Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Pathol Oncol Res ; 18(4): 783-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22729835

ABSTRACT

RUNX3 aberrations play a pivotal role in the oncogenesis of breast, gastric, colon, skin and lung tissues. The aim of this study was to characterize further the expression of RUNX3 in lung cancers. To achieve this, a lung cancer tissue microarray (TMA), frozen lung cancer tissues and lung cell lines were examined for RUNX3 expression by immunohistochemistry, while the TMA was also examined for EGFR and p53 expression. RUNX3 promoter methylation status, and EGFR and KRAS mutation status were also investigated. Inactivation of RUNX3 was observed in 70% of the adenocarcinoma samples, and this was associated with promoter hypermethylation but not biased to EGFR/KRAS mutations. Our results suggest a central role of RUNX3 downregulation in pulmonary adenocarcinoma, which may not be dependent of other established cancer-causing pathways and may have important diagnostic and screening implications.


Subject(s)
Adenocarcinoma/genetics , Core Binding Factor Alpha 3 Subunit/genetics , ErbB Receptors/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics , Adenocarcinoma/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Carcinoma, Squamous Cell/chemistry , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Core Binding Factor Alpha 3 Subunit/metabolism , DNA Methylation , DNA Mutational Analysis , Down-Regulation , ErbB Receptors/metabolism , Humans , Immunohistochemistry , Lung Neoplasms/chemistry , Lung Neoplasms/metabolism , Mutation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Tissue Array Analysis , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism
2.
Clin Cancer Res ; 15(4): 1435-42, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19228744

ABSTRACT

PURPOSE: Cell cycle dysregulation resulting in expression of antiapoptotic genes and uncontrolled proliferation is a feature of undifferentiated nasopharyngeal carcinoma. The pharmacodynamic effects of seliciclib, a cyclin-dependent kinase (CDK) inhibitor, were studied in patients with nasopharyngeal carcinoma. EXPERIMENTAL DESIGN: Patients with treatment-naïve locally advanced nasopharyngeal carcinoma received seliciclib at 800 mg or 400 mg twice daily on days 1 to 3 and 8 to 12. Paired tumor samples obtained at baseline and on day 13 were assessed by light microscopy, immunohistochemistry, and transcriptional profiling using real-time PCR low-density array consisting of a panel of 380 genes related to cell cycle inhibition, apoptosis, signal transduction, and cell proliferation. RESULTS: At 800 mg bd, one patient experienced grade 3 liver toxicity and another had grade 2 vomiting; no significant toxicities were experienced in 13 patients treated at 400 mg bd. Seven of fourteen evaluable patients had clinical evidence of tumor reduction. Some of these responses were associated with increased tumor apoptosis, necrosis, and decreases in plasma EBV DNA posttreatment. Reduced protein expression of Mcl-1, cyclin D1, phosphorylated retinoblastoma protein pRB (T821), and significant transcriptional down-regulation of genes related to cellular proliferation and survival were shown in some patients posttreatment, indicative of cell cycle modulation by seliciclib, more specifically inhibition of cdk2/cyclin E, cdk7/cyclin H, and cdk9/cyclin T. CONCLUSIONS: Brief treatment with this regimen of seliciclib in patients with nasopharyngeal carcinoma is tolerable at 400 mg bd and associated with tumor pharmacodynamic changes consistent with cdk inhibition, and warrants further efficacy studies in this tumor.


Subject(s)
Antineoplastic Agents/therapeutic use , Cell Cycle/drug effects , Nasopharyngeal Neoplasms/drug therapy , Purines/therapeutic use , Administration, Oral , Adult , Aged , Apoptosis/drug effects , Cyclin D1/analysis , DNA, Viral/blood , Female , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Proto-Oncogene Proteins c-bcl-2/analysis , Purines/adverse effects , Purines/blood , Roscovitine
3.
J Transl Med ; 7: 8, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19152710

ABSTRACT

BACKGROUND: Members of the TRIP-Br/SERTAD family of mammalian transcriptional coregulators have recently been implicated in E2F-mediated cell cycle progression and tumorigenesis. We, herein, focus on the detailed functional characterization of the least understood member of the TRIP-Br/SERTAD protein family, TRIP-Br2 (SERTAD2). METHODS: Oncogenic potential of TRIP-Br2 was demonstrated by (1) inoculation of NIH3T3 fibroblasts, which were engineered to stably overexpress ectopic TRIP-Br2, into athymic nude mice for tumor induction and (2) comprehensive immunohistochemical high-throughput screening of TRIP-Br2 protein expression in multiple human tumor cell lines and human tumor tissue microarrays (TMAs). Clinicopathologic analysis was conducted to assess the potential of TRIP-Br2 as a novel prognostic marker of human cancer. RNA interference of TRIP-Br2 expression in HCT-116 colorectal carcinoma cells was performed to determine the potential of TRIP-Br2 as a novel chemotherapeutic drug target. RESULTS: Overexpression of TRIP-Br2 is sufficient to transform murine fibroblasts and promotes tumorigenesis in nude mice. The transformed phenotype is characterized by deregulation of the E2F/DP-transcriptional pathway through upregulation of the key E2F-responsive genes CYCLIN E, CYCLIN A2, CDC6 and DHFR. TRIP-Br2 is frequently overexpressed in both cancer cell lines and multiple human tumors. Clinicopathologic correlation indicates that overexpression of TRIP-Br2 in hepatocellular carcinoma is associated with a worse clinical outcome by Kaplan-Meier survival analysis. Small interfering RNA-mediated (siRNA) knockdown of TRIP-Br2 was sufficient to inhibit cell-autonomous growth of HCT-116 cells in vitro. CONCLUSION: This study identifies TRIP-Br2 as a bona-fide protooncogene and supports the potential for TRIP-Br2 as a novel prognostic marker and a chemotherapeutic drug target in human cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Humans , Mice , Mice, Nude
4.
Pathology ; 40(5): 441-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18604728

