Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
AIDS Care ; 29(6): 675-685, 2017 06.
Article in English | MEDLINE | ID: mdl-27998178

ABSTRACT

HIV stigma can inhibit uptake of HIV testing and antiretroviral therapy as well as negatively affect mental health. Efforts to reduce discrimination against people living with HIV (LWH) have contributed to greater acceptance of the infection. Female sex workers (FSW) LWH may experience overlapping stigma due to both their work and HIV status, although this is poorly understood. We examined HIV and sex-work stigma experienced by FSW LWH in Zimbabwe. Using the SAPPH-IRe cluster-randomised trial baseline survey, we analysed the data from 1039 FSW self-reporting HIV. The women were recruited in 14 sites using respondent-driven sampling. We asked five questions to assess internalised and experienced stigma related to working as a sex worker, and the same questions were asked in reference to HIV. Among all FSW, 91% reported some form of sex-work stigma. This was not associated with sociodemographic or sex-work characteristics. Rates of sex-work stigma were higher than those of HIV-related stigma. For example, 38% reported being "talked badly about" for LWH compared with 77% for their involvement in sex work. Those who reported any sex-work stigma also reported experiencing more HIV stigma compared to those who did not report sex-work stigma, suggesting a layering effect. FSW in Zimbabwe experience stigma for their role as "immoral" women and this appears more prevalent than HIV stigma. As HIV stigma attenuates, other forms of social stigma associated with the disease may persist and continue to pose barriers to effective care.


Subject(s)
HIV Infections/epidemiology , Sex Work/psychology , Sex Workers/psychology , Social Stigma , Adult , Female , HIV Infections/psychology , Health Surveys , Humans , Middle Aged , Sampling Studies , Young Adult , Zimbabwe
2.
Opt Express ; 23(10): 12562-72, 2015 May 18.
Article in English | MEDLINE | ID: mdl-26074511

ABSTRACT

Precise tailoring of optical vector beams is demonstrated, shaping their focal electric fields and used to create complex laser micro-patterning on a metal surface. A Spatial Light Modulator (SLM) and a micro-structured S-waveplate were integrated with a picosecond laser system and employed to structure the vector fields into radial and azimuthal polarizations with and without a vortex phase wavefront as well as superposition states. Imprinting Laser Induced Periodic Surface Structures (LIPSS) elucidates the detailed vector fields around the focal region. In addition to clear azimuthal and radial plasmon surface structures, unique, variable logarithmic spiral micro-structures with a pitch Λ âˆ¼1µm, not observed previously, were imprinted on the surface, confirming unambiguously the complex 2D focal electric fields. We show clearly also how the Orbital Angular Momentum(OAM) associated with a helical wavefront induces rotation of vector fields along the optic axis of a focusing lens and confirmed by the observed surface micro-structures.

3.
Opt Express ; 21(18): 21198-207, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-24103993

ABSTRACT

We report on new developments in wavefront and polarization control for ultrashort-pulse laser microprocessing. We use two Spatial Light Modulators in combination to structure the optical fields of a picosecond-pulse laser beam, producing vortex wavefronts and radial or azimuthal polarization states. We also carry out the first demonstration of multiple first-order beams with vortex wavefronts and radial or azimuthal polarization states, produced using Computer Generated Holograms. The beams produced are used to nano-structure a highly polished metal surface. Laser Induced Periodic Surface Structures are observed and used to directly verify the state of polarization in the focal plane and help to characterize the optical properties of the setup.

4.
Opt Express ; 21(21): 25333-43, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24150374

ABSTRACT

The polarization state of an ultrafast laser is dynamically controlled using two Spatial Light Modulators and additional waveplates. Consequently, four states of polarization, linear horizontal and vertical, radial and azimuthal, all with a ring intensity distribution, were dynamically switched at a frequency ν = 12.5 Hz while synchronized with a motion control system. This technique, demonstrated here for the first time, enables a remarkable level of real-time control of the properties of light waves and applied to real-time surface patterning, shows that highly controlled nanostructuring is possible. Laser ablation of Induced Periodic Surface Structures is used to directly verify the state of polarization at the focal plane.

