cells , In Situ Hybridization, Fluorescence , Lod Score , Lung Neoplasms/genetics , Melanoma/genetics , Mice , Proto-Oncogene Protein c-ets-2 , Rats , Skin Neoplasms/genetics
17.
Biochem Biophys Res Commun ; 247(3): 580-6, 1998 Jun 29.
Article in English | MEDLINE | ID: mdl-9647736

ABSTRACT

The normal epithelial cell-specific 1 (NES1) gene is a recently identified novel serine protease-like gene which is down-regulated during breast cancer progression. The gene product has 34-42% identity with the members of three distinct serine protease families: the trypsin-like family, activators of kringle domain-containing growth factors, and the kallikrein family (X. L. Liu et al., (1996) Cancer Res 56, 3371-3379). Although the cDNA of this gene has been cloned, its genomic structure and chromosomal position are not as yet known. Here, we report the genomic characterization and mapping of the NES1 gene. By subcloning and sequencing a PAC clone containing the complete NES1 gene, we were able to characterize the structure of this gene. The NES1 gene spans 5.5 kb and is composed of five coding exons and one untranslated exon. The positions of the introns were similar to trypsinogen, prostate specific antigen (PSA), and tissue plasminogen activator (TPA). NES1 gene was also localized with somatic cell mapping, radiation hybrid mapping, and fluorescence in situ hybridization techniques to chromosome 19q13.3-q13.4, the same region where the human kallikrein gene family resides. Taken together, our results suggest that the NES1 gene originates from the same ancestor as trypsinogen, PSA, and TPA, but remains in close proximity to PSA.


Subject(s)
Kallikreins , Neoplasm Proteins/chemistry , Amino Acid Sequence , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 19/genetics , Cloning, Molecular , Evolution, Molecular , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Prostate-Specific Antigen/chemistry , Sequence Analysis, DNA , Serine Endopeptidases/chemistry , Tissue Plasminogen Activator/chemistry , Trypsinogen/chemistry
18.
Oncogene ; 16(19): 2459-67, 1998 May 14.
Article in English | MEDLINE | ID: mdl-9627112

ABSTRACT

To define genes that are essential to the initiation and progression of breast cancer we utilized subtractive hybridization and differential display cloning techniques and isolated over 950 cDNAs from breast cell-lines derived from matched normal and tumor tissue. Of these, 102 cDNAs were characterized by DNA sequencing and Northern blot analysis. GenBank searches showed that one of these genes, T1A12 is identical to mac25, an insulin-like growth factor-binding protein related gene. Antibodies generated against the C-terminal region of the T1A12/mac25 protein were used to investigate its expression in 60 primary breast tissues. Sections of 12 benign, 16 ductal carcinoma in situ and 32 infiltrating ductal carcinoma specimens were examined. Strong immunoperoxidase staining was observed in luminal epithelial cells of normal lobules and ducts, in apocrine cells of cysts and fibroadenomas. Moderate to weak protein expression was found in hyperplastic and DCIS cells, but no specific staining was detected in invasive carcinoma cells. FISH mapping using a PAC clone localized the T1A12/mac25 gene to 4q12-13. Microsatellite length polymorphism was studied using markers for 4q in paired normal and tumor breast tissues. Thirty-three per cent (10/30) of the samples were found to be polymorphic with D4S189 and D4S231 microsatellite markers and LOH was detected in 50% (5/10) of these informative samples. Our data indicate that T1A12/mac25 expression is abrogated during breast cancer progression concomitant with loss of heterozygosity on chromosome 4q. T1A12/mac25 may therefore have a tumor suppressor-like function and its expression could indicate a disease with a more favorable status, having a better prognosis.


Subject(s)
Breast Neoplasms/genetics , Carrier Proteins/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Insulin-Like Growth Factor Binding Proteins , Breast Neoplasms/pathology , Cell Line , Chromosomes, Human, Pair 4 , Disease Progression , Female , Humans , Loss of Heterozygosity , Tumor Cells, Cultured
19.
Hum Mol Genet ; 7(3): 501-5, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9467010

ABSTRACT

While the presence of a lipoyl-containing protein (protein X) separate from lipoyl transacetylase in the pyruvate dehydrogenase complex (PDC) has been known for some time, until recently only the cDNA for the yeast enzyme has been cloned. We have cloned, sequenced and characterized the cDNA encoding the human protein X and localized the protein X gene to chromosome 11p13. We also report here a new case of protein X deficiency identified immunologically, with decreased activity of PDC and without mutations in the E1alpha subunit or E1beta subunit. We report that the cDNA and gene of this patient for protein X has a homozygous 4 bp deletion, specifically in the putative mitochondrial targeting signal sequence which results in a premature stop codon. This is the first documented case of a molecular defect in pyruvate dehydrogenase protein X.


Subject(s)
Chromosomes, Human, Pair 11 , Homozygote , Peptides/genetics , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics , Pyruvate Dehydrogenase Complex/genetics , Sequence Deletion , Amino Acid Sequence , Base Sequence , Child , Chromosome Mapping , Cloning, Molecular , Codon, Terminator , Humans , Macromolecular Substances , Male , Molecular Sequence Data , Peptides/chemical synthesis , Pyruvate Dehydrogenase Complex/biosynthesis , Pyruvate Dehydrogenase Complex/chemical synthesis , Recombinant Proteins/biosynthesis , Sequence Homology, Amino Acid
20.
Mamm Genome ; 9(2): 136-43, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9457675

ABSTRACT

We have cloned and characterized the Na,K-ATPase beta 3 subunit gene (ATP1B3), and a beta 3 subunit pseudogene (ATP1B3P1), from a human PAC genomic library. The beta 3 subunit gene is > 50 kb in size and is split into 7 exons. The exon/intron organization of the beta 3 subunit gene is identical to that of the Na,K-ATPase beta 3 subunit gene, indicating that these two genes evolved from a common evolutionary ancestor. Comparison of the promoter region of the human and mouse beta 3 subunit gene reveals a high degree of homology within a 300-bp segment located immediately upstream of the translation start site, suggesting that control elements that serve to regulate the cell-specific expression of the beta 3 subunit gene are likely to be located within this conserved region. Dot blot analysis of beta 3 subunit transcripts revealed expression within virtually all human tissues, while in situ hybridization showed expression of beta 3 mRNA in both neurons and glia of rat brain. Fluorescence in situ hybridization with PAC DNA clones localized ATP1B3 to the q22-->23 region of Chromosome (Chr) 3, and the beta 3 pseudogene to the p13-->15 region of Chr 2.


Subject(s)
Chromosomes, Human, Pair 2/genetics , Chromosomes, Human, Pair 3/genetics , Pseudogenes , Sodium-Potassium-Exchanging ATPase/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Cloning, Molecular , Exons , Gene Expression , Gene Library , Humans , In Situ Hybridization, Fluorescence , Introns , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Rats , Sodium-Potassium-Exchanging ATPase/chemistry , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...