ABSTRACT

Formalin fixed and paraffin embedded tissue (FFPE) collections in pathology departments are the largest resource for retrospective biomedical research studies. Based on the literature analysis of FFPE related research, as well as our own technical validation, we present the Translational Research Arrays (TRARESA), a tissue microarray centred, hospital based, translational research conceptual framework for both validation and/or discovery of novel biomarkers. TRARESA incorporates the analysis of protein, DNA and RNA in the same samples, correlating with clinical and pathological parameters from each case, and allowing (a) the confirmation of new biomarkers, disease hypotheses and drug targets, and (b) the postulation of novel hypotheses on disease mechanisms and drug targets based on known biomarkers. While presenting TRARESA, we illustrate the use of such a comprehensive approach. The conceptualisation of the role of FFPE-based studies in translational research allows the utilisation of this commodity, and adds to the hypothesis-generating armamentarium of existing high-throughput technologies.


Subject(s)
Biomarkers/analysis , Tissue Array Analysis/methods , DNA/analysis , Formaldehyde , Hospitals , Humans , Paraffin Embedding , RNA/analysis , Tissue Fixation , Validation Studies as Topic
5.
Biotechnol J ; 2(11): 1360-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17806102

ABSTRACT

The NAD(+)-dependent protein deacetylase SIRT1 is linked to cellular survival pathways by virtue of keeping the tumor suppressor gene p53 and members of the forkhead transcription factor family deacetylated. To validate SIRT1 as a therapeutic anti-cancer target, we performed immunohistochemistry experiments to study the in vivo expression of SIRT1 in cancer specimens. We show that human SIRT1 is highly expressed in cancer cell lines as well as in tissue samples from colon carcinoma patients. Interestingly, there is a strong cytosolic component in the SIRT1 expression pattern. We further characterized SIRT1 in p53-wild-type and -mutant cell lines and show that SIRT1 mRNA-knockdown leads to a p53-independent decrease of cell proliferation and induction of apoptosis. In addition, SIRT1 expression has been found to be inducible upon DNA damage. A previously discovered small molecule SIRT1 inhibitor with nanomolar in vitro activity has been tested in cancer relevant assays. The SIRT1 inhibitory compound showed no potent anti-proliferative activity despite hitting its molecular target within tumor cells. From these studies we conclude that it may not be sufficient to block the catalytic function of SIRT1, and that its survival effects may be mainly brought about by means other then the deacetylase function. The increased cytosolic expression of SIRT1 in cancer cells could be an indicator of such novel functions.


Subject(s)
Neoplasms/metabolism , Sirtuins/metabolism , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytosol/metabolism , DNA Damage , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Immunohistochemistry , Mutation , Neoplasms/genetics , Neoplasms/pathology , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sirtuin 1 , Sirtuins/genetics , Tissue Array Analysis , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
Neurosci Lett ; 407(2): 124-6, 2006 Oct 23.
Article in English | MEDLINE | ID: mdl-16978776

ABSTRACT

Astrocytic tumors are the most common intracranial neoplasms. Their prognoses correlate with a conventional morphological grading system that suffers from diagnostic subjectivity and hence, inter-observer inconsistency. A molecular marker that provides an objective reference for classification and prognostication of astrocytic tumors would be useful in diagnostic pathology. RhoA, a GTPase protein involved in cell migration and adhesion, has been shown to be upregulated in a variety of human cancers. Based on direct analysis of clinical materials, our study demonstrates increased expression of RhoA in high-grade astrocytomas. This observation may be relevant to astrocytoma biology and the development of potential therapeutics against high-grade astrocytomas. Of more immediate consequence, utilization of this marker may aid in the routine pathological grading (and hence prognostication) of astrocytomas.


Subject(s)
Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , rhoA GTP-Binding Protein/biosynthesis , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Immunohistochemistry , Infant , Male , Middle Aged , Neoplasm Invasiveness/pathology , Paraffin Embedding , Tissue Fixation , Up-Regulation
7.
Cancer Res ; 65(17): 7743-50, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140942

ABSTRACT

Loss of RUNX3 expression is suggested to be causally related to gastric cancer as 45% to 60% of gastric cancers do not express RUNX3 mainly due to hypermethylation of the RUNX3 promoter. Here, we examined for other defects in the properties of RUNX3 in gastric cancers that express RUNX3. Ninety-seven gastric cancer tumor specimens and 21 gastric cancer cell lines were examined by immunohistochemistry using novel anti-RUNX3 monoclonal antibodies. In normal gastric mucosa, RUNX3 was expressed most strongly in the nuclei of chief cells as well as in surface epithelial cells. In chief cells, a significant portion of the protein was also found in the cytoplasm. RUNX3 was not detectable in 43 of 97 (44%) cases of gastric cancers tested and a further 38% showed exclusive cytoplasmic localization, whereas only 18% showed nuclear localization. Evidence is presented suggesting that transforming growth factor-beta is an inducer of nuclear translocation of RUNX3, and RUNX3 in the cytoplasm of cancer cells is inactive as a tumor suppressor. RUNX3 was found to be inactive in 82% of gastric cancers through either gene silencing or protein mislocalization to the cytoplasm. In addition to the deregulation of mechanisms controlling gene expression, there would also seem to be at least one other mechanism controlling nuclear translocation of RUNX3 that is impaired frequently in gastric cancer.


Subject(s)
DNA-Binding Proteins/metabolism , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , Core Binding Factor Alpha 3 Subunit , Cytoplasm/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Epithelial Cells/metabolism , Female , Gastric Mucosa/metabolism , Gene Silencing , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/immunology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...