5.
Oncogene ; 30(40): 4152-62, 2011 Oct 06.
Article in English | MEDLINE | ID: mdl-21499300

ABSTRACT

Ovarian endometrioid adenocarcinomas (OEAs) frequently exhibit constitutive activation of canonical WNT signaling, usually as a result of oncogenic mutations that stabilize and dysregulate the ß-catenin protein. In previous work, we used microarray-based methods to compare gene expression in OEAs with and without dysregulated ß-catenin as a strategy for identifying novel ß-catenin/TCF target genes with important roles in ovarian cancer pathogenesis. Among the genes highlighted by the microarray studies was MSX2, which encodes a homeobox transcription factor. We found MSX2 expression was markedly increased in primary human and murine OEAs with dysregulated ß-catenin compared with OEAs with intact ß-catenin regulation. WNT pathway activation by WNT3a ligand or GSK3ß inhibitor treatment potently induced MSX2 and ectopic expression of a dominant negative form of TCF4 inhibited MSX2 expression in ovarian cancer cells. Chromatin immunoprecipitation studies demonstrated that ß-catenin/TCF directly regulates MSX2 expression via binding to TCF binding elements in multiple regions of the MSX2 gene. Notably, ectopic MSX2 expression was found to promote neoplastic transformation of the rodent RK3E model epithelial cell line and to enhance the invasiveness of immortalized human ovarian epithelial cells in vitro and ovarian carcinoma cells in vivo. Inhibition of endogenous MSX2 expression in ovarian endometrioid cancer cells carrying a ß-catenin mutation using shRNA approaches inhibited neoplastic properties of the cells in vitro and in vivo. Expression of MSX2 in selected ovarian carcinoma cells induced changes suggestive of epithelial-mesenchymal transition (EMT), but based on analysis of ovarian cell lines and primary tumor tissues, effects of MSX2 on EMT appear to be complex and context-dependent. Our findings indicate MSX2 is a direct downstream transcriptional target of ß-catenin/TCF and has a key contributing role in the cancer phenotype of OEAs carrying WNT/ß-catenin pathway defects.


Subject(s)
Carcinoma, Endometrioid/metabolism , Homeodomain Proteins/metabolism , Oncogenes , Ovarian Neoplasms/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , Carcinoma, Endometrioid/pathology , Cell Transformation, Neoplastic , Female , Homeodomain Proteins/genetics , Humans , Mice , Ovarian Neoplasms/pathology , RNA, Messenger/genetics , beta Catenin/metabolism
6.
Oncogene ; 27(27): 3875-9, 2008 Jun 19.
Article in English | MEDLINE | ID: mdl-18246119

ABSTRACT

In epithelial cells, the tight junction divides the plasma membrane into distinct apical and basolateral domains. Polarization is essential for epithelial cell function, and apico-basal cell polarity is lost during the epithelial to mesenchymal transition (EMT), a program of events characterized not only by loss of cell polarity, but also by enhanced cell motility and increased cell invasion. Among several apically localized protein complexes, the Crumbs and Par protein complexes have pivotal roles in control of epithelial polarity and apical membrane formation. Here, we demonstrate that the Snail transcriptional repressor antagonizes expression of the Crumbs polarity complex. We show that Snail abolishes localization of the Crumbs and Par complexes to the tight junction, decreases Crumbs complex protein levels and suppresses Crumbs3 transcription. Evidence that Snail acts directly to antagonize Crumbs3 promoter activity is presented. Strikingly, we note that reexpression of exogenous Crumbs3 in Snail-expressing Madin-Darby Canine Kidney cells partially restores cell-cell junctions. Moreover, we find that the EMT inducer transforming growth factor-beta elicits transcriptional repression of Crumbs3 and results in a measurable loss of Crumbs3 protein. Our findings provide new insights into the links between the transcriptional repression function of Snail and its role in antagonizing key apico-basal polarity factors during EMT.


