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.
Methods Mol Biol ; 986: 325-37, 2013.
Article in English | MEDLINE | ID: mdl-23436421

ABSTRACT

Proper target selection and validation are crucial to the discovery of new anti-cancer agents. Since tumors depend on a suitable microenvironment for their growth, once a putative target has been identified, its validation should be performed whenever possible in vivo. This chapter deals with the generation of human xenograft mouse models genetically modified to induce the modulation of cancer-related genes as an approach to validate oncology targets.


Subject(s)
Drug Design , Genetic Engineering/methods , RNA Interference , Xenograft Model Antitumor Assays/methods , Animals , Disease Models, Animal , Humans , Mice , Neoplasm Transplantation , RNA, Small Interfering
2.
Cell Cycle ; 11(13): 2507-17, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22713237

ABSTRACT

Targeting Chk1 protein kinase can enhance the antitumor effects of radio- and chemotherapy. Recent evidence disclosed a role of Chk1 in unperturbed cell proliferation and survival, implying that Chk1 inhibitors could also be effective as single agents in tumors with a specific genetic background. To identify genes in synthetic lethality with Chk1, we did a high-throughput screening using a siRNA library directed against 719 human protein kinases in the human ovarian cancer cell line OVCAR-5, resistant to Chk1 inhibitors. Wee1 tyrosine kinase was the most significant gene in synthetic lethality with Chk1. Treatment with non-toxic concentrations of a Chk1 inhibitor (PF-00477736) and a Wee1 inhibitor (MK-1775) confirmed the marked synergistic effect in various human cancer cell lines (breast, ovarian, colon, prostate), independently of the p53 status. Detailed molecular analysis showed that the combination caused cancer cells to undergo premature mitosis before the end of DNA replication, with damaged DNA leading to cell death partly by apoptosis. In vivo treatment of mice bearing OVCAR-5 xenografts with the combination of Chk1 and Wee1 inhibitors led to greater tumor growth inhibition than with the inhibitors used as single agents with no toxicity. These data provide a strong rationale for the clinical investigation of the combination of a Chk1 and a Wee1 inhibitor.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins/metabolism , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , Benzodiazepinones/pharmacology , Benzodiazepinones/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Checkpoint Kinase 1 , DNA Replication/drug effects , Drug Synergism , Drug Therapy, Combination , Female , Humans , Mice , Mice, Nude , Mitosis/drug effects , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Ovarian Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/chemistry , Protein Kinases/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrimidinones , RNA Interference , RNA, Small Interfering/metabolism , Transplantation, Heterologous
3.
J Proteomics ; 75(8): 2417-30, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22387315

ABSTRACT

Recent studies showed that food-contaminant non-dioxin-like polychlorinated biphenyls (NDL-PCBs) congeners (PCB52, PCB138, PCB180) have neurotoxic potential, but the cellular and molecular mechanisms underlying neuronal damage are not entirely known. The aim of this study was to assess whether in-vitro exposure to NDL-PCBs may alter the proteome profile of primary cerebellar neurons in order to expand our knowledge on NDL-PCBs neurotoxicity. Comparison of proteome from unexposed and exposed rat cerebellar neurons was performed using state-of-the-art label-free semi-quantitative mass-spectrometry method. We observed significant changes in the abundance of several proteins, that fall into two main classes: (i) novel targets for both PCB138 and 180, mediating the dysregulation of CREB pathways and ubiquitin-proteasome system; (ii) different congeners-specific targets (alpha-actinin-1 for PCB138; microtubule-associated-protein-2 for PCB180) that might lead to similar deleterious consequences on neurons cytoskeleton organization. Interference of the PCB congeners with synaptic formation was supported by the increased expression of pre- and post-synaptic proteins quantified by western blot and immunocytochemistry. Expression alteration of synaptic markers was confirmed in the cerebellum of rats developmentally exposed to these congeners, suggesting an adaptive response to neurodevelopmental toxicity on brain structures. As such, our work is expected to lead to new insights into the mechanisms of NDL-PCBs neurotoxicity.


Subject(s)
Brain/drug effects , Nerve Tissue Proteins/analysis , Polychlorinated Biphenyls/toxicity , Proteome/drug effects , Animals , Animals, Newborn , Brain/metabolism , Brain Chemistry/drug effects , Cells, Cultured , Cerebellum/chemistry , Cerebellum/drug effects , Cerebellum/metabolism , Dioxins/chemistry , Dioxins/toxicity , Environmental Pollutants/chemistry , Environmental Pollutants/toxicity , Food Contamination , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Neurons/chemistry , Neurons/drug effects , Neurons/metabolism , Polychlorinated Biphenyls/chemistry , Proteome/analysis , Proteome/metabolism , Rats , Rats, Wistar
4.
Cancer Res ; 71(13): 4573-84, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21602434

ABSTRACT

The phosphatidylinositol-3-kinase (PI3K)/Akt/mTOR pathway is a major target for cancer therapy. As a strategy to induce the maximal inhibition of this pathway in cancer cells, we combined allosteric mTOR inhibitors (rapamycin and RAD001) with a dual PI3K/mTOR kinase inhibitor (PI-103). Both in vitro and in vivo, the combination exhibited more activity than single agents in human ovarian and prostate cancer cells that harbor alterations in the pathway. At the molecular level, combined inhibition of mTOR prevented the rebound activation of Akt that is seen after treatment with rapamycin and its analogues and caused more sustained inhibition of Akt phosphorylation. Furthermore, the combination strongly inhibited the expression of PI3K/Akt/mTOR downstream proteins. In particular, it showed greater activity than the single agents in inhibiting the phosphorylation of 4EBP1, both in vitro and in vivo, resulting in selective inhibition of CAP-dependent translation. A proteomic approach was used to confirm the identification of c-Myc as the key regulator for the reduction in downstream proteins affected by the combined inhibition of mTOR. In conclusion, the combination of a catalytic and an allosteric inhibitor of mTOR shows greater activity, without a concomitant increase in toxicity, than either drug alone, and this may have therapeutic implications for inhibiting this pathway in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Administration Schedule , Drug Synergism , Female , Furans/administration & dosage , Furans/pharmacology , Humans , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Nude , Multiprotein Complexes , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/administration & dosage , Pyridines/pharmacology , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Sirolimus/administration & dosage , Sirolimus/pharmacology
5.
Cancer Prev Res (Phila) ; 3(8): 929-39, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20628003

