Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
J Exp Med ; 204(6): 1453-61, 2007 Jun 11.
Article in English | MEDLINE | ID: mdl-17535972

ABSTRACT

Tumor suppressor p53-dependent apoptosis is critical in suppressing tumorigenesis. Previously, we reported that DNA double-strand breaks (DSBs) at the V(D)J recombination loci induced genomic instability in the developing lymphocytes of nonhomologous end-joining (NHEJ)-deficient, p53-deficient mice, which led to rapid lymphomagenesis. To test the ability of p53-dependent cell cycle arrest to suppress tumorigenesis in the absence of apoptosis in vivo, we crossbred NHEJ-deficient mice into a mutant p53R172P background; these mice have defects in apoptosis induction, but not cell cycle arrest. These double-mutant mice survived longer than NHEJ/p53 double-null mice and, remarkably, were completely tumor free. We detected accumulation of aberrant V(D)J recombination-related DSBs at the T cell receptor (TCR) locus, and high expression levels of both mutant p53 and cell cycle checkpoint protein p21, but not the apoptotic protein p53-upregulated modulator of apoptosis. In addition, a substantial number of senescent cells were observed among both thymocytes and bone marrow cells. Cytogenetic studies revealed euploidy and limited chromosomal breaks in these lymphoid cells. The results indicate that precursor lymphocytes, which normally possess a high proliferation potential, are able to withdraw from the cell cycle and undergo senescence in response to the persistence of DSBs in a p53-p21-dependent pathway; this is sufficient to inhibit oncogenic chromosomal abnormality and suppress tumorigenesis.


Subject(s)
Cellular Senescence/physiology , DNA Breaks, Double-Stranded , Disease Models, Animal , Neoplasms/physiopathology , Animals , Blotting, Western , Cellular Senescence/genetics , Crosses, Genetic , Cytogenetic Analysis , DNA Ligase ATP , DNA Ligases/genetics , DNA Ligases/metabolism , Immunohistochemistry , In Situ Hybridization, Fluorescence , In Situ Nick-End Labeling , Mice , Mice, Inbred C57BL , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
Blood ; 110(1): 267-77, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17356134

ABSTRACT

The proteasome has been successfully targeted for the treatment of multiple myeloma and mantle cell lymphoma; however, in other hematologic malignancies, bortezomib has been less effective as a single agent. Here, we describe effects of NPI-0052, a novel proteasome inhibitor, in leukemia model systems. In cell lines, NPI-0052 inhibits all 3 proteolytic activities associated with the proteasome: chymotrypsin-, trypsin-, and caspase-like. NPI-0052 also induces DNA fragmentation in leukemia lines and in mononuclear cells from a Ph + acute lymphoblastic leukemia (ALL) patient. Caspase-3 activation by NPI-0052 was seen in wild-type Jurkat cells, but was significantly lessened in Fas-associated death domain (FADD)-deficient or caspase-8-deficient counterparts. NPI-0052-induced apoptosis was further probed using caspase-8 inhibitors, which were more protective than caspase-9 inhibitors. N-acetyl cysteine (NAC) also conferred protection against NPI-0052-induced apoptosis, indicating a role for oxidative stress by NPI-0052. In support of the drug's in vitro activities, biweekly treatment with NPI-0052 lessened total white blood cell (WBC) burden over 35 days in leukemic mice. Interestingly, combining NPI-0052 with either MS-275 or valproic acid (VPA) induced greater levels of cell death than the combination of bortezomib with these histone deacetylase inhibitors (HDACi). These effects of NPI-0052, alone and in combination with HDACi, warrant further testing to determine the compound's clinical efficacy in leukemia.


Subject(s)
Apoptosis/drug effects , Histone Deacetylase Inhibitors , Lactones/pharmacology , Leukemia/drug therapy , Proteasome Inhibitors , Pyrroles/pharmacology , Animals , Caspase 8/metabolism , Cell Line, Tumor , Humans , Lactones/administration & dosage , Leukemia/pathology , Mice , Oxidative Stress , Protease Inhibitors/administration & dosage , Protease Inhibitors/pharmacology , Pyrroles/administration & dosage , Reactive Oxygen Species/metabolism , Tumor Burden/drug effects , Tumor Cells, Cultured
3.
Clin Cancer Res ; 12(15): 4671-7, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16899617

ABSTRACT

PURPOSE: It has previously been reported that the patient response to gefitinib depends on the presence of mutations within the kinase domain of epidermal growth factor receptor (EGFR) or the expression of its truncated form, EGFR variant III (EGFRvIII). The focus of this study was to determine if these alterations are present within the tyrosine kinase and ligand-binding domain of EGFR in urothelial carcinoma. EXPERIMENTAL DESIGN: The kinase domain found within exons 18 to 21 of the EGFR from 11 bladder cancer cell lines and 75 patient tumors were subjected to automated sequencing. EGFRvIII expression was determined by immunohistochemistry using a urothelial carcinoma tissue microarray, and its expression was subsequently verified by reverse transcription PCR, real-time PCR, and Western blot analysis, using an EGFRvIII-transfected glioblastoma cell line and glioblastoma tumors as positive controls. RESULTS: Our analysis failed to detect mutations within the tyrosine kinase domain of EGFR in the 11 cell lines and 75 patients tested. The initial analysis of EGFRvIII expression by immunohistochemistry revealed that at least 50% of the patient tumors expressed EGFRvIII in a urothelial carcinoma tissue microarray. Conflicting reports exist, however, regarding the extent of EGFRvIII expression in tissues owing to the specificity of the antibodies and the methodologies used. Therefore, we sought to validate this observation by reverse transcription PCR, real-time PCR, and Western blot analysis. In these assays, none of the samples were positive for EGFRvIII except for control transfectants and glioblastomas. CONCLUSIONS: When our results are taken together, we conclude that alterations within the tyrosine kinase domain and expression of EGFRvIII are rare events in bladder cancer. The present study has clinical implications in selecting tyrosine kinase inhibitors for the therapy of urothelial carcinoma.


Subject(s)
ErbB Receptors/genetics , Point Mutation , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/genetics , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cohort Studies , DNA Mutational Analysis/methods , Drug Screening Assays, Antitumor , ErbB Receptors/biosynthesis , Exons , Gene Expression Profiling , Humans , Immunohistochemistry , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Sequence Deletion , Structure-Activity Relationship , Tissue Array Analysis/methods , Urinary Bladder Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...