Subject(s)
Membrane Glycoproteins/genetics , Transcription Factors/metabolism , Animals , Cell Line , Cell Membrane/physiology , Cell Movement/physiology , Cell Polarity/genetics , Dogs , Epithelial Cells/cytology , Epithelial Cells/physiology , Gene Expression Regulation, Neoplastic , Kidney , Mesoderm/cytology , Mesoderm/physiology , Snail Family Transcription Factors , Tight Junctions/genetics , Tight Junctions/physiology
7.
Gut ; 51(2): 184-90, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12117877

ABSTRACT

BACKGROUND: The Cdx genes are expressed in the colorectal epithelium and are frequently downregulated during tumorigenesis. Overexpression of Cdx genes has been shown previously to result in cellular differentiation. AIM: To study expression of CDX2 in normal and neoplastic human colon using a newly isolated monoclonal antibody. To define expression of CDX1 and CDX2 in an in vitro model system of colorectal tumour progression and to ascertain whether these are subject to regulation during differentiation. METHODS: Normal and neoplastic human colon was immunostained for CDX2. CDX1 and CDX2 expression was assayed in cell lines derived from premalignant colonic adenomas by western blotting. Differentiation was induced by sodium butyrate treatment or post confluent growth, and changes in CDX expression compared with carcinoma cell lines with low levels of CDX expression. RESULTS: CDX2 protein displayed no gradient of expression within the colonic crypt. Cell lines derived from adenomas, with high levels of CDX1 and CDX2, showed no regulation of these proteins when induced to differentiate by butyrate or confluency. CDX expression in these cell lines was independent of their APC or Ras status. CDX1 and CDX2 were expressed at very low levels in some carcinoma cell lines and were modestly upregulated on differentiation but were not restored to levels seen in adenoma cells. CONCLUSION: The lack of significant regulation on cellular differentiation and the absence of a detectable gradient in the crypt implies that CDX2 may confer tissue specificity but may not play the previously suggested role in crypt patterning.


Subject(s)
Adenoma/chemistry , Colon/chemistry , Colonic Neoplasms/chemistry , Homeodomain Proteins/analysis , Adenoma/metabolism , Animals , Blotting, Western , Butyrates , CDX2 Transcription Factor , Cell Differentiation , Colon/metabolism , Colonic Neoplasms/metabolism , Disease Progression , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry/methods , Mice , Trans-Activators , Tumor Cells, Cultured
8.
Am J Pathol ; 159(6): 2239-48, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11733373

ABSTRACT

Most large bowel cancers are moderately to well-differentiated adenocarcinomas comprised chiefly or entirely of glands lined by tall columnar cells. We have identified a subset of poorly differentiated colon carcinomas with a distinctive histopathological appearance that we term large cell minimally differentiated carcinomas (LCMDCs). These tumors likely include a group of poorly differentiated carcinomas previously described by others as medullary adenocarcinomas. To better understand the pathogenesis of these uncommon neoplasms, we compared molecular features of 15 LCMDCs to those present in 25 differentiated adenocarcinomas (DACs) of the colon. Tumors were examined for alterations commonly seen in typical colorectal carcinomas, including increased p53 and beta-catenin immunoreactivity, K-ras gene mutations, microsatellite instability, and loss of heterozygosity of markers on chromosomes 5q, 17p, and 18q. In addition, tumors were evaluated by immunohistochemistry for CDX2, a homeobox protein whose expression in normal adult tissues is restricted to intestinal and colonic epithelium. Markedly reduced or absent CDX2 expression was noted in 13 of 15 (87%) LCMDCs, whereas only 1 of the 25 (4%) DACs showed reduced CDX2 expression (P < 0.001). Nine of 15 (60%) LCMDCs had the high-frequency microsatellite instability phenotype, but only 2 of 25 (8%) DACs had the high-frequency microsatellite instability phenotype (P = 0.002). Our findings provide support for the hypothesis that the molecular pathogenesis of LCMDCs is distinct from that of most DACs. CDX2 alterations and DNA mismatch repair defects have particularly prominent roles in the development of LCMDCs.