ABSTRACT

Some naturally occurring flavonols, exemplified by quercetin, seem to possess experimental cancer chemopreventive efficacy. Modulation of p53 is a mechanism thought to contribute to their activity. The hypothesis was tested that a synthetic flavonol, 3',4',5'-trimethoxyflavonol (TMFol), can interfere with tumor development and p53 expression in two models of colorectal carcinogenesis, Apc(Min) mice and human-derived HCT116 adenocarcinoma-bearing nude mice. Mice received TMFol with their diet (0.2%) from weaning to week 16 in the case of Apc(Min) or from either day 7 before ("TMFol early") or day 7 after ("TMFol late") tumor inoculation in HCT116 mice. The ability of TMFol to affect tumor proliferation or apoptosis, as reflected by staining for Ki-67 or cleaved caspase-3, respectively, was studied in HCT116 tumors. TMFol tumor levels were measured by high-performance liquid chromatography. Consumption of TMFol reduced small intestinal adenoma burden in Apc(Min) mice by 47%, compared with control mice (P < 0.002). The TMFol early regimen approximately halved HCT116 tumor size (P < 0.05), decreased tumor proliferation, and increased apoptosis, whereas the TMFol late regimen had no significant effect when compared with controls. In tumor tissues from mice, in which TMFol reduced tumor development, p53 expression was increased 3-fold in Apc(Min) and 1.5-fold in HCT116 tumor-bearing mice (P = 0.02). TMFol increased p53 also in cells derived from these tumors. TMFol was detected in HCT116 tumors, but levels did not correlate with tumor burden. TMFol was not mutagenic in the Ames test. The results suggest that chemical modification of the flavonol structure may generate safe and efficacious cancer chemopreventive agents.


Subject(s)
Carcinoma/prevention & control , Colorectal Neoplasms/prevention & control , Drug Resistance, Neoplasm/genetics , Flavonoids/therapeutic use , Genes, p53/physiology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma/genetics , Chemoprevention , Colorectal Neoplasms/genetics , Drug Evaluation, Preclinical , HCT116 Cells , Humans , Mice , Mice, Nude , Mice, Transgenic , Quercetin/analogs & derivatives , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Cancer Res ; 68(24): 10187-96, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-19074886

ABSTRACT

Cell motility and invasion play an essential role in the development of metastasis. Evidence suggests that the enzyme phospholipase Cgamma1 (PLCgamma1) may be involved in tumor progression and possibly development of metastasis. In this study, we show that down-regulation of PLCgamma1 expression severely impairs activation of the small GTP-binding protein Rac and cell invasion in breast cancer cell lines and U87 in vitro. Experimental metastasis assays in nude mice show that inducible knockdown of PLCgamma1 strongly inhibits development of MDA-MB-231-derived lung metastasis and reverts metastasis formation. In addition, analysis of 60 breast cancer patients' tissues revealed an increase of PLCgamma1 expression in metastasis compared with the primary tumor in 50% of tissues analyzed. These data show a critical role of PLCgamma1 in the metastatic potential of cancer cells, and they further indicate that PLCgamma1 inhibition has a therapeutic potential in the treatment of metastasis dissemination.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Phospholipase C gamma/biosynthesis , Animals , Breast Neoplasms/genetics , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/genetics , Cytoskeleton/metabolism , Cytoskeleton/pathology , Disease Progression , Down-Regulation , Female , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Phospholipase C gamma/antagonists & inhibitors , Phospholipase C gamma/genetics , RNA Interference , rac1 GTP-Binding Protein/metabolism
7.
Nucleic Acids Res ; 35(6): 2093-104, 2007.
Article in English | MEDLINE | ID: mdl-17344317

ABSTRACT

Tumour-derived p53 mutants are thought to have acquired 'gain-of-function' properties that contribute to oncogenicity. We have tested the hypothesis that p53 mutants suppress p53-target gene expression, leading to enhanced cellular growth. Silencing of mutant p53 expression in several human cell lines was found to lead to the upregulation of wild-type p53-target genes such as p21, gadd45, PERP and PTEN. The expression of these genes was also suppressed in H1299-based isogenic cell lines expressing various hot-spot p53 mutants, and silencing of mutant p53, but not TAp73, abrogated the suppression. Consistently, these hot-spot p53 mutants were able to suppress a variety of p53-target gene promoters. Analysis using the proto-type p21 promoter construct indicated that the p53-binding sites are dispensable for mutant p53-mediated suppression. However, treatment with the histone deacetylase inhibitor trichostatin-A resulted in relief of mutant p53-mediated suppression, suggesting that mutant p53 may induce hypo-acetylation of target gene promoters leading to the suppressive effects. Finally, we show that stable down-regulation of mutant p53 expression resulted in reduced cellular colony growth in human cancer cells, which was found to be due to the induction of apoptosis. Together, the results demonstrate another mechanism through which p53 mutants could promote cellular growth.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, p53 , Mutation , Neoplasms/genetics , Apoptosis , Binding Sites , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Promoter Regions, Genetic , RNA Interference , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...