Subject(s)
Carcinoma, Large Cell/pathology , Colonic Neoplasms/pathology , DNA-Binding Proteins , Homeodomain Proteins/biosynthesis , Microsatellite Repeats/genetics , Trans-Activators , Adaptor Proteins, Signal Transducing , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , CDX2 Transcription Factor , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/metabolism , Carrier Proteins , Chromosomes, Human, Pair 17/genetics , Chromosomes, Human, Pair 18/genetics , Chromosomes, Human, Pair 5/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Cytoskeletal Proteins/analysis , Female , Genes, ras/genetics , Humans , Immunohistochemistry , Loss of Heterozygosity , Male , Middle Aged , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Mutation , Neoplasm Proteins/analysis , Nuclear Proteins , Proto-Oncogene Proteins/analysis , Tumor Suppressor Protein p53/analysis , beta Catenin
9.
Cancer Res ; 61(22): 8247-55, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719457

ABSTRACT

Clinical and molecular findings suggest that the four major histological subtypes of ovarian carcinoma (serous, clear cell, mucinous, and endometrioid) likely represent distinct disease entities. Prior studies have shown that ovarian endometrioid adenocarcinomas (OEAs) often carry mutations in the CTNNB1 gene, which encodes beta-catenin, a critical component of the Wnt signaling pathway. However, the nature of other defects in the Wnt signaling pathway in ovarian carcinomas remains largely unknown. Thus, in 45 primary OEAs and two OEA-derived cell lines, we sought to comprehensively address the prevalence of and mechanisms underlying beta-catenin and Wnt pathway deregulation. CTNNB1 missense mutations were detected in 14 primary tumors. All mutations affected the NH(2)-terminal regulatory domain of beta-catenin, presumably rendering the mutant proteins resistant to degradation. Immunohistochemical studies revealed nuclear accumulation of beta-catenin in all but two tumors with CTNNB1 mutations. Two primary tumors lacking CTNNBI mutations showed strong nuclear immunoreactivity for beta-catenin. In one of the two tumors, biallelic inactivation of the APC gene was found. In the remaining 29 primary OEAs, unequivocal nuclear beta-catenin immunoreactivity was not observed, though a nonsense mutation in AXIN1 was observed in one tumor and a truncating frameshift mutation in AXIN2 was seen in another case. Both OEA-derived cell lines studied (TOV-112D and MDAH-2774) had elevated constitutive T-cell factor/lymphoid enhancer factor transcriptional activity. TOV-112D cells were shown to harbor mutant beta-catenin, whereas a missense AXIN1 sequence alteration was identified in MDAH-2774 cells. Collectively, our findings demonstrate frequent defects of the Wnt signaling pathway in a particular subtype of ovarian carcinomas, i.e., OEAs. Although mutations in the CTNNB1 gene are the most common mechanism of beta-catenin deregulation in OEAs, beta-catenin deregulation may also result from mutations in the APC, AXIN1, and AXIN2 genes.


Subject(s)
Carcinoma, Endometrioid/genetics , Cytoskeletal Proteins/genetics , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/genetics , Repressor Proteins , Trans-Activators , Adult , Aged , Axin Protein , Carcinoma, Endometrioid/metabolism , Carcinoma, Endometrioid/pathology , Cell Nucleus/metabolism , Cytoskeletal Proteins/biosynthesis , DNA Mutational Analysis , DNA-Binding Proteins/physiology , Female , Genes, APC , Humans , Lymphoid Enhancer-Binding Factor 1 , Middle Aged , Mutation , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Proteins/genetics , Transcription Factors/physiology , Transcription, Genetic , Tumor Cells, Cultured , beta Catenin
10.
Am J Pathol ; 159(4): 1231-8, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11583950

ABSTRACT

Molecular classification of tumors based on their gene expression profiles promises to significantly refine diagnosis and management of cancer patients. The establishment of organ-specific gene expression patterns represents a crucial first step in the clinical application of the molecular approach. Here, we report on the gene expression profiles of 154 primary adenocarcinomas of the lung, colon, and ovary. Using high-density oligonucleotide arrays with 7129 gene probe sets, comprehensive gene expression profiles of 57 lung, 51 colon, and 46 ovary adenocarcinomas were generated and subjected to principle component analysis and to a cross-validated prediction analysis using nearest neighbor classification. These statistical analyses resulted in the classification of 152 of 154 of the adenocarcinomas in an organ-specific manner and identified genes expressed in a putative tissue-specific manner for each tumor type. Furthermore, two tumors were identified, one in the colon group and another in the ovarian group, that did not conform to their respective organ-specific cohorts. Investigation of these outlier tumors by immunohistochemical profiling revealed the ovarian tumor was consistent with a metastatic adenocarcinoma of colonic origin and the colonic tumor was a pleomorphic mesenchymal tumor, probably a leiomyosarcoma, rather than an epithelial tumor. Our results demonstrate the ability of gene expression profiles to classify tumors and suggest that determination of organ-specific gene expression profiles will play a significant role in a wide variety of clinical settings, including molecular diagnosis and classification.


Subject(s)
Adenocarcinoma/genetics , Colonic Neoplasms/genetics , Gene Expression Profiling , Lung Neoplasms/genetics , Ovarian Neoplasms/genetics , Adenocarcinoma/classification , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Colonic Neoplasms/classification , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Diagnosis, Differential , Female , Gene Expression , Humans , Immunohistochemistry , Lung Neoplasms/classification , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Ovarian Neoplasms/classification , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology
11.
Biochem Biophys Res Commun ; 286(5): 1087-97, 2001 Sep 07.
Article in English | MEDLINE | ID: mdl-11527412

ABSTRACT

Expression of the DCC (deleted in colorectal cancer) protein is strongly induced during the neural differentiation of mouse P19 embryonal carcinoma (EC) cells that occurs when these cells are treated with retinoic acid (RA). Myc-associated zinc finger protein (MAZ) is a DNA-binding protein that is widely expressed and functions in human, mouse and hamster cells as an activator, an initiator or a terminator of transcription. However, the biological functions of MAZ remain elusive. We report here that MAZ associates with the cytoplasmic domain of the DCC protein in vivo and in vitro. Yeast two-hybrid assays confirmed this association. An immunofluorescence study demonstrated that DCC protein is expressed at elevated levels in neuron-like P19 EC cells, in particular in axons, in which the MAZ protein is also expressed. We found that MAZ was translocated from the nucleus to the cytoplasm during the RA-induced terminal differentiation of P19 EC cells with resultant loss of the ability of MAZ to bind to the ME1a1 site of the c-myc promoter. Taken together, our observations imply that the DCC protein might play a critical role as a signaling molecule in the regulation of the transcriptional activity of MAZ during the neural differentiation of P19 EC cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Neurons/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Binding Sites , Blotting, Northern , Blotting, Western , Cell Differentiation , Cell Line , Cell Nucleus/metabolism , Cytoplasm/metabolism , DCC Receptor , DNA-Binding Proteins , Genes, myc/genetics , Humans , Immunohistochemistry , Microscopy, Fluorescence , Models, Biological , Molecular Sequence Data , Plasmids/metabolism , Precipitin Tests , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , RNA/metabolism , RNA, Messenger/metabolism , Receptors, Cell Surface , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Tretinoin/metabolism , Two-Hybrid System Techniques , Xenopus , Zinc Fingers
12.
Genes Chromosomes Cancer ; 30(3): 316-21, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11170292

ABSTRACT

Constitutive activation of the Wnt signaling pathway as a result of genetic alterations of APC, AXIN1, and CTNNB1 has been found in various human cancers, including those of the colon, liver, endometrium, ovary, prostate, and stomach. To investigate the pathogenetic significance of constitutive activation of the Wnt signaling pathway in human lung carcinogenesis, CTNNB1 alterations in exon 3, a region known to represent a mutation hot spot, were screened in 46 lung cancer cell lines and 47 primary lung cancers. Missense mutations causing substitutions of Ser/Thr residues critical for regulation by GSK-3beta were detected in one (2%) of the cell lines, A427, and two (4%) of the surgical specimens. The three lung cancers with CTNNB1 mutations were adenocarcinomas. To explore the prevalence of constitutive activation of the Wnt signaling pathway in human lung cancer, we assessed 15 lung cancer cell lines representing major histological subtypes of lung cancers for constitutive Tcf transcriptional activity (CTTA). CTTA was observed only in the A427 adenocarcinoma cell line, but not in the remaining 14 cell lines. The data indicate that constitutive activation of the Wnt signaling pathway caused by CTNNB1 mutation is involved in the development and/or progression of a subset of lung carcinoma, preferentially in adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Cytoskeletal Proteins/genetics , Lung Neoplasms/genetics , Mutation, Missense/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Trans-Activators , Zebrafish Proteins , Adenocarcinoma/enzymology , Cadherins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Enzyme Activation/genetics , Glycogen Synthase Kinase 3 , Humans , Lung Neoplasms/enzymology , Tumor Cells, Cultured , Wnt Proteins , beta Catenin
13.
Proc Natl Acad Sci U S A ; 97(23): 12613-8, 2000 Nov 07.
Article in English | MEDLINE | ID: mdl-11050151

ABSTRACT

The beta-catenin protein plays a critical role in embryonic development and mature tissue homeostasis through its effects on E-cadherin-mediated cell adhesion and Wnt-dependent signal transduction. In colon and other cancers, mutations of beta-catenin or the adenomatous polyposis coli (APC) tumor suppressor appear to stabilize beta-catenin and enhance its interaction with T cell factor (TCF) or lymphoid enhancer factor (Lef) transcription factors. At present, a complete picture of the means by which beta-catenin's interactions with TCF/Lef proteins contribute to neoplastic transformation is lacking. We report that the transcriptional coactivator p300 interacts with beta-catenin in vitro and in vivo and is critical for beta-catenin-mediated neoplastic transformation. p300 synergistically activates beta-catenin/TCF transcription, and their biochemical association requires the CH1 domain of p300 and a region of beta-catenin that includes its NH(2)-terminal transactivation domain and the first two armadillo repeats. Lowering of cellular p300 levels by using a ribozyme directed against p300 reduced TCF transcriptional activity and inhibited the neoplastic growth properties of a beta-catenin-transformed rat epithelial cell line and a human colon carcinoma line with a beta-catenin mutation. These findings demonstrate a critical role for p300 in beta-catenin/TCF transcription and in cancers arising from defects in beta-catenin regulation.


Subject(s)
Cell Transformation, Neoplastic , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/genetics , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Transcription Factors/genetics , Animals , Cell Line , Cytoskeletal Proteins/genetics , E1A-Associated p300 Protein , Gene Expression Regulation , Humans , Jurkat Cells , Lymphoid Enhancer-Binding Factor 1 , Mice , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Transcription, Genetic , Tumor Cells, Cultured , beta Catenin
14.
Genes Dev ; 14(11): 1319-31, 2000 Jun 01.
Article in English | MEDLINE | ID: mdl-10837025

ABSTRACT

beta-Catenin and gamma-catenin (plakoglobin), vertebrate homologs of Drosophila armadillo, function in cell adhesion and the Wnt signaling pathway. In colon and other cancers, mutations in the APC tumor suppressor protein or beta-catenin's amino terminus stabilize beta-catenin, enhancing its ability to activate transcription of Tcf/Lef target genes. Though beta- and gamma-catenin have analogous structures and functions and like binding to APC, evidence that gamma-catenin has an important role in cancer has been lacking. We report here that APC regulates both beta- and gamma-catenin and gamma-catenin functions as an oncogene. In contrast to beta-catenin, for which only amino-terminal mutated forms transform RK3E epithelial cells, wild-type and several amino-terminal mutated forms of gamma-catenin had similar transforming activity. gamma-Catenin's transforming activity, like beta-catenin's, was dependent on Tcf/Lef function. However, in contrast to beta-catenin, gamma-catenin strongly activated c-Myc expression and c-Myc function was crucial for gamma-catenin transformation. Our findings suggest APC mutations alter regulation of both beta- and gamma-catenin, perhaps explaining why the frequency of APC mutations in colon cancer far exceeds that of beta-catenin mutations. Elevated c-Myc expression in cancers with APC defects may be due to altered regulation of both beta- and gamma-catenin. Furthermore, the data imply beta- and gamma-catenin may have distinct roles in Wnt signaling and cancer via differential effects on downstream target genes.


Subject(s)
Cytoskeletal Proteins/metabolism , Neoplasms/metabolism , Trans-Activators , Zebrafish Proteins , Adenomatous Polyposis Coli Protein , Alleles , Animals , Cell Adhesion , Cell Division , Cell Line , Cell Line, Transformed , Colonic Neoplasms/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/physiology , DNA-Binding Proteins/metabolism , Desmoplakins , Fluorescent Antibody Technique , Gene Expression Regulation , Humans , Lymphoid Enhancer-Binding Factor 1 , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/biosynthesis , Signal Transduction , Time Factors , Transcription Factors/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Wnt Proteins , beta Catenin , gamma Catenin
16.
Genes Chromosomes Cancer ; 27(4): 353-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10719364

ABSTRACT

Loss of heterozygosity (LOH) of chromosome arm 18q is frequent in gastrointestinal cancers. Over 90% of pancreatic carcinomas have 18q LOH. Bi-allelic inactivation of the MADH4/DPC4/SMAD4 gene at 18q21.1 is seen in about half of pancreatic carcinomas with 18q LOH. In the remaining tumors with 18q LOH, MADH4 is not mutated and its expression is unaffected, and no alterations in MADH2/SMAD2, a MADH4-related gene at 18q12.3, have been found. A controversial candidate tumor-suppressor gene at 18q21.2 is DCC (deleted in colorectal carcinoma), which encodes a netrin-1 receptor component with functions in cell migration and apoptosis. Reduced or absent DCC expression has been observed in many cancers, but few somatic mutations that would clearly inactivate DCC function have been reported. We studied a panel of 115 pancreatic and 14 biliary cancers for homozygous deletions of DCC exons and flanking 18q regions. Seven homozygous deletions were seen in the region that includes the DCC gene. In two tumors, the deletions inactivate DCC but not MADH4. A physical and transcript map of the deleted regions was constructed, and DCC was the only known gene affected by all seven deletions. These data are the strongest mutational evidence presented yet in support of the hypothesis that DCC or another gene in the region distal to MADH4 is inactivated, playing a causal role in cancer development.


Subject(s)
Biliary Tract Neoplasms/genetics , DNA-Binding Proteins/genetics , Gene Deletion , Genes, DCC , Homozygote , Pancreatic Neoplasms/genetics , Trans-Activators/genetics , Adenocarcinoma/genetics , Animals , Chromosomes, Human, Pair 18/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Humans , Loss of Heterozygosity , Mice , Mice, Nude , Receptors, Cell Surface/genetics , Smad4 Protein , Transplantation, Heterologous , Tumor Cells, Cultured
17.
J Biol Chem ; 275(15): 10943-53, 2000 Apr 14.
Article in English | MEDLINE | ID: mdl-10753894

ABSTRACT

The WT1 tumor suppressor gene encodes a transcription factor that can activate and repress gene expression. Transcriptional targets relevant for the growth suppression functions of WT1 are poorly understood. We found that mesenchymal NIH 3T3 fibroblasts stably expressing WT1 exhibit growth suppression and features of epithelial differentiation including up-regulation of E-cadherin mRNA. Acute expression of WT1 in NIH 3T3 fibroblasts after retroviral infection induced murine E-cadherin expression. In transient transfection experiments, the human and murine E-cadherin promoters were activated by co-expression of WT1. E-cadherin promoter activity was increased in cells overexpressing WT1 and was blocked by a dominant negative form of WT1. WT1 activated the murine E-cadherin promoter through a conserved GC-rich sequence similar to an EGR-1 binding site as well as through a CAAT box sequence. WT1 produced in vitro or derived from nuclear extracts bound to the WT1-response element within the murine E-cadherin promoter, but not the CAAT box. E-cadherin, a gene important in epithelial differentiation and neoplastic transformation, represents a downstream target gene that links the roles of the WT1 in differentiation and growth control.


Subject(s)
Cadherins/genetics , DNA-Binding Proteins/genetics , Transcription Factors/genetics , 3T3 Cells , Animals , Base Sequence , DNA-Binding Proteins/physiology , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Transcription Factors/physiology , Transfection , WT1 Proteins
18.
Oncogene ; 18(51): 7274-9, 1999 Dec 02.
Article in English | MEDLINE | ID: mdl-10602481

ABSTRACT

Inactivation of the E-cadherin cell adhesion molecule is believed critical in the development and behavior of many epithelial cancers, though mutations in the E-cadherin gene account for inactivation in only a fraction of cases. In many breast cancer lines, E-cadherin transcription is extinguished, but the role and significance of alterations in trans-acting transcription factors, promoter hypermethylation, and chromatin changes remain unresolved. To gain further insights into mechanisms underlying E-cadherin inactivation in breast cancer, we analysed somatic cell hybrids resulting from pairwise fusions between breast cancer lines with intact E-cadherin transcription (E-cad+) and lines lacking E-cadherin transcription (E-cad-). All hybrid lines failed to express E-cadherin transcripts and protein, despite the fact that E-cadherin alleles from E-cad+ lines were present in the hybrids. Elements in the proximal 108 bp of the E-cadherin promoter, when present in reporter gene constructs, were sufficient to direct strong transcription in E-cad+ breast lines, but displayed weak activity in E-cad- parental lines and hybrids. E-cadherin expression could not be restored in E-cad- lines or hybrids by treatment with a DNA demethylating agent and/or a histone deacetylase inhibitor. Our findings suggest loss of E-cadherin expression in some breast cancers may be due to dominant repression of the trans-acting pathways that regulate E-cadherin transcription.


Subject(s)
Breast Neoplasms/genetics , Cadherins/genetics , Gene Expression Regulation, Neoplastic , Promoter Regions, Genetic/genetics , Base Sequence , Breast Neoplasms/metabolism , Cadherins/biosynthesis , Female , Humans , Molecular Sequence Data , Transcription Factors/genetics , Tumor Cells, Cultured
20.
Mol Cell Biol ; 19(8): 5696-706, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10409758

ABSTRACT

Current models predict that beta-catenin (beta-cat) functions in Wnt signaling via activation of Tcf/Lef target genes and that its abundance is regulated by the adenomatous polyposis coli (APC) and glycogen synthase kinase 3beta (GSK3beta) proteins. In colon and other cancers, mutations in APC or presumptive GSK3beta phosphorylation sites of beta-cat are associated with constitutive activation of Tcf/Lef transcription. In spite of assumptions about its oncogenic potential, prior efforts to demonstrate that mutated beta-cat will induce neoplastic transformation have yielded equivocal results. We report here that mutated, but not wild-type, beta-cat proteins induced neoplastic transformation of RK3E, an adenovirus E1A-immortalized epithelial cell line. Analysis of the properties of mutant beta-cat proteins and studies with a dominant negative Tcf-4 mutant indicated that the ability of beta-cat to bind and activate Tcf/Lef factors is crucial for transformation. c-myc has recently been implicated as a critical Tcf-regulated target gene. However, c-myc was not consistently activated in beta-cat-transformed RK3E cells, and a dominant negative c-Myc mutant protein failed to inhibit beta-cat transformation. Our findings underscore the role of beta-cat mutations and Tcf/Lef activation in cancer and illustrate a useful system for defining critical factors in beta-cat transformation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , Genes, myc , Trans-Activators , Transcription Factors/genetics , Transcription, Genetic , Zebrafish Proteins , Adenoviridae/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Line, Transformed/metabolism , Cell Transformation, Viral , Cytoskeletal Proteins/physiology , Epithelial Cells , Genes, APC , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Kidney , Lymphoid Enhancer-Binding Factor 1 , Mutagenesis, Site-Directed , Proto-Oncogene Proteins/physiology , Rats , Signal Transduction , Wnt Proteins , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...